1
|
Schultz TE, Mathmann CD, Domínguez Cadena LC, Muusse TW, Kim H, Wells JW, Ulett GC, Hamerman JA, Brooks AJ, Kobe B, Sweet MJ, Stacey KJ, Blumenthal A. TLR4 endocytosis and endosomal TLR4 signaling are distinct and independent outcomes of TLR4 activation. EMBO Rep 2025:10.1038/s44319-025-00444-2. [PMID: 40204912 DOI: 10.1038/s44319-025-00444-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Toll-like receptor 4 (TLR4) signaling at the plasma membrane and in endosomes results in distinct contributions to inflammation and host defence. Current understanding indicates that endocytosis of cell surface-activated TLR4 is required to enable subsequent signaling from endosomes. Contrary to this prevailing model, our data show that endosomal TLR4 signaling is not reliant on cell surface-expressed TLR4 or ligand-induced TLR4 endocytosis. Moreover, previously recognized requirements for the accessory molecule CD14 in TLR4 endocytosis and endosomal signaling are likely attributable to CD14 binding as well as trafficking and transferring lipopolysaccharide (LPS) to TLR4 at different subcellular localizations. TLR4 endocytosis requires the TLR4 intracellular signaling domain, contributions by phospholipase C gamma 2, spleen tyrosine kinase, E1/E2 ubiquitination enzymes, but not canonical TLR signaling adaptors and cascades. Thus, our study identifies independently operating TLR4 signaling modes that control TLR4 endocytosis, pro-inflammatory cell surface-derived, as well as endosomal TLR4 signaling. This revised understanding of how TLR4 functions within cells might be harnessed to selectively amplify or restrict TLR4 activation for the development of adjuvants, vaccines and therapeutics.
Collapse
Affiliation(s)
- Thomas E Schultz
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Carmen D Mathmann
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | | | - Timothy W Muusse
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Hyoyoung Kim
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - James W Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4215, Australia
| | - Jessica A Hamerman
- Immunology Program, Benaroya Research Institute, Seattle, WA, 98101, USA
| | - Andrew J Brooks
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Katryn J Stacey
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Antje Blumenthal
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
2
|
Li Y, Uhelski ML, North RY, Farson LB, Bankston CB, Roland GH, Fan DH, Sheffield KN, Jia A, Orlando D, Heles M, Yaksh TL, Miller YI, Kosten TA, Dougherty PM. ApoA-I binding protein (AIBP) regulates transient receptor potential vanilloid 1 (TRPV1) activity in rat dorsal root ganglion neurons by selective disruption of toll-like receptor 4 (TLR4)-lipid rafts. Brain Behav Immun 2025; 123:644-655. [PMID: 39414176 DOI: 10.1016/j.bbi.2024.10.017] [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: 05/06/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Toll-like receptor 4 (TLR4) and the transient receptor potential vanilloid subtype 1 (TRPV1) are both upregulated and play key roles in the induction and expression of paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Using Apolipoprotein A-I binding protein, non-specific cholesterol depletion, TLR4 mis-sense rats and a TLR4 inhibitor, we demonstrate that co-localization of TRPV1 with TLR4 to cholesterol-rich lipid membrane rafts in nociceptors is essential for its normal activation as well as for its exaggerated activation that underlies the development and expression of CIPN. The findings suggest that TLR4-lipid rafts may have an essential role in numerous neuroinflammatory and neuropathic pain conditions. This mechanism is also generalized to female rats for the first time.
Collapse
Affiliation(s)
- Yan Li
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Megan L Uhelski
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Robert Y North
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, the United States of America
| | - Luke B Farson
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Christopher B Bankston
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Gavin H Roland
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Dwight H Fan
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | | | - Amy Jia
- Northwestern University, Evanston, IL 60208, the United States of America
| | - Dana Orlando
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Mario Heles
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Tony L Yaksh
- The Department of Anesthesiology, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Yury I Miller
- Department of Medicine, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Therese A Kosten
- Department of Psychology, Health Building 1, 4349 Martin Luther King Blvd, Houston, TX 77204, the United States of America
| | - Patrick M Dougherty
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America.
| |
Collapse
|
3
|
Kong J, Yang J, He C, Zhou B, Fang S, Salinas M, Mohabbat AB, Bauer BA, Wang X. Regulation of endotoxemia through the gut microbiota: The role of the Mediterranean diet and its components. APMIS 2024; 132:948-955. [PMID: 39370693 DOI: 10.1111/apm.13473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/12/2024] [Indexed: 10/08/2024]
Abstract
Endotoxemia is closely related to many diseases. As the largest endotoxin reservoir in the human body, the gut microbiota should be a key target for alleviating endotoxemia. The intestinal microbiota is believed to cause endotoxemia directly or indirectly by modifying the intestinal barrier function through dysbiosis, changing intestinal mucosal permeability and bacterial translocation. Diet is known to be the main environmental factor affecting the intestinal microbiota, and different diets and food components have a large impact on the gut microbiota. The Mediterranean diet, which received much attention in recent years, is believed to be able to regulate the gut microbiota, thereby maintaining the function of the intestinal barrier and alleviating endotoxemia. In this review, we focus on the relationship between the gut microbiota and endotoxemia, and how the Mediterranean dietary (MD) pattern can interfere with endotoxemia through the gut microbiota.
Collapse
Affiliation(s)
- Jing Kong
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Juan Yang
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Cong He
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bingduo Zhou
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengquan Fang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Manisha Salinas
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Arya B Mohabbat
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Brent A Bauer
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xiaosu Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Chaudhary N, Kasiewicz LN, Newby AN, Arral ML, Yerneni SS, Melamed JR, LoPresti ST, Fein KC, Strelkova Petersen DM, Kumar S, Purwar R, Whitehead KA. Amine headgroups in ionizable lipids drive immune responses to lipid nanoparticles by binding to the receptors TLR4 and CD1d. Nat Biomed Eng 2024; 8:1483-1498. [PMID: 39363106 PMCID: PMC11863198 DOI: 10.1038/s41551-024-01256-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Lipid nanoparticles (LNPs) are the most clinically advanced delivery vehicle for RNA therapeutics, partly because of established lipid structure-activity relationships focused on formulation potency. Yet such knowledge has not extended to LNP immunogenicity. Here we show that the innate and adaptive immune responses elicited by LNPs are linked to their ionizable lipid chemistry. Specifically, we show that the amine headgroups in ionizable lipids drive LNP immunogenicity by binding to Toll-like receptor 4 and CD1d and by promoting lipid-raft formation. Immunogenic LNPs favour a type-1 T-helper-cell-biased immune response marked by increases in the immunoglobulins IgG2c and IgG1 and in the pro-inflammatory cytokines tumour necrosis factor, interferon γ and the interleukins IL-6 and IL-2. Notably, the inflammatory signals originating from these receptors inhibit the production of anti-poly(ethylene glycol) IgM antibodies, preventing the often-observed loss of efficacy in the LNP-mediated delivery of siRNA and mRNA. Moreover, we identified computational methods for the prediction of the structure-dependent innate and adaptive responses of LNPs. Our findings may help accelerate the discovery of well-tolerated ionizable lipids suitable for repeated dosing.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Lisa N Kasiewicz
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alexandra N Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Mariah L Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Jilian R Melamed
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Samuel T LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Katherine C Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Sushant Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Rahul Purwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
White CS, Dilger RN. Immunomodulatory potential of dietary soybean-derived saponins. J Anim Sci 2024; 102:skae349. [PMID: 39529449 PMCID: PMC11630861 DOI: 10.1093/jas/skae349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Soybeans are widely recognized as a valuable crop, often included as a high-quality protein source in production animal diets. In addition to contributing to the macronutrient composition of the diet, soybeans also contain many minor bioactive components which can influence the health and growth of animals. This review examined the immunomodulatory potential of soy saponins and their specific effects on the inflammatory response, oxidative stress, and intestinal barrier function. Saponins are amphiphilic molecules, a property imparted by their polar carbohydrate chains that attach to a nonpolar aglycone backbone. This structure also complicates their isolation, thus most research investigating soy saponins has been performed in models that only require small amounts of isolated material. Many experiments conducted in vitro or in rodents reported that saponins can reduce damage, particularly in conditions where a challenge was first introduced to stimulate inflammation or oxidative stress. It appears that saponins can exert their anti-inflammatory effects through modulation of the NF-κB pathway, reducing its activation and the release of pro-inflammatory molecules later in the cascade. Furthermore, soy saponins can influence levels of important anti-oxidative enzymes and reduce the generation of reactive oxygen species, thus attenuating levels of oxidative stress in the model. As these results were obtained from experiments done in vitro or in rodents, they neglect to provide a good representation of how soy saponins may affect some of the greatest consumers of soy-based products, with those being production animals. The work that has been done seems to indicate that soy saponins may exert similar anti-inflammatory and anti-oxidative effects in production animals as those observed in other research models along with immunostimulatory activity that may help boost host defense systems. Overall, there is a dearth of research regarding the effects of soy saponins on species that commonly consume soy products, which begins by developing more effective methods of saponin extraction.
Collapse
Affiliation(s)
- Cameron S White
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| |
Collapse
|
6
|
Tagaeva R, Efimova S, Ischenko A, Zhakhov A, Shevtsov M, Ostroumova O. A new look at Hsp70 activity in phosphatidylserine-enriched membranes: chaperone-induced quasi-interdigitated lipid phase. Sci Rep 2023; 13:19233. [PMID: 37932471 PMCID: PMC10628215 DOI: 10.1038/s41598-023-46131-x] [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: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 11/08/2023] Open
Abstract
70 kDa heat shock protein Hsp70 (also termed HSP70A1A) is the major stress-inducible member of the HSP70 chaperone family, which is present on the plasma membranes of various tumor cells, but not on the membranes of the corresponding normal cells. The exact mechanisms of Hsp70 anchoring in the membrane and its membrane-related functions are still under debate, since the protein does not contain consensus signal sequence responsible for translocation from the cytosol to the lipid bilayer. The present study was focused on the analysis of the interaction of recombinant human Hsp70 with the model phospholipid membranes. We have confirmed that Hsp70 has strong specificity toward membranes composed of negatively charged phosphatidylserine (PS), compared to neutral phosphatidylcholine membranes. Using differential scanning calorimetry, we have shown for the first time that Hsp70 affects the thermotropic behavior of saturated PS and leads to the interdigitation that controls membrane thickness and rigidity. Hsp70-PS interaction depended on the lipid phase state; the protein stabilized ordered domains enriched with high-melting PS, increasing their area, probably due to formation of quasi-interdigitated phase. Moreover, the ability of Hsp70 to form ion-permeable pores in PS membranes may also be determined by the bilayer thickness. These observations contribute to a better understanding of Hsp70-PS interaction and biological functions of membrane-bound Hsp70 in cancer cells.
Collapse
Affiliation(s)
- Ruslana Tagaeva
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, Saint Petersburg, 197341, Russia
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave. 4, Saint Petersburg, 194064, Russia
| | - Svetlana Efimova
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave. 4, Saint Petersburg, 194064, Russia
| | - Alexander Ischenko
- Saint-Petersburg Pasteur Institute, Mira Str. 14, Saint Petersburg, 197101, Russia
| | - Alexander Zhakhov
- Saint-Petersburg Pasteur Institute, Mira Str. 14, Saint Petersburg, 197101, Russia
| | - Maxim Shevtsov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, Saint Petersburg, 197341, Russia.
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave. 4, Saint Petersburg, 194064, Russia.
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Olga Ostroumova
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave. 4, Saint Petersburg, 194064, Russia
| |
Collapse
|
7
|
Kim SH, Kahng DY, Kim KS, Min HJ. Extracellular Hsp70 Is Involved in the CXCL12/CXCR4 Pathway in Primary Human Nasal Epithelial Cells: A Preliminary Study. JOURNAL OF RHINOLOGY 2023; 30:135-138. [PMID: 39664952 PMCID: PMC11524344 DOI: 10.18787/jr.2023.00042] [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: 06/09/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 12/13/2024] Open
Abstract
Background and Objectives To date, no studies have been conducted on the interaction between extracellular heat shock protein 70 (Hsp70) and C-X-C chemokine receptor type 4 (CXCR4) in the upper airway. We aimed to evaluate the relationship between extracellular Hsp70 and CXCR4 and their role in the primary human nasal epithelium. Methods We cultured primary human nasal epithelial (HNE) cells in an air-liquid interface. Macrogen performed single-cell quantitative polymerase chain reaction and sequencing. We conducted western blot analysis for the CXCR4 and mitogen-activated protein kinase (MAPK) pathways. Results Extracellular Hsp70 treatment significantly increased the genetic expression and protein levels of CXCR4 in primary HNE cells. Phospho-ERK expression was increased by cotreatment with Hsp70 and CXCL12, but inhibited by pretreatment with AMD3100, a CXCR4 inhibitor. Pretreatment with an anti-Hsp70 antibody reduced phospho-ERK expression upregulation induced by cotreatment with Hsp70 and CXCL12. Conclusion Extracellular Hsp70 participates in the activation of the CXCR4-dependent downstream signaling pathway in HNE cells. Further studies should evaluate the extracellular Hsp70-CXCL12/CXCR4 axis and the role of its components in the development of inflammatory diseases.
Collapse
Affiliation(s)
- Seong Hee Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Dong Young Kahng
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Kyung Soo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jin Min
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
Skandalis SS. CD44 Intracellular Domain: A Long Tale of a Short Tail. Cancers (Basel) 2023; 15:5041. [PMID: 37894408 PMCID: PMC10605500 DOI: 10.3390/cancers15205041] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
CD44 is a single-chain transmembrane receptor that exists in multiple forms due to alternative mRNA splicing and post-translational modifications. CD44 is the main cell surface receptor of hyaluronan as well as other extracellular matrix molecules, cytokines, and growth factors that play important roles in physiological processes (such as hematopoiesis and lymphocyte homing) and the progression of various diseases, the predominant one being cancer. Currently, CD44 is an established cancer stem cell marker in several tumors, implying a central functional role in tumor biology. The present review aims to highlight the contribution of the CD44 short cytoplasmic tail, which is devoid of any enzymatic activity, in the extraordinary functional diversity of the receptor. The interactions of CD44 with cytoskeletal proteins through specific structural motifs within its intracellular domain drives cytoskeleton rearrangements and affects the distribution of organelles and transport of molecules. Moreover, the CD44 intracellular domain specifically interacts with various cytoplasmic effectors regulating cell-trafficking machinery, signal transduction pathways, the transcriptome, and vital cell metabolic pathways. Understanding the cell type- and context-specificity of these interactions may unravel the high complexity of CD44 functions and lead to novel improved therapeutic interventions.
Collapse
Affiliation(s)
- Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
9
|
Garcia C, Andersen CJ, Blesso CN. The Role of Lipids in the Regulation of Immune Responses. Nutrients 2023; 15:3899. [PMID: 37764683 PMCID: PMC10535783 DOI: 10.3390/nu15183899] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Lipid metabolism plays a major role in the regulation of the immune system. Exogenous (dietary and microbial-derived) and endogenous (non-microbial-derived) lipids play a direct role in regulating immune cell activation, differentiation and expansion, and inflammatory phenotypes. Understanding the complexities of lipid-immune interactions may have important implications for human health, as certain lipids or immune pathways may be beneficial in circumstances of acute infection yet detrimental in chronic inflammatory diseases. Further, there are key differences in the lipid effects between specific immune cell types and location (e.g., gut mucosal vs. systemic immune cells), suggesting that the immunomodulatory properties of lipids may be tissue-compartment-specific, although the direct effect of dietary lipids on the mucosal immune system warrants further investigation. Importantly, there is recent evidence to suggest that lipid-immune interactions are dependent on sex, metabolic status, and the gut microbiome in preclinical models. While the lipid-immune relationship has not been adequately established in/translated to humans, research is warranted to evaluate the differences in lipid-immune interactions across individuals and whether the optimization of lipid-immune interactions requires precision nutrition approaches to mitigate or manage disease. In this review, we discuss the mechanisms by which lipids regulate immune responses and the influence of dietary lipids on these processes, highlighting compelling areas for future research.
Collapse
Affiliation(s)
| | | | - Christopher N. Blesso
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (C.G.); (C.J.A.)
| |
Collapse
|
10
|
Matthews CEP, Fussner LA, Yaeger M, Aloor JJ, Reece SW, Kilburg-Basnyat BJ, Varikuti S, Luo B, Inks M, Sergin S, Schmidt CA, Neufer PD, Pennington ER, Fisher-Wellman KH, Chowdhury SM, Fessler MB, Fenton JI, Anderson EJ, Shaikh SR, Gowdy KM. The prohibitin complex regulates macrophage fatty acid composition, plasma membrane packing, and lipid raft-mediated inflammatory signaling. Prostaglandins Leukot Essent Fatty Acids 2023; 190:102540. [PMID: 36706677 PMCID: PMC9992117 DOI: 10.1016/j.plefa.2023.102540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/28/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
Prohibitins (PHB1 and PHB2) are ubiquitously expressed proteins which play critical roles in multiple biological processes, and together form the ring-like PHB complex found in phospholipid-rich cellular compartments including lipid rafts. Recent studies have implicated PHB1 as a mediator of fatty acid transport as well as a membrane scaffold mediating B lymphocyte and mast cell signal transduction. However, the specific role of PHBs in the macrophage have not been characterized, including their role in fatty acid uptake and lipid raft-mediated inflammatory signaling. We hypothesized that the PHB complex regulates macrophage inflammatory signaling through the formation of lipid rafts. To evaluate our hypothesis, RAW 264.7 macrophages were transduced with shRNA against PHB1, PHB2, or scrambled control (Scr), and then stimulated with lipopolysaccharide (LPS) or tumor necrosis factor-alpha (TNF-α), which activate lipid raft-dependent receptor signaling (CD14/TLR4 and TNFR1, respectively). PHB1 knockdown was lethal, whereas PHB2 knockdown (PHB2kd), which also resulted in decreased PHB1 expression, led to attenuated nuclear factor-kappa-B (NF-κB) activation and subsequent cytokine and chemokine production. PHB2kd macrophages also had decreased cell surface TNFR1, CD14, TLR4, and lipid raft marker ganglioside GM1 at baseline and post-stimuli. Post-LPS, PHB2kd macrophages did not increase the concentration of cellular saturated, monounsaturated, and polyunsaturated fatty acids. This was accompanied by decreased lipid raft formation and modified plasma membrane molecular packing, further supporting the PHB complex's importance in lipid raft formation. Taken together, these data suggest a critical role for PHBs in regulating macrophage inflammatory signaling via maintenance of fatty acid composition and lipid raft structure. SUMMARY: Prohibitins are proteins found in phospholipid-rich cellular compartments, including lipid rafts, that play important roles in signaling, transcription, and multiple other cell functions. Macrophages are key cells in the innate immune response and the presence of membrane lipid rafts is integral to signal transduction, but the role of prohibitins in macrophage lipid rafts and associated signaling is unknown. To address this question, prohibitin knockdown macrophages were generated and responses to lipopolysaccharide and tumor necrosis factor-alpha, which act through lipid raft-dependent receptors, were analyzed. Prohibitin knockdown macrophages had significantly decreased cytokine and chemokine production, transcription factor activation, receptor expression, lipid raft assembly and membrane packing, and altered fatty acid remodeling. These data indicate a novel role for prohibitins in macrophage inflammatory signaling through regulation of fatty acid composition and lipid raft formation.
Collapse
Affiliation(s)
- Christine E Psaltis Matthews
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Lynn A Fussner
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Michael Yaeger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Jim J Aloor
- Diabetes and Obesity Institute, Department of Physiology, East Carolina University, Greenville, NC, United States
| | - Sky W Reece
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Brita J Kilburg-Basnyat
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Sanjay Varikuti
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Bin Luo
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Morgan Inks
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Selin Sergin
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Cameron A Schmidt
- Diabetes and Obesity Institute, Department of Physiology, East Carolina University, Greenville, NC, United States
| | - P Darrell Neufer
- Diabetes and Obesity Institute, Department of Physiology, East Carolina University, Greenville, NC, United States
| | - Edward Ross Pennington
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Kelsey H Fisher-Wellman
- Diabetes and Obesity Institute, Department of Physiology, East Carolina University, Greenville, NC, United States
| | - Saiful M Chowdhury
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, TX, United States
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, United States
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Ethan J Anderson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, FOE Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
11
|
Zhou X, Jin S, Pan J, Lin Q, Yang S, Lu Y, Qiu M, Ambe PC, Basharat Z, Zimmer V, Wang W, Hong W. Relationship between Cholesterol-Related Lipids and Severe Acute Pancreatitis: From Bench to Bedside. J Clin Med 2023; 12:jcm12051729. [PMID: 36902516 PMCID: PMC10003000 DOI: 10.3390/jcm12051729] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
It is well known that hypercholesterolemia in the body has pro-inflammatory effects through the formation of inflammasomes and augmentation of TLR (Toll-like receptor) signaling, which gives rise to cardiovascular disease and neurodegenerative diseases. However, the interaction between cholesterol-related lipids and acute pancreatitis (AP) has not yet been summarized before. This hinders the consensus on the existence and clinical importance of cholesterol-associated AP. This review focuses on the possible interaction between AP and cholesterol-related lipids, which include total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) and apolipoprotein (Apo) A1, from the bench to the bedside. With a higher serum level of total cholesterol, LDL-C is associated with the severity of AP, while the persistent inflammation of AP is allied with a decrease in serum levels of cholesterol-related lipids. Therefore, an interaction between cholesterol-related lipids and AP is postulated. Cholesterol-related lipids should be recommended as risk factors and early predictors for measuring the severity of AP. Cholesterol-lowering drugs may play a role in the treatment and prevention of AP with hypercholesterolemia.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Shengchun Jin
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jingyi Pan
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qingyi Lin
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Shaopeng Yang
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yajing Lu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Minhao Qiu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Peter C. Ambe
- Department of General Surgery, Visceral Surgery and Coloproctology, Vinzenz-Pallotti-Hospital Bensberg, Vinzenz-Pallotti-Str. 20–24, 51429 Bensberg, Germany
| | - Zarrin Basharat
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Vincent Zimmer
- Department of Medicine, Marienhausklinik St. Josef Kohlhof, 66539 Neunkirchen, Germany
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Wandong Hong
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Correspondence: ; Tel./Fax: +86-0577-55579122
| |
Collapse
|
12
|
Research progress on the role of cholesterol in hepatocellular carcinoma. Eur J Pharmacol 2022; 938:175410. [DOI: 10.1016/j.ejphar.2022.175410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
|
13
|
Chávez-Aguilar LA, Ávila-Castro D, Merino-Pasaye LE, Peña-Vélez R. Children With Asparaginase-associated Pancreatitis Present Elevated Levels of Insulin, Total Cholesterol, and HOMA-IR Before Starting Acute Lymphoblastic Leukemia Treatment. J Pediatr Hematol Oncol 2022; 44:342-344. [PMID: 34966097 DOI: 10.1097/mph.0000000000002368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/19/2021] [Indexed: 11/26/2022]
Abstract
Asparaginase-associated pancreatitis frequently occurs in children with cancer. It is unknown if other factors can influence the development of pancreatitis. A total of 33 pediatric patients with a confirmed diagnosis of acute lymphoblastic leukemia were included in this study. Before acute lymphoblastic leukemia drug treatment, the metabolic parameters (glucose, insulin, homeostasis model assessment insulin resistance, total cholesterol, triglycerides) and body mass index percentile were compared. Children who had acute pancreatitis had higher levels of insulin, homeostasis model assessment insulin resistance, and total cholesterol, compared with children who did not develop acute pancreatitis. These metabolic alterations could play a role in the development of pancreatitis.
Collapse
Affiliation(s)
| | - David Ávila-Castro
- Department of Pediatric Hematology, Centro Médico Nacional 20 de Noviembre, Mexico City
| | - Laura E Merino-Pasaye
- Department of Pediatric Hematology, Centro Médico Nacional 20 de Noviembre, Mexico City
| | - Rubén Peña-Vélez
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Hospital General de Puebla "Dr Eduardo Vázquez N", Puebla, Mexico
| |
Collapse
|
14
|
Immunometabolic rewiring of tubular epithelial cells in kidney disease. Nat Rev Nephrol 2022; 18:588-603. [PMID: 35798902 DOI: 10.1038/s41581-022-00592-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 12/20/2022]
Abstract
Kidney tubular epithelial cells (TECs) have a crucial role in the damage and repair response to acute and chronic injury. To adequately respond to constant changes in the environment, TECs have considerable bioenergetic needs, which are supported by metabolic pathways. Although little is known about TEC metabolism, a number of ground-breaking studies have shown that defective glucose metabolism or fatty acid oxidation in the kidney has a key role in the response to kidney injury. Imbalanced use of these metabolic pathways can predispose TECs to apoptosis and dedifferentiation, and contribute to lipotoxicity and kidney injury. The accumulation of lipids and aberrant metabolic adaptations of TECs during kidney disease can also be driven by receptors of the innate immune system. Similar to their actions in innate immune cells, pattern recognition receptors regulate the metabolic rewiring of TECs, causing cellular dysfunction and lipid accumulation. TECs should therefore be considered a specialized cell type - like cells of the innate immune system - that is subject to regulation by immunometabolism. Targeting energy metabolism in TECs could represent a strategy for metabolically reprogramming the kidney and promoting kidney repair.
Collapse
|
15
|
Horlock AD, Ormsby TJR, Clift MJD, Santos JEP, Bromfield JJ, Sheldon IM. Cholesterol supports bovine granulosa cell inflammatory responses to lipopolysaccharide. Reproduction 2022; 164:109-123. [PMID: 35900358 DOI: 10.1530/rep-22-0032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/14/2022] [Indexed: 11/08/2022]
Abstract
During bacterial infections of the bovine uterus or mammary gland, ovarian granulosa cells mount inflammatory responses to lipopolysaccharide (LPS). In vitro, LPS stimulates granulosa cell secretion of the cytokines IL-1α and IL-1β, and the chemokine IL-8. These LPS-stimulated inflammatory responses depend on culturing granulosa cells with serum, but the mechanism is unclear. Here we tested the hypothesis that cholesterol supports inflammatory responses to LPS in bovine granulosa cells. We used granulosa cells isolated from 4-8 mm and > 8.5 mm diameter ovarian follicles and manipulated the availability of cholesterol. We found that serum or follicular fluid containing cholesterol increased LPS-stimulated secretion of IL-1α and IL-1β from granulosa cells. Conversely, depleting cholesterol using methyl-β-cyclodextrin diminished LPS-stimulated secretion of IL-1α, IL-1β and IL-8 from granulosa cells cultured in serum. Follicular fluid contained more high-density lipoprotein cholesterol than low-density lipoprotein cholesterol, and granulosa cells expressed the receptor for high-density lipoprotein, scavenger receptor class B member 1 (SCARB1). Furthermore, culturing granulosa cells with high-density lipoprotein cholesterol, but not low-density lipoprotein or very low-density lipoprotein cholesterol, increased LPS-stimulated inflammation in granulosa cells. Cholesterol biosynthesis also played a role in granulosa cell inflammation because RNA interference of mevalonate pathway enzymes inhibited LPS-stimulated inflammation. Finally, treatment with follicle-stimulating hormone, but not luteinizing hormone, increased LPS-stimulated granulosa cell inflammation, and follicle-stimulating hormone increased SCARB1 protein. However, changes in inflammation were not associated with changes in oestradiol or progesterone secretion. Taken together these findings imply that cholesterol supports inflammatory responses to LPS in granulosa cells.
Collapse
Affiliation(s)
- Anthony D Horlock
- A Horlock, Swansea University Medical School, Swansea University, Swansea, United Kingdom of Great Britain and Northern Ireland
| | - Thomas J R Ormsby
- T Ormsby, Swansea University Medical School, Swansea University, Swansea, United Kingdom of Great Britain and Northern Ireland
| | - Martin J D Clift
- M Clift, Swansea University Medical School, Swansea University, Swansea, United Kingdom of Great Britain and Northern Ireland
| | - Jose E P Santos
- J Santos, Department of Animal Sciences, University of Florida, Gainesville, United States
| | - John J Bromfield
- J Bromfield, Department of Animal Sciences, University of Florida, Gainesville, United States
| | - Iain Martin Sheldon
- I Sheldon, Swansea University Medical School, Swansea University, Swansea, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
16
|
Woo JH, Park SJ, Park SM, Joe E, Jou I. Interleukin‐6 signaling requires EHD1‐mediated alteration of membrane rafts. FEBS J 2022; 289:5914-5932. [DOI: 10.1111/febs.16458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/25/2022] [Accepted: 04/12/2022] [Indexed: 01/09/2023]
Affiliation(s)
- Joo Hong Woo
- Inflamm‐aging Translational Research Center Ajou University School of Medicine Suwon Korea
| | - Soo Jung Park
- Inflamm‐aging Translational Research Center Ajou University School of Medicine Suwon Korea
| | - Sang Myun Park
- Department of Pharmacology Ajou University School of Medicine Suwon Korea
- Center for Convergence Research of Neurological Disorders Ajou University School of Medicine Suwon Korea
| | - Eun‐hye Joe
- Department of Pharmacology Ajou University School of Medicine Suwon Korea
- Center for Convergence Research of Neurological Disorders Ajou University School of Medicine Suwon Korea
| | - Ilo Jou
- Inflamm‐aging Translational Research Center Ajou University School of Medicine Suwon Korea
- Department of Pharmacology Ajou University School of Medicine Suwon Korea
| |
Collapse
|
17
|
Yang DJ, Lu HM, Liu Y, Li M, Hu WM, Zhou ZG. Development and validation of a prediction model for moderately severe and severe acute pancreatitis in pregnancy. World J Gastroenterol 2022; 28:1588-1600. [PMID: 35582133 PMCID: PMC9048464 DOI: 10.3748/wjg.v28.i15.1588] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/02/2022] [Accepted: 03/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The severity of acute pancreatitis in pregnancy (APIP) is correlated with higher risks of maternal and fetal death.
AIM To develop a nomogram that could predict moderately severe and severe acute pancreatitis in pregnancy (MSIP).
METHODS Patients with APIP admitted to West China Hospital between January 2012 and December 2018 were included in this study. They were divided into mild acute pancreatitis in pregnancy (MAIP) and MSIP. Characteristic parameters and laboratory results were collected. The training set and test set were randomly divided at a ratio of 7:3. Least absolute shrinkage and selection operator regression was used to select potential prognostic factors. A nomogram was developed by logistic regression. A random forest model was used to validate the stability of the prediction factors. Receiver operating characteristic curves and calibration curves were used to evaluate the model’s predictive performance.
RESULTS A total of 190 patients were included in this study. A total of 134 patients (70.5%) and 56 patients (29.5%) were classified as having MAIP and MSIP, respectively. Four independent predictors (lactate dehydrogenase, triglyceride, cholesterol, and albumin levels) were identified for MSIP. A nomogram prediction model based on these factors was established. The model had areas under the curve of 0.865 and 0.853 in the training and validation sets, respectively. The calibration curves showed that the nomogram has a good consistency.
CONCLUSION A nomogram including lactate dehydrogenase, triglyceride, cholesterol, and albumin levels as independent predictors was built with good performance for MSIP prediction.
Collapse
Affiliation(s)
- Du-Jiang Yang
- Department of Gastroenterological Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hui-Min Lu
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yong Liu
- Department of Gastroenterological Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mao Li
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wei-Ming Hu
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zong-Guang Zhou
- Department of Gastroenterological Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
18
|
Charó N, Jerez H, Tatti S, Romero EL, Schattner M. The Anti-Inflammatory Effect of Nanoarchaeosomes on Human Endothelial Cells. Pharmaceutics 2022; 14:736. [PMID: 35456570 PMCID: PMC9027062 DOI: 10.3390/pharmaceutics14040736] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 01/14/2023] Open
Abstract
Archaebacterias are considered a unique source of novel biomaterials of interest for nanomedicine. In this perspective, the effects of nanoarchaeosomes (ARC), which are nanovesicles prepared from polar lipids extracted from the extreme halophilic Halorubrum tebenquinchense, on human umbilical vein endothelial cells (HUVEC) were investigated in physiological and under inflammatory static conditions. Upon incubation, ARC (170 nm mean size, -41 mV ζ) did not affect viability, cell proliferation, and expression of intercellular adhesion molecule-1 (ICAM-1) and E-selectin under basal conditions, but reduced expression of both molecules and secretion of IL-6 induced by lypopolysaccharide (LPS), Pam3CSK4 or Escherichia coli. Such effects were not observed with TNF-α or IL-1β stimulation. Interestingly, ARC significantly decreased basal levels of von Willebrand factor (vWF) and levels induced by all stimuli. None of these parameters was altered by liposomes of hydrogenated phosphatidylcholine and cholesterol of comparable size and concentration. Only ARC were endocytosed by HUVEC and reduced mRNA expression of ICAM-1 and vWF via NF-ĸB and ERK1/2 in LPS-stimulated cells. This is the first report of the anti-inflammatory effect of ARC on endothelial cells and our data suggest that its future use in vascular disease may hopefully be of particular interest.
Collapse
Affiliation(s)
- Nancy Charó
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina;
| | - Horacio Jerez
- Center for Research and Development in Nanomedicines (CIDEN), National University of Quilmes, Roque Saenz Peña, Bernal 1876, Argentina;
| | - Silvio Tatti
- Department of Obstetrics and Gynecology, Clinical Hospital, Av. Córdoba 2351, Buenos Aires 1120, Argentina;
| | - Eder Lilia Romero
- Center for Research and Development in Nanomedicines (CIDEN), National University of Quilmes, Roque Saenz Peña, Bernal 1876, Argentina;
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina;
| |
Collapse
|
19
|
Kulkarni R, Wiemer EAC, Chang W. Role of Lipid Rafts in Pathogen-Host Interaction - A Mini Review. Front Immunol 2022; 12:815020. [PMID: 35126371 PMCID: PMC8810822 DOI: 10.3389/fimmu.2021.815020] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/31/2021] [Indexed: 12/25/2022] Open
Abstract
Lipid rafts, also known as microdomains, are important components of cell membranes and are enriched in cholesterol, glycophospholipids and receptors. They are involved in various essential cellular processes, including endocytosis, exocytosis and cellular signaling. Receptors are concentrated at lipid rafts, through which cellular signaling can be transmitted. Pathogens exploit these signaling mechanisms to enter cells, proliferate and egress. However, lipid rafts also play an important role in initiating antimicrobial responses by sensing pathogens via clustered pathogen-sensing receptors and triggering downstream signaling events such as programmed cell death or cytokine production for pathogen clearance. In this review, we discuss how both host and pathogens use lipid rafts and associated proteins in an arms race to survive. Special attention is given to the involvement of the major vault protein, the main constituent of a ribonucleoprotein complex, which is enriched in lipid rafts upon infection with vaccinia virus.
Collapse
Affiliation(s)
- Rakesh Kulkarni
- Molecular and Cell Biology, Taiwan International Graduate Program, National Defense Medical Center, Academia Sinica and Graduate Institute of Life Science, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- *Correspondence: Rakesh Kulkarni, ; Wen Chang,
| | - Erik A. C. Wiemer
- Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
| | - Wen Chang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- *Correspondence: Rakesh Kulkarni, ; Wen Chang,
| |
Collapse
|
20
|
Sun S, Yao Y, Huang C, Xu H, Zhao Y, Wang Y, Zhu Y, Miao Y, Feng X, Gao X, Zheng J, Zhang Q. CD36 regulates LPS-induced acute lung injury by promoting macrophages M1 polarization. Cell Immunol 2022; 372:104475. [PMID: 35063904 DOI: 10.1016/j.cellimm.2021.104475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 12/02/2021] [Accepted: 12/31/2021] [Indexed: 01/11/2023]
|
21
|
Chatterjee R, Chowdhury AR, Mukherjee D, Chakravortty D. Lipid larceny: channelizing host lipids for establishing successful pathogenesis by bacteria. Virulence 2021; 12:195-216. [PMID: 33356849 PMCID: PMC7808437 DOI: 10.1080/21505594.2020.1869441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 12/03/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022] Open
Abstract
Lipids are complex organic compounds made up of carbon, oxygen, and hydrogen. These play a diverse and intricate role in cellular processes like membrane trafficking, protein sorting, signal transduction, and bacterial infections. Both Gram-positive bacteria (Staphylococcus sp., Listeria monocytogenes, etc.) and Gram-negative bacteria (Chlamydia sp., Salmonella sp., E. coli, etc.) can hijack the various host-lipids and utilize them structurally as well as functionally to mount a successful infection. The pathogens can deploy with various arsenals to exploit host membrane lipids and lipid-associated receptors as an attachment for toxins' landing or facilitate their entry into the host cellular niche. Bacterial species like Mycobacterium sp. can also modulate the host lipid metabolism to fetch its carbon source from the host. The sequential conversion of host membrane lipids into arachidonic acid and prostaglandin E2 due to increased activity of cPLA-2 and COX-2 upon bacterial infection creates immunosuppressive conditions and facilitates the intracellular growth and proliferation of bacteria. However, lipids' more debatable role is that they can also be a blessing in disguise. Certain host-lipids, especially sphingolipids, have been shown to play a crucial antibacterial role and help the host in combating the infections. This review shed light on the detailed role of host lipids in bacterial infections and the current understanding of the lipid in therapeutics. We have also discussed potential prospects and the need of the hour to help us cope in this race against deadly pathogens and their rapidly evolving stealthy virulence strategies.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Atish Roy Chowdhury
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Debapriya Mukherjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
22
|
Souza TN, Valdez AF, Rizzo J, Zamith-Miranda D, Guimarães AJ, Nosanchuk JD, Nimrichter L. Host cell membrane microdomains and fungal infection. Cell Microbiol 2021; 23:e13385. [PMID: 34392593 PMCID: PMC8664998 DOI: 10.1111/cmi.13385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/14/2021] [Accepted: 07/24/2021] [Indexed: 01/13/2023]
Abstract
Lipid microdomains or lipid rafts are dynamic and tightly ordered regions of the plasma membrane. In mammalian cells, they are enriched in cholesterol, glycosphingolipids, Glycosylphosphatidylinositol-anchored and signalling-related proteins. Several studies have suggested that mammalian pattern recognition receptors are concentrated or recruited to lipid domains during host-pathogen association to enhance the effectiveness of host effector processes. However, pathogens have also evolved strategies to exploit these domains to invade cells and survive. In fungal organisms, a complex cell wall network usually mediates the first contact with the host cells. This cell wall may contain virulence factors that interfere with the host membrane microdomains dynamics, potentially impacting the infection outcome. Indeed, the microdomain disruption can dampen fungus-host cell adhesion, phagocytosis and cellular immune responses. Here, we provide an overview of regulatory strategies employed by pathogenic fungi to engage with and potentially subvert the lipid microdomains of host cells. TAKE AWAY: Lipid microdomains are ordered regions of the plasma membrane enriched in cholesterol, glycosphingolipids (GSL), GPI-anchored and signalling-related proteins. Pathogen recognition by host immune cells can involve lipid microdomain participation. During this process, these domains can coalesce in larger complexes recruiting receptors and signalling proteins, significantly increasing their signalling abilities. The antifungal innate immune response is mediated by the engagement of pathogen-associated molecular patterns to pattern recognition receptors (PRRs) at the plasma membrane of innate immune cells. Lipid microdomains can concentrate or recruit PRRs during host cell-fungi association through a multi-interactive mechanism. This association can enhance the effectiveness of host effector processes. However, virulence factors at the fungal cell surface and extracellular vesicles can re-assembly these domains, compromising the downstream signalling and favouring the disease development. Lipid microdomains are therefore very attractive targets for novel drugs to combat fungal infections.
Collapse
Affiliation(s)
- Taiane N Souza
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F Valdez
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Rizzo
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, Paris, France
| | - Daniel Zamith-Miranda
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Allan Jefferson Guimarães
- Departamento de Microbiologia e Parasitologia-MIP, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Joshua D Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leonardo Nimrichter
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Yang SC. A New Perspective on Fish Oil: The Prevention of Alcoholic Liver Disease. J Oleo Sci 2021; 70:1531-1538. [PMID: 34732632 DOI: 10.5650/jos.ess21216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mechanisms of alcoholic liver diseases (ALD) are very complex and interrelated, including abnormal lipid metabolism, oxidative stress, and gut-derived endotoxin pathway. On the other hand, fish oil is rich in n-3 polyunsaturated fatty acids (PUFAs), such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which decrease blood triglyceride concentration in hypertriglycemia patients and show protective effects against fatty liver. However, there is limited evidence from studies of the relationship between fish oil and ALD based on the viewpoint of the intestinal integrity and microflora. Therefore, this review discusses the mechanism of amelioration for ALD by fish oil. Based on our previous studies, partial replacement of olive oil by fish oil in alcohol-containing liquid diet ameliorated the liver damage including fatty liver and inflammation in rats. Based on these results, the mechanisms of hepatoprotective effects due to fish oil substitution were discussed in three parts, such as regulating lipid metabolism, decreasing oxidative stress and maintaining intestinal health. First of all, we found that fish oil substitution increased plasma adiponectin levels, and then increasing MCAD and CPT-1 mRNA levels to accelerate fatty acid oxidation in liver, then further prevent ethanol-induced hepatosteatosis in rats with chronic alcohol-feeding. Fish oil replacement also enhanced hepatic autophagy flux, which enhanced lipid degradation, then inhibited lipid accumulation in liver. Secondly, the appreciable proportion of fish oil decreased lipid peroxidation by reducing the protein expression of cytochrome p450 2E1 in chronic alcohol-feeding rats. We also speculated that the appropriate proportion of n-6 and n-3 PUFAs is very important for preventing alcoholic liver disease. At last, substituting fish oil for olive oil normalized the intestinal permeability and fecal microbiota composition, thus providing a low plasma endotoxin level and inflammatory responses, which exert ameliorative effects on ethanol-induced liver injuries in rats.
Collapse
Affiliation(s)
- Suh-Ching Yang
- School of Nutrition and Health Sciences, Taipei Medical University.,Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University.,Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University.,School of Gerontology Health Management, College of Nursing, Taipei Medical University.,Nutrition Research Center, Taipei Medical University Hospital
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW This study reviews the mechanisms of HDL cholesterol immunomodulation in the context of the mechanisms of chronic inflammation and immunosuppression causing persistent inflammation, immunosuppression and catabolism syndrome (PICS) and describes potential therapies and gaps in current research. RECENT FINDINGS Low HDL cholesterol is predictive of acute sepsis severity and outcome. Recent research has indicated apolipoprotein is a prognostic indicator of long-term outcomes. The pathobiologic mechanisms of PICS have been elucidated in the past several years. Recent research of the interaction of HDL pathways in related chronic inflammatory diseases may provide insights into further mechanisms and therapeutic targets. SUMMARY HDL significantly influences innate and adaptive immune pathways relating to chronic disease and inflammation. Further research is needed to better characterize these interactions in the setting of PICS.
Collapse
Affiliation(s)
- Grant Barker
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Julia R Winer
- University of Florida College of Medicine, Gainesville, Florida
| | - Faheem W Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Srinivasa Reddy
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| |
Collapse
|
25
|
Yokoyama N, Hanafusa K, Hotta T, Oshima E, Iwabuchi K, Nakayama H. Multiplicity of Glycosphingolipid-Enriched Microdomain-Driven Immune Signaling. Int J Mol Sci 2021; 22:9565. [PMID: 34502474 PMCID: PMC8430928 DOI: 10.3390/ijms22179565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
Glycosphingolipids (GSLs), together with cholesterol, sphingomyelin (SM), and glycosylphosphatidylinositol (GPI)-anchored and membrane-associated signal transduction molecules, form GSL-enriched microdomains. These specialized microdomains interact in a cis manner with various immune receptors, affecting immune receptor-mediated signaling. This, in turn, results in the regulation of a broad range of immunological functions, including phagocytosis, cytokine production, antigen presentation and apoptosis. In addition, GSLs alone can regulate immunological functions by acting as ligands for immune receptors, and exogenous GSLs can alter the organization of microdomains and microdomain-associated signaling. Many pathogens, including viruses, bacteria and fungi, enter host cells by binding to GSL-enriched microdomains. Intracellular pathogens survive inside phagocytes by manipulating intracellular microdomain-driven signaling and/or sphingolipid metabolism pathways. This review describes the mechanisms by which GSL-enriched microdomains regulate immune signaling.
Collapse
Affiliation(s)
- Noriko Yokoyama
- Institute for Environmental and Gender-Specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan; (N.Y.); (K.H.); (T.H.); (E.O.); (K.I.)
| | - Kei Hanafusa
- Institute for Environmental and Gender-Specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan; (N.Y.); (K.H.); (T.H.); (E.O.); (K.I.)
| | - Tomomi Hotta
- Institute for Environmental and Gender-Specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan; (N.Y.); (K.H.); (T.H.); (E.O.); (K.I.)
| | - Eriko Oshima
- Institute for Environmental and Gender-Specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan; (N.Y.); (K.H.); (T.H.); (E.O.); (K.I.)
| | - Kazuhisa Iwabuchi
- Institute for Environmental and Gender-Specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan; (N.Y.); (K.H.); (T.H.); (E.O.); (K.I.)
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Chiba 279-0023, Japan
- Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, Urayasu, Chiba 279-0023, Japan
| | - Hitoshi Nakayama
- Institute for Environmental and Gender-Specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan; (N.Y.); (K.H.); (T.H.); (E.O.); (K.I.)
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Chiba 279-0023, Japan
- Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, Urayasu, Chiba 279-0023, Japan
| |
Collapse
|
26
|
Olona A, Hateley C, Muralidharan S, Wenk MR, Torta F, Behmoaras J. Sphingolipid metabolism during Toll-like receptor 4 (TLR4)-mediated macrophage activation. Br J Pharmacol 2021; 178:4575-4587. [PMID: 34363204 DOI: 10.1111/bph.15642] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/18/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophage activation in response to stimulation of Toll-like receptor 4 (TLR4) provides a paradigm for investigating energy metabolism that regulates the inflammatory response. TLR4-mediated pro-inflammatory macrophage activation is characterized by increased glycolysis and altered mitochondrial metabolism, supported by selective amino acid uptake and/or usage. Fatty acid metabolism remains as a highly complex rewiring that accompanies classical macrophage activation. TLR4 activation leads to de novo synthesis of fatty acids, which flux into sphingolipids, complex lipids that form the building blocks of eukaryotic cell membranes and regulate cell function. Here, we review the importance of TLR4-mediated de novo synthesis of membrane sphingolipids in macrophages. We first highlight fatty acid metabolism during TLR4-driven macrophage immunometabolism. We then focus on the temporal dynamics of sphingolipid biosynthesis and emphasize the modulatory role of some sphingolipid species (i.e. sphingomyelins, ceramides and glycosphingolipids) on the pro-inflammatory and pro-resolution phases of LPS/TLR4 activation in macrophages.
Collapse
Affiliation(s)
- Antoni Olona
- Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Charlotte Hateley
- Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | | | - Markus R Wenk
- SLING, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Federico Torta
- SLING, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jacques Behmoaras
- Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, UK.,Programme in Cardiovascular and Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School Singapore, Republic of Singapore
| |
Collapse
|
27
|
Rodriguez-Coira J, Villaseñor A, Izquierdo E, Huang M, Barker-Tejeda TC, Radzikowska U, Sokolowska M, Barber D. The Importance of Metabolism for Immune Homeostasis in Allergic Diseases. Front Immunol 2021; 12:692004. [PMID: 34394086 PMCID: PMC8355700 DOI: 10.3389/fimmu.2021.692004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022] Open
Abstract
There is increasing evidence that the metabolic status of T cells and macrophages is associated with severe phenotypes of chronic inflammation, including allergic inflammation. Metabolic changes in immune cells have a crucial role in their inflammatory or regulatory responses. This notion is reinforced by metabolic diseases influencing global energy metabolism, such as diabetes or obesity, which are known risk factors of severity in inflammatory conditions, due to the metabolic-associated inflammation present in these patients. Since several metabolic pathways are closely tied to T cell and macrophage differentiation, a better understanding of metabolic alterations in immune disorders could help to restore and modulate immune cell functions. This link between energy metabolism and inflammation can be studied employing animal, human or cellular models. Analytical approaches rank from classic immunological studies to integrated analysis of metabolomics, transcriptomics, and proteomics. This review summarizes the main metabolic pathways of the cells involved in the allergic reaction with a focus on T cells and macrophages and describes different models and platforms of analysis used to study the immune system and its relationship with metabolism.
Collapse
Affiliation(s)
- Juan Rodriguez-Coira
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Alma Villaseñor
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Elena Izquierdo
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Tomás Clive Barker-Tejeda
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Domingo Barber
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| |
Collapse
|
28
|
Sun M, Han X, Zhou D, Zhong J, Liu L, Wang Y, Ni J, Shen X, Liang C, Fang H. BIG1 mediates sepsis-induced lung injury by modulating lipid raft-dependent macrophage inflammatory responses. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1088-1097. [PMID: 34153089 DOI: 10.1093/abbs/gmab085] [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: 02/26/2021] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome with high mortality. It has been reported that brefeldin A-inhibited guanine nucleotide-exchange factor 1 (BIG1) is involved in the pathogenesis of sepsis. However, the mechanism is not fully elucidated. In the present study, we explored the role of BIG1 in mediating lipid raft-dependent macrophage inflammatory response and its impact on lung injury in murine sepsis. In vitro studies revealed that BIG1 deficiency reduces the upregulation and secretion of tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and IL-1β and inhibits the activation of the toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88-dependent nuclear factor kappa-B signaling pathway induced by the lipopolysaccharide (LPS) treatment. Further experiments revealed that the inhibitory effects of BIG1 deficiency on LPS-induced inflammation are due to the upregulation of adenosine triphosphate-binding cassette transporter A1. This promotes the free-cholesterol efflux from lipid rafts and results in the reduction of lipid raft TLR4 content. The decrease in TLR4 content in lipid raft thereby inhibits the LPS-induced inflammatory response. Furthermore, using the cecal ligation and puncture-induced polymicrobial sepsis mouse model, we found that conditional knockout (cKO) of the myeloid cell BIG1 significantly reduced the serum concentrations of TNF-α, IL-6, and IL-1β, and downregulated their mRNA expressions in the lungs. Pathological analysis confirmed that the BIG1 cKO alleviated the sepsis-induced lung injury. These results revealed the crucial new role of BIG1 in mediating lipid raft-dependent macrophage inflammatory response. Hence, BIG1 may be a potential promising therapeutic target for the treatment of septic lung injury.
Collapse
Affiliation(s)
- Minli Sun
- Department of Anesthesiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200031, China
| | - Xiaodan Han
- Department of Anesthesiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200031, China
| | - Di Zhou
- Department of Anesthesiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200031, China
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200031, China
| | - Lixin Liu
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 200032, China
| | - Yirui Wang
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 200032, China
| | - Jiahui Ni
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 200032, China
| | - Xiaoyan Shen
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 200032, China
| | - Chao Liang
- Department of Anesthesiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200031, China
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200031, China
| |
Collapse
|
29
|
Cortés HD, Gómez FA, Marshall SH. The Phagosome-Lysosome Fusion Is the Target of a Purified Quillaja saponin Extract (PQSE) in Reducing Infection of Fish Macrophages by the Bacterial Pathogen Piscirickettsia salmonis. Antibiotics (Basel) 2021; 10:antibiotics10070847. [PMID: 34356768 PMCID: PMC8300623 DOI: 10.3390/antibiotics10070847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/20/2022] Open
Abstract
Piscirickettsia salmonis, the etiological agent of Piscirickettsiosis, is a Gram-negative and facultative intracellular pathogen that has affected the Chilean salmon industry since 1989. The bacterium is highly aggressive and can survive and replicate within fish macrophages using the Dot/Icm secretion system to evade the host’s immune response and spread systemically. To date, no efficient control measures have been developed for this disease; therefore, the producers use large amounts of antibiotics to control this pathogen. In this frame, this work has focused on evaluating the use of saponins from Quillaja saponaria as a new alternative to control the Piscirickettsiosis. It has been previously reported that purified extract of Q. saponaria (PQSE) displays both antimicrobial activity against pathogenic bacteria and viruses and adjuvant properties. Our results show that PQSE does not present antimicrobial activity against P. salmonis, although it reduces P. salmonis infection in an in vitro model, promoting the phagosome–lysosome fusion. Additionally, we demonstrate that PQSE modulates the expression of IL-12 and IL-10 in infected cells, promoting the immune response against the pathogen and reducing the expression of pathogen virulence genes. These results together strongly argue for specific anti-invasion and anti-intracellular replication effects induced by the PQSE in macrophages.
Collapse
|
30
|
Schromm AB, Paulowski L, Kaconis Y, Kopp F, Koistinen M, Donoghue A, Keese S, Nehls C, Wernecke J, Garidel P, Sevcsik E, Lohner K, Sanchez-Gomez S, Martinez-de-Tejada G, Brandenburg K, Brameshuber M, Schütz GJ, Andrä J, Gutsmann T. Cathelicidin and PMB neutralize endotoxins by multifactorial mechanisms including LPS interaction and targeting of host cell membranes. Proc Natl Acad Sci U S A 2021; 118:e2101721118. [PMID: 34183393 PMCID: PMC8271772 DOI: 10.1073/pnas.2101721118] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Antimicrobial peptides (AMPs) contribute to an effective protection against infections. The antibacterial function of AMPs depends on their interactions with microbial membranes and lipids, such as lipopolysaccharide (LPS; endotoxin). Hyperinflammation induced by endotoxin is a key factor in bacterial sepsis and many other human diseases. Here, we provide a comprehensive profile of peptide-mediated LPS neutralization by systematic analysis of the effects of a set of AMPs and the peptide antibiotic polymyxin B (PMB) on the physicochemistry of endotoxin, macrophage activation, and lethality in mice. Mechanistic studies revealed that the host defense peptide LL-32 and PMB each reduce LPS-mediated activation also via a direct interaction of the peptides with the host cell. As a biophysical basis, we demonstrate modifications of the structure of cholesterol-rich membrane domains and the association of glycosylphosphatidylinositol (GPI)-anchored proteins. Our discovery of a host cell-directed mechanism of immune control contributes an important aspect in the development and therapeutic use of AMPs.
Collapse
Affiliation(s)
- Andra B Schromm
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany;
| | - Laura Paulowski
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Yani Kaconis
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Franziska Kopp
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Max Koistinen
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Annemarie Donoghue
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Susanne Keese
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Christian Nehls
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Julia Wernecke
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Deutsches Elektronen-Synchrotron, D-22607 Hamburg, Germany
| | - Patrick Garidel
- Biophysikalische Chemie, Martin-Luther-Universität Halle-Wittenberg, D-06108 Halle, Germany
| | - Eva Sevcsik
- Institute of Applied Physics at TU Wien, Vienna 1040, Austria
| | - Karl Lohner
- Institute of Molecular Biosciences, Biophysics Division, University of Graz, A-8010 Graz, Austria
- BioTechMed-Graz, A-8010 Graz, Austria
| | - Susana Sanchez-Gomez
- Department of Microbiology and Parasitology, University of Navarra, E-31008 Pamplona, Spain
| | - Guillermo Martinez-de-Tejada
- Department of Microbiology and Parasitology, University of Navarra, E-31008 Pamplona, Spain
- Navarra Institute for Health Research, E-31008 Pamplona, Spain
| | - Klaus Brandenburg
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | | | | | - Jörg Andrä
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Department of Biotechnology, Faculty of Life Sciences, Hamburg University of Applied Sciences, D-21033 Hamburg, Germany
| | - Thomas Gutsmann
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| |
Collapse
|
31
|
Flotillin-Dependent Membrane Microdomains Are Required for Functional Phagolysosomes against Fungal Infections. Cell Rep 2021; 32:108017. [PMID: 32814035 PMCID: PMC10054021 DOI: 10.1016/j.celrep.2020.108017] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/03/2020] [Accepted: 07/17/2020] [Indexed: 11/23/2022] Open
Abstract
Lipid rafts form signaling platforms on biological membranes with incompletely characterized role in immune response to infection. Here we report that lipid-raft microdomains are essential components of phagolysosomal membranes of macrophages and depend on flotillins. Genetic deletion of flotillins demonstrates that the assembly of both major defense complexes vATPase and NADPH oxidase requires membrane microdomains. Furthermore, we describe a virulence mechanism leading to dysregulation of membrane microdomains by melanized wild-type conidia of the important human-pathogenic fungus Aspergillus fumigatus resulting in reduced phagolysosomal acidification. We show that phagolysosomes with ingested melanized conidia contain a reduced amount of free Ca2+ ions and that inhibition of Ca2+-dependent calmodulin activity led to reduced lipid-raft formation. We identify a single-nucleotide polymorphism in the human FLOT1 gene resulting in heightened susceptibility for invasive aspergillosis in hematopoietic stem cell transplant recipients. Collectively, flotillin-dependent microdomains on the phagolysosomal membrane play an essential role in protective antifungal immunity.
Collapse
|
32
|
Wei X, Zhang L, Zhang R, Wu R, Petitte JN, Hou Y, Si D, Ahmad B, Guo H, Zhang M, Cheng Q, Tong Y. Targeting the TLR2 Receptor With a Novel Thymopentin-Derived Peptide Modulates Immune Responses. Front Immunol 2021; 12:620494. [PMID: 34122400 PMCID: PMC8191578 DOI: 10.3389/fimmu.2021.620494] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 05/07/2021] [Indexed: 11/17/2022] Open
Abstract
The innate and adaptive immune systems act in concert to protect us from infectious agents and other harmful substances. As a state of temporary or permanent immune dysfunction, immunosuppression can make an organism more susceptible to infection, organ injury, and cancer due to damage to the immune system. It takes a long time to develop new immunomodulatory agents to prevent and treat immunosuppressive diseases, with slow progress. Toll-like receptor 2 (TLR2) agonists have been reported as potential immunomodulatory candidates due to their effective activation of immune responses. It has been demonstrated that thymopentin (TP5) could modulate immunity by binding to the TLR2 receptor. However, the fairly short half-life of TP5 greatly reduces its pharmacological potential for immunosuppression therapy. Although peptide cathelicidin 2 (CATH2) has a long half-life, it shows poor immunomodulatory activity and severe cytotoxicity, which seriously hampers its clinical development. Peptide hybridization is an effective approach for the design and engineering of novel functional peptides because hybrid peptides combine the advantages and benefits of various native peptides. In this study, to overcome all these challenges faced by the parental peptides, six hybrid peptides (CaTP, CbTP, CcTP, TPCa, TPCb, and TPCc) were designed by combining the full-length TP5 with different active fragments of CATH2. CbTP, the most potent TLR2 agonist among the six hybrid peptides, was effectively screened through in silico analysis and in vitro experiments. The CbTP peptide exhibited lower cytotoxicity than either CATH2 or TP5. Furthermore, the immunomodulatory effects of CbTP were confirmed in a CTX-immunosuppressed mouse model, which showed that CbTP has increased immunopotentiating activity and physiological stability compared to the parental peptides. CbTP successfully inhibited immunosuppression and weight loss, increased immune organ indices, and improved CD4+/CD8+ T lymphocyte subsets. In addition, CbTP significantly increased the production of the cytokine TNF-α and IL-6, and the immunoglobulins IgA, IgM, and IgG. The immunoenhancing effects of CbTP were attributed to its TLR2-binding activity, promoting the formation of the TLR2 cluster, the activation of the TLR2 receptor, and thus activation of the downstream MyD88-NF-кB signaling pathway.
Collapse
Affiliation(s)
- Xubiao Wei
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lulu Zhang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rijun Zhang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rujuan Wu
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - James N Petitte
- Prestage Department of Poultry Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, United States
| | - Yanfei Hou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Dayong Si
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Baseer Ahmad
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Henan Guo
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Manyi Zhang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiang Cheng
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yucui Tong
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
33
|
Lycopene Inhibits Toll-Like Receptor 4-Mediated Expression of Inflammatory Cytokines in House Dust Mite-Stimulated Respiratory Epithelial Cells. Molecules 2021; 26:molecules26113127. [PMID: 34073777 PMCID: PMC8197212 DOI: 10.3390/molecules26113127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 01/31/2023] Open
Abstract
House dust mites (HDM) are critical factors in airway inflammation. They activate respiratory epithelial cells to produce reactive oxygen species (ROS) and activate Toll-like receptor 4 (TLR4). ROS induce the expression of inflammatory cytokines in respiratory epithelial cells. Lycopene is a potent antioxidant nutrient with anti-inflammatory activity. The present study aimed to investigate whether HDM induce intracellular and mitochondrial ROS production, TLR4 activation, and pro-inflammatory cytokine expression (IL-6 and IL-8) in respiratory epithelial A549 cells. Additionally, we examined whether lycopene inhibits HDM-induced alterations in A549 cells. The treatment of A549 cells with HDM activated TLR4, induced the expression of IL-6 and IL-8, and increased intracellular and mitochondrial ROS levels. TAK242, a TLR4 inhibitor, suppressed both HDM-induced ROS production and cytokine expression. Furthermore, lycopene inhibited the HDM-induced TLR4 activation and cytokine expression, along with reducing the intracellular and mitochondrial ROS levels in HDM-treated cells. These results collectively indicated that the HDM induced TLR4 activation and increased intracellular and mitochondrial ROS levels, thus resulting in the induction of cytokine expression in respiratory epithelial cells. The antioxidant lycopene could inhibit HDM-induced cytokine expression, possibly by suppressing TLR4 activation and reducing the intracellular and mitochondrial ROS levels in respiratory epithelial cells.
Collapse
|
34
|
Chéneau C, Eichholz K, Tran TH, Tran TTP, Paris O, Henriquet C, Bajramovic JJ, Pugniere M, Kremer EJ. Lactoferrin Retargets Human Adenoviruses to TLR4 to Induce an Abortive NLRP3-Associated Pyroptotic Response in Human Phagocytes. Front Immunol 2021; 12:685218. [PMID: 34093588 PMCID: PMC8173049 DOI: 10.3389/fimmu.2021.685218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
Despite decades of clinical and preclinical investigations, we still poorly grasp our innate immune response to human adenoviruses (HAdVs) and their vectors. In this study, we explored the impact of lactoferrin on three HAdV types that are being used as vectors for vaccines. Lactoferrin is a secreted globular glycoprotein that influences direct and indirect innate immune response against a range of pathogens following a breach in tissue homeostasis. The mechanism by which lactoferrin complexes increases HAdV uptake and induce maturation of human phagocytes is unknown. We show that lactoferrin redirects HAdV types from species B, C, and D to Toll-like receptor 4 (TLR4) cell surface complexes. TLR4-mediated internalization of the HAdV-lactoferrin complex induced an NLRP3-associated response that consisted of cytokine release and transient disruption of plasma membrane integrity, without causing cell death. These data impact our understanding of HAdV immunogenicity and may provide ways to increase the efficacy of HAdV-based vectors/vaccines.
Collapse
Affiliation(s)
- Coraline Chéneau
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Karsten Eichholz
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Tuan Hiep Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Thi Thu Phuong Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Océane Paris
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Corinne Henriquet
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université Montpellier, Institut Régional du Cancer, Montpellier, France
| | | | - Martine Pugniere
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université Montpellier, Institut Régional du Cancer, Montpellier, France
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
35
|
Tseng YW, Chang CC, Chang YC. Novel Virulence Role of Pneumococcal NanA in Host Inflammation and Cell Death Through the Activation of Inflammasome and the Caspase Pathway. Front Cell Infect Microbiol 2021; 11:613195. [PMID: 33777832 PMCID: PMC7991587 DOI: 10.3389/fcimb.2021.613195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/05/2021] [Indexed: 01/15/2023] Open
Abstract
Streptococcus pneumoniae is one of most deadly Gram-positive bacterium that causes significant mortality and morbidity worldwide. Intense inflammation and cytotoxicity is a hallmark of invasive pneumococcal disease. Pneumococcal NanA has been shown to exaggerate the production of inflammatory cytokines via unmasking of inhibitory Siglec-5 from its sialyl cis-ligands. To further investigate the mechanistic role of NanA and Siglec-5 in pneumococccal diseases, we systemically analyzed genes and signaling pathways differentially regulated in macrophages infected with wild type and NanA-deficient pneumococcus. We found that NanA-mediated desialylation impairs the Siglec-5-TLR-2 interaction and reduces the recruitment of phosphatase SHP-1 to Siglec-5. This dysregulated crosstalk between TLR-2 and inhibitory Siglec-5 exaggerated multiple inflammatory and death signaling pathways and consequently caused excessive inflammation and cytotoxicity in the infected macrophage. Collectively, our results reveal a novel virulence role of NanA in pneumococcal pathogenesis and suggest that targeting NanA activity may ameliorate the pneumococcus-mediated inflammation and cytotoxicity in severe invasive pneumococcal diseases.
Collapse
Affiliation(s)
- Yu-Wen Tseng
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Chi Chang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Chi Chang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
36
|
Li M, Yu Y. Innate immune receptor clustering and its role in immune regulation. J Cell Sci 2021; 134:134/4/jcs249318. [PMID: 33597156 DOI: 10.1242/jcs.249318] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The discovery of receptor clustering in the activation of adaptive immune cells has revolutionized our understanding of the physical basis of immune signal transduction. In contrast to the extensive studies of adaptive immune cells, particularly T cells, there is a lesser, but emerging, recognition that the formation of receptor clusters is also a key regulatory mechanism in host-pathogen interactions. Many kinds of innate immune receptors have been found to assemble into nano- or micro-sized domains on the surfaces of cells. The clusters formed between diverse categories of innate immune receptors function as a multi-component apparatus for pathogen detection and immune response regulation. Here, we highlight these pioneering efforts and the outstanding questions that remain to be answered regarding this largely under-explored research topic. We provide a critical analysis of the current literature on the clustering of innate immune receptors. Our emphasis is on studies that draw connections between the phenomenon of receptor clustering and its functional role in innate immune regulation.
Collapse
Affiliation(s)
- Miao Li
- Department of Chemistry, Indiana University, Bloomington, IN 47401, USA
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN 47401, USA
| |
Collapse
|
37
|
Seclì L, Fusella F, Avalle L, Brancaccio M. The dark-side of the outside: how extracellular heat shock proteins promote cancer. Cell Mol Life Sci 2021; 78:4069-4083. [PMID: 33544155 PMCID: PMC8164615 DOI: 10.1007/s00018-021-03764-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/28/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
Abstract
In addition to exerting several essential house-keeping activities in the cell, heat shock proteins (HSPs) are crucial players in a well-structured molecular program activated in response to stressful challenges. Among the different activities carried out by HSPs during emergency, they reach the extracellular milieu, from where they scout the surroundings, regulate extracellular protein activity and send autocrine and paracrine signals. Cancer cells permanently experience stress conditions due to their altered equilibrium and behaviour, and constantly secrete heat shock proteins as a result. Other than supporting anti-tumour immunity, extracellular heat shock proteins (eHSPs), can also exacerbate cancer cell growth and malignancy by sustaining different cancer hallmarks. eHSPs are implicated in extracellular matrix remodelling, resistance to apoptosis, promotion of cell migration and invasion, induction of epithelial to mesenchymal transition, angiogenesis and activation of stromal cells, supporting ultimately, metastasis dissemination. A broader understanding of eHSP activity and contribution to tumour development and progression is leading to new opportunities in the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Laura Seclì
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.
| | - Federica Fusella
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.
| |
Collapse
|
38
|
Blevins LK, Crawford RB, Azzam DJ, Guilarte TR, Kaminski NE. Surface translocator protein 18 kDa (TSPO) localization on immune cells upon stimulation with LPS and in ART-treated HIV + subjects. J Leukoc Biol 2020; 110:123-140. [PMID: 33205494 DOI: 10.1002/jlb.3a1219-729rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 11/09/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a well-known outer mitochondrial membrane protein and it is widely used as a biomarker of neuroinflammation and brain injury. Although it is thought that TSPO plays key roles in a multitude of host cell functions, including steroid biosynthesis, apoptosis, generation of reactive oxygen species, and proliferation, some of these functions have recently been questioned. Here, we report the unexpected finding that circulating immune cells differentially express basal levels of TSPO on their cell surface, with a high percentage of monocytes and neutrophils expressing cell surface TSPO. In vitro stimulation of monocytes with LPS significantly increases the frequency of cells with surface TSPO expression in the absence of altered gene expression. Importantly, the LPS increase in TSPO cell surface expression in monocytes appears to be selective for LPS because two other distinct monocyte activators failed to increase the frequency of cells with surface TSPO. Finally, when we quantified immune cell TSPO surface expression in antiretroviral therapy-treated HIV+ donors, a chronic inflammatory disease, we found significant increases in the frequency of TSPO surface localization, which could be pharmacologically suppressed with ∆9 -tetrahydrocannabinol. These findings suggest that cell surface TSPO in circulating leukocytes could serve as a peripheral blood-based biomarker of inflammation.
Collapse
Affiliation(s)
- Lance K Blevins
- Department of Pharmacology and Toxicology, Center for Research on Ingredient Safety, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Robert B Crawford
- Department of Pharmacology and Toxicology, Center for Research on Ingredient Safety, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, Florida, USA
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, Florida, USA
| | - Norbert E Kaminski
- Department of Pharmacology and Toxicology, Center for Research on Ingredient Safety, Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
39
|
Kim SJ, Howe C, Mitchell J, Choo J, Powers A, Oikonomopoulos A, Pothoulakis C, Hommes DW, Im E, Rhee SH. Autotaxin loss accelerates intestinal inflammation by suppressing TLR4-mediated immune responses. EMBO Rep 2020; 21:e49332. [PMID: 32875703 DOI: 10.15252/embr.201949332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 07/20/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
Autotaxin (ATX) converts lysophosphatidylcholine and sphingosyl-phosphorylcholine into lysophosphatidic acid and sphingosine 1-phosphate, respectively. Despite the pivotal function of ATX in lipid metabolism, mechanisms by which ATX regulates immune and inflammatory disorders remain elusive. Here, using myeloid cell lineage-restricted Atx knockout mice, we show that Atx deficiency disrupts membrane microdomains and lipid rafts, resulting in the inhibition of Toll-like receptor 4 (TLR4) complex formation and the suppression of adaptor recruitment, thereby inhibiting TLR4-mediated responses in macrophages. Accordingly, TLR4-induced innate immune functions, including phagocytosis and iNOS expression, are attenuated in Atx-deficient macrophages. Consequently, Atx-/- mice exhibit a higher bacterial prevalence in the intestinal mucosa compared to controls. When combined with global Il10-/- mice, which show spontaneous colitis due to the translocation of luminal commensal microbes into the mucosa, myeloid cell lineage-restricted Atx knockout accelerates colitis development compared to control littermates. Collectively, our data reveal that Atx deficiency compromises innate immune responses, thereby promoting microbe-associated gut inflammation.
Collapse
Affiliation(s)
- Su Jin Kim
- Department of Biological Sciences, Oakland University, Rochester, MI, USA.,College of Pharmacy, Pusan National University, Busan, Korea
| | - Cody Howe
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Jonathon Mitchell
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Jieun Choo
- College of Pharmacy, Pusan National University, Busan, Korea
| | - Alexandra Powers
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Angelos Oikonomopoulos
- Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Charalabos Pothoulakis
- Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Daniel W Hommes
- Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan, Korea
| | - Sang Hoon Rhee
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| |
Collapse
|
40
|
Mustra Rakic J, Wang XD. Role of lycopene in smoke-promoted chronic obstructive pulmonary disease and lung carcinogenesis. Arch Biochem Biophys 2020; 689:108439. [PMID: 32504553 DOI: 10.1016/j.abb.2020.108439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 12/30/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer are a major cause of morbidity and mortality worldwide, with cigarette smoking being the single most important risk factor for both. Emerging evidence indicates alterations in reverse cholesterol transport-mediated removal of excess cholesterol from lung, and intracellular cholesterol overload to be involved in smoke-promoted COPD and lung cancer development. Since there are currently few effective treatments for COPD and lung cancer, it is important to identify food-derived, biologically active compounds, which can protect against COPD and lung cancer development. High intake of the carotenoid lycopene, as one of phytochemicals, is associated with a decreased risk of chronic lung lesions. This review article summarizes and discusses epidemiologic evidence, in vitro and in vivo studies regarding the prevention of smoke-promoted COPD and lung carcinogenesis through dietary lycopene as an effective intervention strategy. We focus on the recent research implying that lycopene preventive effect is through targeting the main genes involved in reverse cholesterol transport. This review also indicates gaps in knowledge about the function of lycopene against COPD and lung cancer, offering directions for further research.
Collapse
Affiliation(s)
- Jelena Mustra Rakic
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, MA, USA; Biochemical and Molecular Nutrition Program, Friedman School of Nutrition and Policy, Tufts University, Boston, MA, USA
| | - Xiang-Dong Wang
- Nutrition and Cancer Biology Lab, Jean Mayer USDA-Human Nutrition Research Center on Aging (HNRCA) at Tufts University, Boston, MA, USA; Biochemical and Molecular Nutrition Program, Friedman School of Nutrition and Policy, Tufts University, Boston, MA, USA.
| |
Collapse
|
41
|
Caveolar communication with xenobiotic-stalled ribosomes compromises gut barrier integrity. Commun Biol 2020; 3:270. [PMID: 32461676 PMCID: PMC7253476 DOI: 10.1038/s42003-020-0994-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
In response to internal and external insults, the intestinal lining undergoes various types of epithelial adaptation or pathologic distress via stress-responsive eIF2α kinase signaling and subsequent cellular reprogramming. As a vital platform for growth factor-linked adaptive signaling, caveolae were evaluated for epithelial modulation of the insulted gut. Patients under ulcerative insult displayed enhanced expression of caveolin-1, the main structural component of caveolae, which was positively associated with expression of protein kinase R (PKR), the ribosomal stress-responsive eIF2α kinase. PKR-linked biological responses were simulated in experimental gut models of ribosome-inactivating stress using mice and Caenorhabditis elegans. Caveolar activation counteracted the expression of wound-protective epidermal growth factor receptor (EGFR) and its target genes, such as chemokines that were pivotal for epithelial integrity in the ribosome-inactivated gut. Mechanistic findings regarding ribosomal inactivation-associated disorders in the gut barrier provide crucial molecular evidence for detrimental caveolar actions against EGFR-mediated epithelial protection in patients with IBD.
Collapse
|
42
|
Savio LEB, de Andrade Mello P, Santos SACS, de Sousa JC, Oliveira SDS, Minshall RD, Kurtenbach E, Wu Y, Longhi MS, Robson SC, Coutinho-Silva R. P2X7 receptor activation increases expression of caveolin-1 and formation of macrophage lipid rafts, thereby boosting CD39 activity. J Cell Sci 2020; 133:jcs.237560. [PMID: 32005701 DOI: 10.1242/jcs.237560] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Macrophages are tissue-resident immune cells that are crucial for the initiation and maintenance of immune responses. Purinergic signaling modulates macrophage activity and impacts cellular plasticity. The ATP-activated purinergic receptor P2X7 (also known as P2RX7) has pro-inflammatory properties, which contribute to macrophage activation. P2X7 receptor signaling is, in turn, modulated by ectonucleotidases, such as CD39 (also known as ENTPD1), expressed in caveolae and lipid rafts. Here, we examined P2X7 receptor activity and determined impacts on ectonucleotidase localization and function in macrophages primed with lipopolysaccharide (LPS). First, we verified that ATP boosts CD39 activity and caveolin-1 protein expression in LPS-primed macrophages. Drugs that disrupt cholesterol-enriched domains - such as nystatin and methyl-β-cyclodextrin - decreased CD39 enzymatic activity in all circumstances. We noted that CD39 colocalized with lipid raft markers (flotillin-2 and caveolin-1) in macrophages that had been primed with LPS followed by treatment with ATP. P2X7 receptor inhibition blocked these ATP-mediated increases in caveolin-1 expression and inhibited the colocalization with CD39. Further, we found that STAT3 activation is significantly attenuated caveolin-1-deficient macrophages treated with LPS or LPS+BzATP. Taken together, our data suggest that P2X7 receptor triggers the initiation of lipid raft-dependent mechanisms that upregulates CD39 activity and could contribute to limit macrophage responses restoring homeostasis.
Collapse
Affiliation(s)
- Luiz Eduardo Baggio Savio
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Paola de Andrade Mello
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Stephanie Alexia Cristina Silva Santos
- Laboratory of Molecular Biology and Biochemistry of Proteins, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Júlia Costa de Sousa
- Laboratory of Molecular Biology and Biochemistry of Proteins, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Suellen D S Oliveira
- Departments of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Richard D Minshall
- Departments of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA.,Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Eleonora Kurtenbach
- Laboratory of Molecular Biology and Biochemistry of Proteins, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Yan Wu
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Maria Serena Longhi
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Simon C Robson
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
43
|
ABC Transporters, Cholesterol Efflux, and Implications for Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:67-83. [DOI: 10.1007/978-981-15-6082-8_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Barnett KC, Kagan JC. Lipids that directly regulate innate immune signal transduction. Innate Immun 2020; 26:4-14. [PMID: 31180799 PMCID: PMC6901815 DOI: 10.1177/1753425919852695] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/28/2022] Open
Abstract
Pattern Recognition Receptors (PRRs) detect evidence of infection and tissue damage. The activation of these receptors and their downstream signal transduction pathways initiate a protective immune response. These signaling pathways are influenced by their spatial context, and precise subcellular positioning of proteins and protein complexes in these pathways is essential for effective immune responses in vivo . This organization is not limited to transmembrane proteins that reside in specific organelles, but also to proteins that engage membrane lipid head groups for proper positioning. In this review, we focus on the role of cell membranes and protein–lipid interactions in innate immune signal transduction and how their mechanisms of localization regulate the immune response. We will discuss how lipids spatially regulate the sensing of damage or infection, mediate effector activity, and serve as messengers of cell death and tissue damage.
Collapse
Affiliation(s)
- Katherine C Barnett
- Harvard Medical School and Division of
Gastroenterology, Boston Children’s Hospital, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of
Gastroenterology, Boston Children’s Hospital, USA
| |
Collapse
|
45
|
Hong W, Zimmer V, Basharat Z, Zippi M, Stock S, Geng W, Bao X, Dong J, Pan J, Zhou M. Association of total cholesterol with severe acute pancreatitis: A U-shaped relationship. Clin Nutr 2020; 39:250-257. [PMID: 30772093 DOI: 10.1016/j.clnu.2019.01.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/09/2019] [Accepted: 01/23/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS There is no consensus on relationship between total cholesterol levels and incidence of severe acute pancreatitis (SAP). The aim of this study was to investigate the relation between total cholesterol (TC) and the disease severity of acute pancreatitis. METHODS We conducted a cross-sectional study on patients with acute pancreatitis between April 2012 and December 2015 in a university hospital. Fasting blood total cholesterol (TC) was assayed within 24 h of admission, as well as 3-5 days, 7-9 days and 13-15 days during hospitalization. Time interval before admission, age, gender, Body Mass Index, hypertension, diabetes mellitus, alcohol consumption, smoking, etiology and albumin were recorded as potential confounding factors. To assess the pattern of relationship of TC and SAP, we used restricted cubic spline analysis with multivariable logistic regression analysis. We also compared total cholesterol concentrations between patients with or without SAP at different time points. RESULTS 648 patients (median age: 47.5 years; 62.4% man) were enrolled. The incidence of SAP was 10%. A U-shaped association of TC level within 24 h of admission with severity was observed in acute pancreatitis. Patients with low TC levels (<160 mg/dL) and high TC levels (>240 mg/dL) had a significantly higher incidence of SAP and protracted hospital stays when compared to moderate TC levels (160-240 mg/dL). Low total cholesterol levels (OR 2.72; 95 %eCI 1.27-5.83; P = 0.01) and high total cholesterol levels (OR 2.54; 95 %eCI 1.09-5.89; P = 0.03), were still independently associated with development of SAP after adjusting for potential confounding factors. Longitudinal cohort study indicated that patients with SAP had lower total cholesterol concentrations among 3-15 days after admission compared to patients without SAP (P < 0.001). CONCLUSIONS Both low TC level (<160 mg/dL) and high TC (>240 mg/dL) within 24 h of admission is independently associated with an increased risk of SAP.
Collapse
Affiliation(s)
- Wandong Hong
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou, Zhejiang, People's Republic of China.
| | - Vincent Zimmer
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, 66424, Germany; Department of Medicine, Marienhausklinik St. Josef Kohlhof, Neunkirchen, 66539, Germany.
| | - Zarrin Basharat
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; Laboratoire Génomique, Bioinformatique et Applications, Conservatoire National des Arts et 11 Métiers, Paris, 75003, France.
| | - Maddalena Zippi
- Unit of Gastroenterology and Digestive Endoscopy, Sandro Pertini Hospital, Rome, Italy.
| | - Simon Stock
- Department of Surgery, World Mate Emergency Hospital, Battambang, Cambodia.
| | - Wujun Geng
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Xueqin Bao
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Junfeng Dong
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Jingye Pan
- Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Mengtao Zhou
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
46
|
Dominant role of splenic marginal zone lipid rafts in the classical complement pathway against S. pneumoniae. Cell Death Discov 2019; 5:133. [PMID: 31531231 PMCID: PMC6733876 DOI: 10.1038/s41420-019-0213-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/11/2019] [Accepted: 08/18/2019] [Indexed: 12/28/2022] Open
Abstract
Lipid rafts (LRs) play crucial roles in complex physiological processes, modulating innate and acquired immune responses to pathogens. The transmembrane C-type lectins human dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) and its mouse homolog SIGN-R1 are distributed in LRs and expressed on splenic marginal zone (MZ) macrophages. The DC-SIGN-C1q or SIGN-R1-C1q complex could mediate the immunoglobulin (Ig)-independent classical complement pathway against Streptococcus pneumoniae. Precise roles of LRs during this complement pathway are unknown. Here we show that LRs are indispensable for accelerating the DC-SIGN- or SIGN-R1-mediated classical complement pathway against S. pneumoniae, thus facilitating rapid clearance of the pathogen. The trimolecular complex of SIGN-R1-C1q-C4 was exclusively enriched in LRs of splenic MZ macrophages and their localization was essential for activating C3 catabolism and enhancing pneumococcal clearance, which were abolished in SIGN-R1-knockout mice. However, DC-SIGN replacement on splenic MZ macrophage’s LRs of SIGN-R1-depleted mice reversed these defects. Disruption of LRs dramatically reduced pneumococcal uptake and decomposition. Additionally, DC- SIGN, C1q, C4, and C3 were obviously distributed in splenic LRs of cadavers. Therefore, LRs on splenic SIGN-R1+ or DC-SIGN+ macrophages could provide spatially confined and optimal bidirectional platforms, not only for usual intracellular events, for example recognition and phagocytosis of pathogens, but also an unusual extracellular event such as the complement system. These findings improve our understanding of the orchestrated roles of the spleen, unraveling a new innate immune system initiated from splenic MZ LRs, and yielding answers to several long-standing problems, including the need to understand the profound role of LRs in innate immunity, the need to identify how such a small portion of splenic SIGN-R1+ macrophages (<0.05% of splenic macrophages) effectively resist S. pneumoniae, and the need to understand how LRs can promote the protective function of DC-SIGN against S. pneumoniae in the human spleen.
Collapse
|
47
|
Lin CY, Lai HL, Chen HM, Siew JJ, Hsiao CT, Chang HC, Liao KS, Tsai SC, Wu CY, Kitajima K, Sato C, Khoo KH, Chern Y. Functional roles of ST8SIA3-mediated sialylation of striatal dopamine D 2 and adenosine A 2A receptors. Transl Psychiatry 2019; 9:209. [PMID: 31455764 PMCID: PMC6712005 DOI: 10.1038/s41398-019-0529-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 05/22/2019] [Accepted: 06/20/2019] [Indexed: 01/20/2023] Open
Abstract
Sialic acids are typically added to the end of glycoconjugates by sialyltransferases. Among the six ST8 α-N-acetyl-neuraminide α-2,8-sialyltransferases (ST8SIA) existing in adult brains, ST8SIA2 is a schizophrenia-associated gene. However, the in vivo substrates and physiological functions of most sialyltransferases are currently unknown. The ST8SIA3 is enriched in the striatum. Here, we showed that ablation of St8sia3 in mice (St8sia3-KO) led to fewer disialylated and trisialylated terminal glycotopes in the striatum of St8sia3-KO mice. Moreover, the apparent sizes of several striatum-enriched G-protein-coupled receptors (GPCRs) (including the adenosine A2A receptor (A2AR) and dopamine D1/D2 receptors (D1R and D2R)) were smaller in St8sia3-KO mice than in WT mice. A sialidase treatment removed the differences in the sizes of these molecules between St8sia3-KO and WT mice, confirming the involvement of sialylation. Expression of ST8SIA3 in the striatum of St8sia3-KO mice using adeno-associated viruses normalized the sizes of these proteins, demonstrating a direct role of ST8SIA3. The lack of ST8SIA3-mediated sialylation altered the distribution of these proteins in lipid rafts and the interaction between D2R and A2AR. Locomotor activity assays revealed altered pharmacological responses of St8sia3-KO mice to drugs targeting these receptors and verified that a greater population of D2R formed heteromers with A2AR in the striatum of St8sia3-KO mice. Since the A2AR-D2R heteromer is an important drug target for several basal ganglia diseases (such as schizophrenia and Parkinson's disease), the present study not only reveals a crucial role for ST8SIA3 in striatal functions but also provides a new drug target for basal ganglia-related diseases.
Collapse
Affiliation(s)
- Chien-Yu Lin
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsing-Lin Lai
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Mei Chen
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jian-Jing Siew
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,0000 0001 0425 5914grid.260770.4Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Cheng-Te Hsiao
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hua-Chien Chang
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Kuo-Shiang Liao
- 0000 0001 2287 1366grid.28665.3fGenomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shih-Chieh Tsai
- grid.36020.37Department of Research and Development, National Laboratory Animal Center, National Applied Research Laboratories, Taipei and Tainan, Taipei, Taiwan
| | - Chung-Yi Wu
- 0000 0001 2287 1366grid.28665.3fGenomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ken Kitajima
- 0000 0001 0943 978Xgrid.27476.30Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-860 Japan
| | - Chihiro Sato
- 0000 0001 0943 978Xgrid.27476.30Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-860 Japan
| | - Kay-Hooi Khoo
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
48
|
Carlin CR. New Insights to Adenovirus-Directed Innate Immunity in Respiratory Epithelial Cells. Microorganisms 2019; 7:microorganisms7080216. [PMID: 31349602 PMCID: PMC6723309 DOI: 10.3390/microorganisms7080216] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
The nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) family of transcription factors is a key component of the host innate immune response to infectious adenoviruses and adenovirus vectors. In this review, we will discuss a regulatory adenoviral protein encoded by early region 3 (E3) called E3-RIDα, which targets NFκB through subversion of novel host cell pathways. E3-RIDα down-regulates an EGF receptor signaling pathway, which overrides NFκB negative feedback control in the nucleus, and is induced by cell stress associated with viral infection and exposure to the pro-inflammatory cytokine TNF-α. E3-RIDα also modulates NFκB signaling downstream of the lipopolysaccharide receptor, Toll-like receptor 4, through formation of membrane contact sites controlling cholesterol levels in endosomes. These innate immune evasion tactics have yielded unique perspectives regarding the potential physiological functions of host cell pathways with important roles in infectious disease.
Collapse
Affiliation(s)
- Cathleen R Carlin
- Department of Molecular Biology and Microbiology and the Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
49
|
Jia GL, Huang Q, Cao YN, Xie CS, Shen YJ, Chen JL, Lu JH, Zhang MB, Li J, Tao YX, Cao H. Cav-1 participates in the development of diabetic neuropathy pain through the TLR4 signaling pathway. J Cell Physiol 2019; 235:2060-2070. [PMID: 31318049 DOI: 10.1002/jcp.29106] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022]
Abstract
This study aims to determine whether caveolin-1 (Cav-1) participates in the process of diabetic neuropathic pain by directly regulating the expression of toll-like receptor 4 (TLR4) and the subsequent phosphorylation of N-methyl-D-aspartate receptor 2B subunit (NR2B) in the spinal cord. Male Sprague-Dawley rats (120-150 g) were continuously fed with high-fat and high-sugar diet for 8 weeks, and received a single low-dose of intraperitoneal streptozocin injection in preparation for the type-II diabetes model. Then, these rats were divided into five groups according to the level of blood glucose, and the mechanical withdrawal threshold and thermal withdrawal latency values. The pain thresholds were measured at 3, 7, and 14 days after animal grouping. Then, eight rats were randomly chosen from each group and killed. Lumbar segments 4-6 of the spinal cord were removed for western blot analysis and immunofluorescence assay. Cav-1 was persistently upregulated in the spinal cord after diabetic neuropathic pain in rats. The downregulation of Cav-1 through the subcutaneous injection of Cav-1 inhibitor daidzein ameliorated the pain hypersensitivity and TLR4 expression in the spinal cord in diabetic neuropathic pain (DNP) rats. Furthermore, it was found that Cav-1 directly bound with TLR4, and the subsequent phosphorylation of NR2B in the spinal cord contributed to the modulation of DNP. These findings suggest that Cav-1 plays a vital role in DNP processing at least in part by directly regulating the expression of TLR4, and through the subsequent phosphorylation of NR2B in the spinal cord.
Collapse
Affiliation(s)
- Gai-Li Jia
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Qi Huang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Yan-Nan Cao
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Ci-Shan Xie
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Yu-Jing Shen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Jia-Li Chen
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Jia-Hui Lu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Mao-Biao Zhang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Jun Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Hong Cao
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Pain Medicine Institute of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
50
|
Kubelkova K, Macela A. Innate Immune Recognition: An Issue More Complex Than Expected. Front Cell Infect Microbiol 2019; 9:241. [PMID: 31334134 PMCID: PMC6616152 DOI: 10.3389/fcimb.2019.00241] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022] Open
Abstract
Primary interaction of an intracellular bacterium with its host cell is initiated by activation of multiple signaling pathways in response to bacterium recognition itself or as cellular responses to stress induced by the bacterium. The leading molecules in these processes are cell surface membrane receptors as well as cytosolic pattern recognition receptors recognizing pathogen-associated molecular patterns or damage-associated molecular patterns induced by the invading bacterium. In this review, we demonstrate possible sequences of events leading to recognition of Francisella tularensis, present findings on known mechanisms for manipulating cell responses to protect Francisella from being killed, and discuss newly published data from the perspective of early stages of host-pathogen interaction.
Collapse
Affiliation(s)
- Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| | | |
Collapse
|