1
|
Chernov AS, Telegin GB, Minakov AN, Kazakov VA, Rodionov MV, Palikov VA, Kudriaeva AA, Belogurov AA. Synthetic Amphipathic Helical Peptide L-37pA Ameliorates the Development of Acute Respiratory Distress Syndrome (ARDS) and ARDS-Induced Pulmonary Fibrosis in Mice. Int J Mol Sci 2024; 25:8384. [PMID: 39125954 PMCID: PMC11312864 DOI: 10.3390/ijms25158384] [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: 06/20/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
In this study, we evaluated the ability of the synthetic amphipathic helical peptide (SAHP), L-37pA, which mediates pathogen recognition and innate immune responses, to treat acute respiratory distress syndrome (ARDS) accompanied by diffuse alveolar damage (DAD) and chronic pulmonary fibrosis (PF). For the modeling of ARDS/DAD, male ICR mice were used. Intrabronchial instillation (IB) of 200 µL of inflammatory agents was performed by an intravenous catheter 20 G into the left lung lobe only, leaving the right lobe unaffected. Intravenous injections (IVs) of L-37pA, dexamethasone (DEX) and physiological saline (saline) were used as therapies for ARDS/DAD. L37pA inhibited the circulating levels of inflammatory cytokines, such as IL-8, TNFα, IL1α, IL4, IL5, IL6, IL9 and IL10, by 75-95%. In all cases, the computed tomography (CT) data indicate that L-37pA reduced lung density faster to -335 ± 23 Hounsfield units (HU) on day 7 than with DEX and saline, to -105 ± 29 HU and -23 ± 11 HU, respectively. The results of functional tests showed that L-37pA treatment 6 h after ARDS/DAD initiation resulted in a more rapid improvement in the physiological respiratory lung by 30-45% functions compared with the comparison drugs. Our data suggest that synthetic amphipathic helical peptide L-37pA blocked a cytokine storm, inhibited acute and chronic pulmonary inflammation, prevented fibrosis development and improved physiological respiratory lung function in the ARDS/DAD mouse model. We concluded that a therapeutic strategy using SAHPs targeting SR-B receptors is a potential novel effective treatment for inflammation-induced ARDS, DAD and lung fibrosis of various etiologies.
Collapse
Affiliation(s)
- Aleksandr S. Chernov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Georgii B. Telegin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Alexey N. Minakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Vitaly A. Kazakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Maksim V. Rodionov
- Medical Radiological Research Center (MRRC) Named after A.F. Tsyb-Branch of the National Medical Radiological Research Center of the Ministry of Health of the Russian Federation, Obninsk 249031, Russia;
| | - Viktor A. Palikov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Anna A. Kudriaeva
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Alexey A. Belogurov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
- Department of Biological Chemistry, Russian University of Medicine of the Ministry of Health of the Russian Federation, Moscow 127473, Russia
| |
Collapse
|
2
|
Chirivi M, Contreras GA. Endotoxin-induced alterations of adipose tissue function: a pathway to bovine metabolic stress. J Anim Sci Biotechnol 2024; 15:53. [PMID: 38581064 PMCID: PMC10998405 DOI: 10.1186/s40104-024-01013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/14/2024] [Indexed: 04/07/2024] Open
Abstract
During the periparturient period, dairy cows exhibit negative energy balance due to limited appetite and increased energy requirements for lactogenesis. The delicate equilibrium between energy availability and expenditure puts cows in a state of metabolic stress characterized by excessive lipolysis in white adipose tissues (AT), increased production of reactive oxygen species, and immune cell dysfunction. Metabolic stress, especially in AT, increases the risk for metabolic and inflammatory diseases. Around parturition, cows are also susceptible to endotoxemia. Bacterial-derived toxins cause endotoxemia by promoting inflammatory processes and immune cell infiltration in different organs and systems while impacting metabolic function by altering lipolysis, mitochondrial activity, and insulin sensitivity. In dairy cows, endotoxins enter the bloodstream after overcoming the defense mechanisms of the epithelial barriers, particularly during common periparturient conditions such as mastitis, metritis, and pneumonia, or after abrupt changes in the gut microbiome. In the bovine AT, endotoxins induce a pro-inflammatory response and stimulate lipolysis in AT, leading to the release of free fatty acids into the bloodstream. When excessive and protracted, endotoxin-induced lipolysis can impair adipocyte's insulin signaling pathways and lipid synthesis. Endotoxin exposure can also induce oxidative stress in AT through the production of reactive oxygen species by inflammatory cells and other cellular components. This review provides insights into endotoxins' impact on AT function, highlighting the gaps in our knowledge of the mechanisms underlying AT dysfunction, its connection with periparturient cows' disease risk, and the need to develop effective interventions to prevent and treat endotoxemia-related inflammatory conditions in dairy cattle.
Collapse
Affiliation(s)
- Miguel Chirivi
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
3
|
Fung KYY, Ho TWW, Xu Z, Neculai D, Beauchemin CAA, Lee WL, Fairn GD. Apolipoprotein A1 and high-density lipoprotein limit low-density lipoprotein transcytosis by binding SR-B1. J Lipid Res 2024; 65:100530. [PMID: 38479648 PMCID: PMC11004410 DOI: 10.1016/j.jlr.2024.100530] [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: 09/06/2022] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/09/2024] Open
Abstract
Atherosclerosis results from the deposition and oxidation of LDL and immune cell infiltration in the sub-arterial space leading to arterial occlusion. Studies have shown that transcytosis transports circulating LDL across endothelial cells lining blood vessels. LDL transcytosis is initiated by binding to either scavenger receptor B1 (SR-B1) or activin A receptor-like kinase 1 on the apical side of endothelial cells leading to its transit and release on the basolateral side. HDL is thought to partly protect individuals from atherosclerosis due to its ability to remove excess cholesterol and act as an antioxidant. Apolipoprotein A1 (APOA1), an HDL constituent, can bind to SR-B1, raising the possibility that APOA1/HDL can compete with LDL for SR-B1 binding, thereby limiting LDL deposition in the sub-arterial space. To examine this possibility, we used in vitro approaches to quantify the internalization and transcytosis of fluorescent LDL in coronary endothelial cells. Using microscale thermophoresis and affinity capture, we find that SR-B1 and APOA1 interact and that binding is enhanced when using the cardioprotective variant of APOA1 termed Milano (APOA1-Milano). In male mice, transiently increasing the levels of HDL reduced the acute deposition of fluorescently labeled LDL in the atheroprone inner curvature of the aorta. Reduced LDL deposition was also observed when increasing circulating wild-type APOA1 or the APOA1-Milano variant, with a more robust inhibition from the APOA1-Milano. The results suggest that HDL may limit SR-B1-mediated LDL transcytosis and deposition, adding to the mechanisms by which it can act as an atheroprotective particle.
Collapse
Affiliation(s)
- Karen Y Y Fung
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Tse Wing Winnie Ho
- Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Zizhen Xu
- Department of Cell Biology, and Department of Pathology Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dante Neculai
- Department of Cell Biology, and Department of Pathology Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Catherine A A Beauchemin
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Interdisciplinary Theoretical and Mathematical Sciences (iTHEMS) program, RIKEN, Wako, Saitama, Japan
| | - Warren L Lee
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
4
|
Wang Y, Fan Y, Jiang Y, Wang E, Song Y, Chen H, Xu F, Xie K, Yu Y. APOA2: New Target for Molecular Hydrogen Therapy in Sepsis-Related Lung Injury Based on Proteomic and Genomic Analysis. Int J Mol Sci 2023; 24:11325. [PMID: 37511084 PMCID: PMC10379236 DOI: 10.3390/ijms241411325] [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: 06/05/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Target biomarkers for H2 at both the protein and genome levels are still unclear. In this study, quantitative proteomics acquired from a mouse model were first analyzed. At the same time, functional pathway analysis helped identify functional pathways at the protein level. Then, bioinformatics on mRNA sequencing data were conducted between sepsis and normal mouse models. Differential expressional genes with the closest relationship to disease status and development were identified through module correlation analysis. Then, common biomarkers in proteomics and transcriptomics were extracted as target biomarkers. Through analyzing expression quantitative trait locus (eQTL) and genome-wide association studies (GWAS), colocalization analysis on Apoa2 and sepsis phenotype was conducted by summary-data-based Mendelian randomization (SMR). Then, two-sample and drug-target, syndrome Mendelian randomization (MR) analyses were all conducted using the Twosample R package. For protein level, protein quantitative trait loci (pQTLs) of the target biomarker were also included in MR. Animal experiments helped validate these results. As a result, Apoa2 protein or mRNA was identified as a target biomarker for H2 with a protective, causal relationship with sepsis. HDL and type 2 diabetes were proven to possess causal relationships with sepsis. The agitation and inhibition of Apoa2 were indicated to influence sepsis and related syndromes. In conclusion, we first proposed Apoa2 as a target for H2 treatment.
Collapse
Affiliation(s)
- Yuanlin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yan Fan
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yi Jiang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Enquan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yu Song
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hongguang Chen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Feier Xu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
5
|
de Queiroz NMGP, de Oliveira LS, Gomes MTR, Carneiro MBH, Vieira LQ, Oliveira SC, Horta MF. Requirement of scavenger receptors for activation of the IRF-3/IFN-β/STAT-1 pathway in TLR4-mediated production of NO by LPS-activated macrophages. Nitric Oxide 2023; 134-135:61-71. [PMID: 37059259 DOI: 10.1016/j.niox.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/02/2023] [Accepted: 04/12/2023] [Indexed: 04/16/2023]
Abstract
Production of nitric oxide (NO) by LPS-activated macrophages is due to a complex cellular signaling initiated by TLR4 that leads to the transcription of IFN-β, which activates IRF-1 and STAT-1, as well as to the activation of NF-κB, required for iNOS transcription. High concentrations of LPS can also be uptaken by scavenger receptors (SRs), which, in concert with TLR4, leads to inflammatory responses. The mechanisms by which TLR4 and SRs interact, and the pathways activated by this interaction in macrophages are not elucidated. Therefore, our main goal was to evaluate the role of SRs, particularly SR-A, in LPS-stimulated macrophages for NO production. We first showed that, surprisingly, LPS can induce the expression of iNOS and the production of NO in TLR4-/- mice, provided exogenous IFN-β is supplied. These results indicate that LPS stimulate receptors other than TLR4. The inhibition of SR-A using DSS or neutralizing antibody to SR-AI showed that SR-A is essential for the expression of iNOS and NO production in stimulation of TLR4 by LPS. The restoration of the ability to express iNOS and produce NO by addition of rIFN-β to inhibited SR-A cells indicated that the role of SR-AI in LPS-induced NO production is to provide IFN-β, probably by mediating the internalization of LPS/TLR4, and the differential inhibition by DSS and neutralizing antibody to SR-AI suggested that other SRs are also involved. Our results reinforce that TLR4 and SR-A act in concert in LPS activation and demonstrated that, for the production of NO, it does mainly by synthesizing IRF-3 and also by activating the TRIF/IRF-3 pathway for IFN-β production, essential for LPS-mediated transcription of iNOS. Consequently STAT-1 is activated, and IRF-1 is expressed, which together with NF-κB from TLR4/MyD88/TIRAP, induce iNOS synthesis and NO production. SUMMARY SENTENCE: TLR4 and SRs act in concert activating IRF-3 to transcribe IFN-β and activate STAT-1 to produce NO by LPS-activated macrophages.
Collapse
Affiliation(s)
- Nina Marí Gual Pimenta de Queiroz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Luciana Souza de Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Marco Tulio Ribeiro Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Matheus Batista Heitor Carneiro
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Leda Quercia Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, Salvador, BA, Brazil
| | - Maria Fátima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
6
|
Taban Q, Ahmad SM, Mumtaz PT, Bhat B, Haq E, Magray S, Saleem S, Shabir N, Muhee A, Kashoo ZA, Zargar MH, Malik AA, Ganai NA, Shah RA. Scavenger receptor B1 facilitates the endocytosis of Escherichia coli via TLR4 signaling in mammary gland infection. Cell Commun Signal 2023; 21:3. [PMID: 36604713 PMCID: PMC9813905 DOI: 10.1186/s12964-022-01014-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/11/2022] [Indexed: 01/06/2023] Open
Abstract
SCARB1 belongs to class B of Scavenger receptors (SRs) that are known to be involved in binding and endocytosis of various pathogens. SRs have emerging role in regulating innate immunity and host-pathogen interactions by acting in co-ordination with Toll-like receptors.Query Little is known about the function of SCARB1 in milk-derived mammary epithelial cells (MECs). This study reports the role of SCARB1 in infection and its potential association in TLR4 signaling on bacterial challenge in Goat mammary epithelial cells (GMECs). The novelty in the establishment of MEC culture lies in the method that aims to enhance the viability of the cells with intact characteristics upto a higher passage number. We represent MEC culture to be used as a potential infection model for deeper understanding of animal physiology especially around the mammary gland. On E.coli challenge the expression of SCARB1 was significant in induced GMECs at 6 h. Endoribonuclease-esiRNA based silencing of SCARB1 affects the expression of TLR4 and its pathways i.e. MyD88 and TRIF pathways on infection. Knockdown also affected the endocytosis of E.coli in GMECs demonstrating that E.coli uses SCARB1 function to gain entry in cells. Furthermore, we predict 3 unique protein structures of uncharacterized SCARB1 (Capra hircus) protein. Overall, we highlight SCARB1 as a main participant in host defence and its function in antibacterial advances to check mammary gland infections. Video Abstract.
Collapse
Affiliation(s)
- Qamar Taban
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
- Department of Biotechnology, University of Kashmir, Hazratbal Srinagar, Jammu and Kashmir, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India.
| | | | - Basharat Bhat
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Ehtishamul Haq
- Department of Biotechnology, University of Kashmir, Hazratbal Srinagar, Jammu and Kashmir, India
| | - Suhail Magray
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Sahar Saleem
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Nadeem Shabir
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Amatul Muhee
- Department of Clinical Veterinary Medicine, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Zahid Amin Kashoo
- Department of Veterinary Microbiology & Immunology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Mahrukh Hameed Zargar
- Department of Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Abrar A Malik
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Nazir A Ganai
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| | - Riaz A Shah
- Division of Animal Biotechnology, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, FV.Sc and A.H, Shuhama, Jammu and Kashmir, India
| |
Collapse
|
7
|
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: 3] [Impact Index Per Article: 1.5] [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
|
8
|
Palmieri M, Joseph TE, O’Brien CA, Gomez-Acevedo H, Manolagas SC, Ambrogini E. Deletion of the scavenger receptor Scarb1 in osteoblast progenitors does not affect bone mass. PLoS One 2022; 17:e0265893. [PMID: 35349600 PMCID: PMC8963559 DOI: 10.1371/journal.pone.0265893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/09/2022] [Indexed: 11/18/2022] Open
Abstract
The scavenger receptor class B member 1 (SR-B1 or Scarb1) is a cell surface receptor for high density lipoproteins. It also binds oxidized low density lipoproteins and phosphocholine-containing oxidized phospholipids (PC-OxPL), which adversely affect bone homeostasis. Overexpression of a single chain form of the antigen-binding domain of E06 IgM-a natural antibody that recognizes PC-OxPL-increases trabecular and cortical bone mass in female and male mice by stimulating bone formation. We have previously reported that Scarb1 is the most abundant scavenger receptor for PC-OxPL in calvaria-derived osteoblastic cells. Additionally, bone marrow- and calvaria-derived osteoblasts from Scarb1 knockout mice (Scarb1 KO) are protected from the pro-apoptotic and anti-differentiating effects of OxPL. Previous skeletal analysis of Scarb1 KO mice has produced contradictory results, with some studies reporting elevated bone mass but another study reporting low bone mass. To clarify the role of Scarb1 in osteoblasts, we deleted Scarb1 specifically in cells of the osteoblast lineage using Osx1-Cre transgenic mice. We observed no difference in bone mineral density measured by DXA in either female or male Osx1-Cre;Scarb1fl/fl mice compared to wild type (WT), Osx1-Cre, or Scarb1fl/fl littermate controls. Additionally, microCT analysis of 6-month-old females and 7-month-old males did not detect any difference in trabecular or cortical bone mass between genotypes. These results indicate that expression of Scarb1 in cells of the osteoblast lineage does not play an important role in bone homeostasis and, therefore, it is not essential for the effects of PC-OxPL on these cells.
Collapse
Affiliation(s)
- Michela Palmieri
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Teenamol E. Joseph
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Charles A. O’Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Horacio Gomez-Acevedo
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Stavros C. Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| |
Collapse
|
9
|
Zhong Q, Chatterjee S, Choudhary JS, Frankel G. EPEC-induced activation of the Ca 2+ transporter TRPV2 leads to pyroptotic cell death. Mol Microbiol 2022; 117:480-492. [PMID: 34897856 DOI: 10.1111/mmi.14863] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/27/2022]
Abstract
The enteropathogenic Escherichia coli (EPEC) type III secretion system effector Tir, which mediates intimate bacterial attachment to epithelial cells, also triggers Ca2+ influx followed by LPS entry and caspase-4-dependent pyroptosis, which could be antagonized by the effector NleF. Here we reveal the mechanism by which EPEC induces Ca2+ influx. We show that in the intestinal epithelial cell line SNU-C5, Tir activates the mechano/osmosensitive cation channel TRPV2 which triggers extracellular Ca2+ influx. Tir-induced Ca2+ influx could be blocked by siRNA silencing of TRPV2, pre-treatment with the TRPV2 inhibitor SET2 or by growing cells in low osmolality medium. Pharmacological activation of TRPV2 in the absence of Tir failed to initiate caspase-4-dependent cell death, confirming the necessity of Tir. Consistent with the model implicating activation on translocation of TRPV2 from the ER to plasma membrane, inhibition of protein trafficking by either brefeldin A or the effector NleA prevented TRPV2 activation and cell death. While infection with EPECΔnleA triggered pyroptotic cell death, this could be prevented by NleF. Taken together this study shows that while integration of Tir into the plasma membrane activates TRPV2, EPEC uses NleA to inhibit TRPV2 trafficking and NleF to inhibit caspase-4 and pyroptosis.
Collapse
Affiliation(s)
- Qiyun Zhong
- Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Sharanya Chatterjee
- Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Jyoti S Choudhary
- Functional Proteomics Group, Chester Beatty Laboratories, The Institute of Cancer Research, London, UK
| | - Gad Frankel
- Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| |
Collapse
|
10
|
Taban Q, Mumtaz PT, Masoodi KZ, Haq E, Ahmad SM. Scavenger receptors in host defense: from functional aspects to mode of action. Cell Commun Signal 2022; 20:2. [PMID: 34980167 PMCID: PMC8721182 DOI: 10.1186/s12964-021-00812-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/27/2021] [Indexed: 12/17/2022] Open
Abstract
Scavenger receptors belong to a superfamily of proteins that are structurally heterogeneous and encompass the miscellaneous group of transmembrane proteins and soluble secretory extracellular domain. They are functionally diverse as they are involved in various disorders and biological pathways and their major function in innate immunity and homeostasis. Numerous scavenger receptors have been discovered so far and are apportioned in various classes (A-L). Scavenger receptors are documented as pattern recognition receptors and known to act in coordination with other co-receptors such as Toll-like receptors in generating the immune responses against a repertoire of ligands such as microbial pathogens, non-self, intracellular and modified self-molecules through various diverse mechanisms like adhesion, endocytosis and phagocytosis etc. Unlike, most of the scavenger receptors discussed below have both membrane and soluble forms that participate in scavenging; the role of a potential scavenging receptor Angiotensin-Converting Enzyme-2 has also been discussed whereby only its soluble form might participate in preventing the pathogen entry and replication, unlike its membrane-bound form. This review majorly gives an insight on the functional aspect of scavenger receptors in host defence and describes their mode of action extensively in various immune pathways involved with each receptor type. Video abstract.
Collapse
Affiliation(s)
- Qamar Taban
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology - Kashmir, Shuhama, 190006, India.,Department of Biotechnology, University of Kashmir, Hazratbal Srinagar, Kashmir, India
| | | | - Khalid Z Masoodi
- Division of Plant Biotechnology, Transcriptomics Laboratory, SKUAST-K, Shalimar, India
| | - Ehtishamul Haq
- Department of Biotechnology, University of Kashmir, Hazratbal Srinagar, Kashmir, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e- Kashmir University of Agricultural Sciences and Technology - Kashmir, Shuhama, 190006, India.
| |
Collapse
|
11
|
Cao H, Huang W. HDL and Sepsis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:129-139. [DOI: 10.1007/978-981-19-1592-5_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
12
|
Su X, Ramírez-Escudero M, Sun F, van den Dikkenberg JB, van Steenbergen MJ, Pieters RJ, Janssen BJC, van Hasselt PM, Hennink WE, van Nostrum CF. Internalization and Transport of PEGylated Lipid-Based Mixed Micelles across Caco-2 Cells Mediated by Scavenger Receptor B1. Pharmaceutics 2021; 13:2022. [PMID: 34959304 PMCID: PMC8703698 DOI: 10.3390/pharmaceutics13122022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to get insight into the internalization and transport of PEGylat-ed mixed micelles loaded by vitamin K, as mediated by Scavenger Receptor B1 (SR-B1) that is abundantly expressed by intestinal epithelium cells as well as by differentiated Caco-2 cells. Inhibition of SR-B1 reduced endocytosis and transport of vitamin-K-loaded 0%, 30% and 50% PEGylated mixed micelles and decreased colocalization of the micelles with SR-B1. Confocal fluorescence microscopy, fluorescence-activated cell sorting (FACS) analysis, and surface plasmon resonance (SPR) were used to study the interaction between the mixed micelles of different compositions (varying vitamin K loading and PEG content) and SR-B1. Interaction of PEGylated micelles was independent of the vitamin K content, indicating that the PEG shell prevented vitamin K exposure at the surface of the micelles and binding with the receptor and that the PEG took over the micelles' ability to bind to the receptor. Molecular docking calculations corroborated the dual binding of both vita-min K and PEG with the binding domain of SR-B1. In conclusion, the improved colloidal stability of PEGylated mixed micelles did not compromise their cellular uptake and transport due to the affinity of PEG for SR-B1. SR-B1 is able to interact with PEGylated nanoparticles and mediates their subsequent internalization and transport.
Collapse
Affiliation(s)
- Xiangjie Su
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (X.S.); (F.S.); (J.B.v.d.D.); (M.J.v.S.); (W.E.H.)
| | - Mercedes Ramírez-Escudero
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.R.-E.); (B.J.C.J.)
| | - Feilong Sun
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (X.S.); (F.S.); (J.B.v.d.D.); (M.J.v.S.); (W.E.H.)
| | - Joep B. van den Dikkenberg
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (X.S.); (F.S.); (J.B.v.d.D.); (M.J.v.S.); (W.E.H.)
| | - Mies J. van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (X.S.); (F.S.); (J.B.v.d.D.); (M.J.v.S.); (W.E.H.)
| | - Roland J. Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Bert J. C. Janssen
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.R.-E.); (B.J.C.J.)
| | - Peter M. van Hasselt
- Department of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands;
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (X.S.); (F.S.); (J.B.v.d.D.); (M.J.v.S.); (W.E.H.)
| | - Cornelus F. van Nostrum
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (X.S.); (F.S.); (J.B.v.d.D.); (M.J.v.S.); (W.E.H.)
| |
Collapse
|
13
|
Fernandes das Neves M, Batuca JR, Delgado Alves J. The role of high-density lipoprotein in the regulation of the immune response: implications for atherosclerosis and autoimmunity. Immunology 2021; 164:231-241. [PMID: 33934336 PMCID: PMC8442240 DOI: 10.1111/imm.13348] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/29/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
Inflammation and immune dysfunction have been increasingly recognized as crucial mechanisms in atherogenesis. Modifications in cell lipid metabolism, plasma dyslipidaemia and particularly low high-density lipoprotein (HDL) levels occur both in atherosclerosis and in autoimmune rheumatic diseases (which are strongly associated with an increased risk of atherosclerosis), suggesting the presence of a crucial link. HDL, the plasma lipoprotein responsible for reverse cholesterol transport, is known for its several protective effects in the context of atherosclerosis. Among these, HDL immunomodulatory effects are possibly the less understood. Through the efflux of cholesterol from plasma cell membranes with the consequent disruption of lipid rafts and the interaction with the cholesterol transporters present in the plasma membrane, HDL affects both the innate and adaptive immune responses. Animal and human studies have demonstrated a predominance of HDL anti-inflammatory effects, despite some pro-inflammatory actions having also been reported. The HDL role on the modulation of the immune response is further suggested by the detection of low levels together with a dysfunctional HDL in patients with autoimmune diseases. Here, we review the current knowledge of the immune mechanisms of atherosclerosis and the modulatory effects HDL may have on them.
Collapse
Affiliation(s)
- Marisa Fernandes das Neves
- Center of the Study of Chronic DiseasesNew University of LisbonLisbonPortugal
- Medicine 4 DepartmentFernando Fonseca HospitalAmadoraPortugal
| | - Joana R. Batuca
- Center of the Study of Chronic DiseasesNew University of LisbonLisbonPortugal
| | - José Delgado Alves
- Center of the Study of Chronic DiseasesNew University of LisbonLisbonPortugal
- Medicine 4 DepartmentFernando Fonseca HospitalAmadoraPortugal
| |
Collapse
|
14
|
Baranova IN, Bocharov AV, Vishnyakova TG, Chen Z, Birukova AA, Ke Y, Hu X, Yuen PST, Star RA, Birukov KG, Patterson AP, Eggerman TL. Class B Scavenger Receptors BI and BII Protect against LPS-Induced Acute Lung Injury in Mice by Mediating LPS. Infect Immun 2021; 89:e0030121. [PMID: 34097506 PMCID: PMC8445172 DOI: 10.1128/iai.00301-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
Recent studies suggest an anti-inflammatory protective role for class B scavenger receptor BI (SR-BI) in endotoxin-induced inflammation and sepsis. Other data, including ours, provide evidence for an alternative role of SR-BI, facilitating bacterial and endotoxin uptake and contributing to inflammation and bacterial infection. Enhanced endotoxin susceptibility of SR-BI-deficient mice due to their anti-inflammatory glucocorticoid deficiency complicates the understanding of SR-BI's role in endotoxemia/sepsis, calling for the use of alternative models. In this study, using human SR-BI (hSR-BI) and hSR-BII transgenic mice, we found that SR-BI and, to a lesser extent, its splicing variant SR-BII protect against LPS-induced lung damage. At 20 h after intratracheal LPS instillation, the extent of pulmonary inflammation and vascular leakage was significantly lower in hSR-BI and hSR-BII transgenic mice than in wild-type mice. Higher bronchoalveolar lavage fluid (BALF) inflammatory cell count and protein content and lung tissue neutrophil infiltration found in wild-type mice were associated with markedly (2 to 3 times) increased proinflammatory cytokine production compared to these parameters in transgenic mice following LPS administration. The markedly lower endotoxin levels detected in BALF of transgenic versus wild-type mice and the significantly increased BODIPY-LPS uptake observed in lungs of hSR-BI and hSR-BII mice 20 h after the i.t. LPS injection suggest that hSR-BI- and hSR-BII-mediated enhanced LPS clearance in the airways could represent the mechanism of their protective role against LPS-induced acute lung injury.
Collapse
Affiliation(s)
- Irina N. Baranova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander V. Bocharov
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Tatyana G. Vishnyakova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhigang Chen
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna A. Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter S. T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Amy P. Patterson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas L. Eggerman
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
- National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
15
|
Abstract
High-density lipoprotein (HDL) particles, best known for their anti-atherosclerotic effects, also may play a beneficial role during acute renal stress. HDL from healthy human beings also shows anti-inflammatory and anti-oxidant capacities, promotes endothelial function and repair, and serves as a systemic signaling mechanism facilitating rapid interorgan communication during times of physiologic stress. Higher concentrations of HDL are associated with less acute kidney injury after sepsis, cardiac and vascular surgery, and contrast-exposure during percutaneous coronary interventions. A better understanding of the interplay between HDL and the kidney both under homeostatic conditions and under acute physiologic stress could lead to the identification of novel risk factors and therapeutic targets for acute kidney injury prevention and treatment in the future.
Collapse
Affiliation(s)
- Loren E Smith
- Division of Multispecialty Adult Anesthesiology, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
16
|
Morris G, Puri BK, Bortolasci CC, Carvalho A, Berk M, Walder K, Moreira EG, Maes M. The role of high-density lipoprotein cholesterol, apolipoprotein A and paraoxonase-1 in the pathophysiology of neuroprogressive disorders. Neurosci Biobehav Rev 2021; 125:244-263. [PMID: 33657433 DOI: 10.1016/j.neubiorev.2021.02.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Lowered high-density lipoprotein (HDL) cholesterol has been reported in major depressive disorder, bipolar disorder, first episode of psychosis, and schizophrenia. HDL, its major apolipoprotein component, ApoA1, and the antioxidant enzyme paraoxonase (PON)1 (which is normally bound to ApoA1) all have anti-atherogenic, antioxidant, anti-inflammatory, and immunomodulatory roles, which are discussed in this paper. The paper details the pathways mediating the anti-inflammatory effects of HDL, ApoA1 and PON1 and describes the mechanisms leading to compromised HDL and PON1 levels and function in an environment of chronic inflammation. The molecular mechanisms by which changes in HDL, ApoA1 and PON1 might contribute to the pathophysiology of the neuroprogressive disorders are explained. Moreover, the anti-inflammatory actions of ApoM-mediated sphingosine 1-phosphate (S1P) signalling are reviewed as well as the deleterious effects of chronic inflammation and oxidative stress on ApoM/S1P signalling. Finally, therapeutic interventions specifically aimed at improving the levels and function of HDL and PON1 while reducing levels of inflammation and oxidative stress are considered. These include the so-called Mediterranean diet, extra virgin olive oil, polyphenols, flavonoids, isoflavones, pomegranate juice, melatonin and the Mediterranean diet combined with the ketogenic diet.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | | | - Chiara C Bortolasci
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.
| | - Andre Carvalho
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia
| | - Estefania G Moreira
- Post-Graduation Program in Health Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Michael Maes
- Deakin University, IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
17
|
Abumrad NA, Cabodevilla AG, Samovski D, Pietka T, Basu D, Goldberg IJ. Endothelial Cell Receptors in Tissue Lipid Uptake and Metabolism. Circ Res 2021; 128:433-450. [PMID: 33539224 DOI: 10.1161/circresaha.120.318003] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipid uptake and metabolism are central to the function of organs such as heart, skeletal muscle, and adipose tissue. Although most heart energy derives from fatty acids (FAs), excess lipid accumulation can cause cardiomyopathy. Similarly, high delivery of cholesterol can initiate coronary artery atherosclerosis. Hearts and arteries-unlike liver and adrenals-have nonfenestrated capillaries and lipid accumulation in both health and disease requires lipid movement from the circulation across the endothelial barrier. This review summarizes recent in vitro and in vivo findings on the importance of endothelial cell receptors and uptake pathways in regulating FAs and cholesterol uptake in normal physiology and cardiovascular disease. We highlight clinical and experimental data on the roles of ECs in lipid supply to tissues, heart, and arterial wall in particular, and how this affects organ metabolism and function. Models of FA uptake into ECs suggest that receptor-mediated uptake predominates at low FA concentrations, such as during fasting, whereas FA uptake during lipolysis of chylomicrons may involve paracellular movement. Similarly, in the setting of an intact arterial endothelial layer, recent and historic data support a role for receptor-mediated processes in the movement of lipoproteins into the subarterial space. We conclude with thoughts on the need to better understand endothelial lipid transfer for fuller comprehension of the pathophysiology of hyperlipidemia, and lipotoxic diseases such as some forms of cardiomyopathy and atherosclerosis.
Collapse
Affiliation(s)
- Nada A Abumrad
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Dmitri Samovski
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Terri Pietka
- Division of Nutritional Sciences, Department of Medicine, Washington University School of Medicine, Saint Louis, MO (N.A.A., D.S., T.P.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine (A.G.C., D.B., I.J.G.)
| |
Collapse
|
18
|
Zhong Q, Roumeliotis TI, Kozik Z, Cepeda-Molero M, Fernández LÁ, Shenoy AR, Bakal C, Frankel G, Choudhary JS. Clustering of Tir during enteropathogenic E. coli infection triggers calcium influx-dependent pyroptosis in intestinal epithelial cells. PLoS Biol 2020; 18:e3000986. [PMID: 33378358 PMCID: PMC7773185 DOI: 10.1371/journal.pbio.3000986] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/24/2020] [Indexed: 11/19/2022] Open
Abstract
Clustering of the enteropathogenic Escherichia coli (EPEC) type III secretion system (T3SS) effector translocated intimin receptor (Tir) by intimin leads to actin polymerisation and pyroptotic cell death in macrophages. The effect of Tir clustering on the viability of EPEC-infected intestinal epithelial cells (IECs) is unknown. We show that EPEC induces pyroptosis in IECs in a Tir-dependent but actin polymerisation-independent manner, which was enhanced by priming with interferon gamma (IFNγ). Mechanistically, Tir clustering triggers rapid Ca2+ influx, which induces lipopolysaccharide (LPS) internalisation, followed by activation of caspase-4 and pyroptosis. Knockdown of caspase-4 or gasdermin D (GSDMD), translocation of NleF, which blocks caspase-4 or chelation of extracellular Ca2+, inhibited EPEC-induced cell death. IEC lines with low endogenous abundance of GSDMD were resistant to Tir-induced cell death. Conversely, ATP-induced extracellular Ca2+ influx enhanced cell death, which confirmed the key regulatory role of Ca2+ in EPEC-induced pyroptosis. We reveal a novel mechanism through which infection with an extracellular pathogen leads to pyroptosis in IECs.
Collapse
Affiliation(s)
- Qiyun Zhong
- Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, United Kingdom
| | - Theodoros I. Roumeliotis
- Functional Proteomics Group, Chester Beatty Laboratories, The Institute of Cancer Research, London, United Kingdom
| | - Zuza Kozik
- Functional Proteomics Group, Chester Beatty Laboratories, The Institute of Cancer Research, London, United Kingdom
| | - Massiel Cepeda-Molero
- Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, United Kingdom
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus UAM-Cantoblanco, Madrid, Spain
| | - Avinash R. Shenoy
- Centre for Molecular Bacteriology & Infection, Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Chris Bakal
- Dynamical Cell Systems, Chester Beatty Laboratories, The Institute of Cancer Research, London, United Kingdom
| | - Gad Frankel
- Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, United Kingdom
| | - Jyoti S. Choudhary
- Functional Proteomics Group, Chester Beatty Laboratories, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
19
|
Kudinov VA, Alekseeva OY, Torkhovskaya TI, Baskaev KK, Artyushev RI, Saburina IN, Markin SS. High-Density Lipoproteins as Homeostatic Nanoparticles of Blood Plasma. Int J Mol Sci 2020; 21:E8737. [PMID: 33228032 PMCID: PMC7699323 DOI: 10.3390/ijms21228737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/14/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023] Open
Abstract
It is well known that blood lipoproteins (LPs) are multimolecular complexes of lipids and proteins that play a crucial role in lipid transport. High-density lipoproteins (HDL) are a class of blood plasma LPs that mediate reverse cholesterol transport (RCT)-cholesterol transport from the peripheral tissues to the liver. Due to this ability to promote cholesterol uptake from cell membranes, HDL possess antiatherogenic properties. This function was first observed at the end of the 1970s to the beginning of the 1980s, resulting in high interest in this class of LPs. It was shown that HDL are the prevalent class of LPs in several types of living organisms (from fishes to monkeys) with high resistance to atherosclerosis and cardiovascular disorders. Lately, understanding of the mechanisms of the antiatherogenic properties of HDL has significantly expanded. Besides the contribution to RCT, HDL have been shown to modulate inflammatory processes, blood clotting, and vasomotor responses. These particles also possess antioxidant properties and contribute to immune reactions and intercellular signaling. Herein, we review data on the structure and mechanisms of the pleiotropic biological functions of HDL from the point of view of their evolutionary role and complex dynamic nature.
Collapse
Affiliation(s)
- Vasily A. Kudinov
- Laboratory of Cell Biology and Developmental Pathology, FSBSI Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
- Experimental Drug Research and Production Department, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (K.K.B.); (R.I.A.)
| | - Olga Yu. Alekseeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia;
- Department of Biochemistry, People’s Friendship University (RUDN University), 117198 Moscow, Russia
| | - Tatiana I. Torkhovskaya
- Laboratory of Phospholipid Transport Systems and Nanomedicines, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | - Konstantin K. Baskaev
- Experimental Drug Research and Production Department, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (K.K.B.); (R.I.A.)
| | - Rafael I. Artyushev
- Experimental Drug Research and Production Department, Institute of Biomedical Chemistry, 119121 Moscow, Russia; (K.K.B.); (R.I.A.)
| | - Irina N. Saburina
- Laboratory of Cell Biology and Developmental Pathology, FSBSI Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
| | - Sergey S. Markin
- Clinical Research Department, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| |
Collapse
|
20
|
Vishnyakova TG, Bocharov AV, Baranova IN, Kurlander R, Drake SK, Chen Z, Amar M, Sviridov D, Vaisman B, Poliakov E, Remaley AT, Eggerman TL, Patterson AP. SR-BI mediates neutral lipid sorting from LDL to lipid droplets and facilitates their formation. PLoS One 2020; 15:e0240659. [PMID: 33057430 PMCID: PMC7561250 DOI: 10.1371/journal.pone.0240659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 09/30/2020] [Indexed: 12/30/2022] Open
Abstract
SR-BI binds various lipoproteins, including HDL, LDL as well as VLDL, and mediates selective cholesteryl ester (CE) uptake. HDL derived CE accumulates in cellular lipid droplets (LDs), which also store triacylglycerol (TAG). We hypothesized that SR-BI could significantly facilitate LD formation, in part, by directly transporting LDL derived neutral lipids (NL) such as CE and TAG into LDs without lipolysis and de novo lipid synthesis. SR-BI overexpression greatly increased LDL uptake and LD formation in stably transfected HeLa cells (SR-BI-HeLa). LDs isolated from SR-BI-HeLa contained 4- and 7-times more CE and TAG, respectively, than mock-transfected HeLa (Mock-HeLa). In contrast, LDL receptor overexpression in HeLa (LDLr-HeLa) greatly increased LDL uptake, degradation with moderate 1.5- and 2-fold increases of CE and TAG, respectively. Utilizing CE and TAG analogs, BODIPY-TAG (BP-TAG) and BODIPY-CE (BP-CE), for tracking LDL NL, we found that after initial binding of LDL to SR-BI-HeLa, apoB remained at the cell surface, while BP-CE and BP-TAG were sorted and simultaneously transported together to LDs. Both lipids demonstrated limited internalization to lysosomes or endoplasmic reticulum in SR-BI-HeLa. In LDLr-HeLa, NLs demonstrated clear lysosomal sequestration without their sorting to LDs. An inhibition of TAG and CE de novo synthesis by 90-95% only reduced TAG and CE LD content by 45-50%, and had little effect on BP-CE and BP-TAG transport to LDs in SR-BI HeLa. Furthermore, intravenous infusion of 1-2 mg of LDL increased liver LDs in normal (WT) but not in SR-BI KO mice. Mice transgenic for human SR-BI demonstrated higher liver LD accumulation than WT mice. Finally, Electro Spray Infusion Mass Spectrometry (ESI-MS) using deuterated d-CE found that LDs accumulated up to 40% of unmodified d-CE LDL. We conclude that SR-BI mediates LDL-induced LD formation in vitro and in vivo. In addition to cytosolic NL hydrolysis and de novo lipid synthesis, this process includes selective sorting and transport of LDL NL to LDs with limited lysosomal NL sequestration and the transport of LDL CE, and TAG directly to LDs independently of de novo synthesis.
Collapse
Affiliation(s)
- Tatyana G. Vishnyakova
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
| | - Alexander V. Bocharov
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
- * E-mail:
| | - Irina N. Baranova
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
| | - Roger Kurlander
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
| | - Steven K. Drake
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
| | - Zhigang Chen
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
| | - Marcelo Amar
- National Heart, Lung and Blood Institute, Bethesda, Maryland, United
States of America
| | - Denis Sviridov
- National Heart, Lung and Blood Institute, Bethesda, Maryland, United
States of America
| | - Boris Vaisman
- National Heart, Lung and Blood Institute, Bethesda, Maryland, United
States of America
| | - Eugenia Poliakov
- National Eye Institute, Bethesda, Maryland, United States of
America
| | - Alan T. Remaley
- National Heart, Lung and Blood Institute, Bethesda, Maryland, United
States of America
| | - Thomas L. Eggerman
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
- National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda,
Maryland, United States of America
| | - Amy P. Patterson
- Clinical Center, The National Institutes of Health, Bethesda, Maryland,
United States of America
- National Heart, Lung and Blood Institute, Bethesda, Maryland, United
States of America
| |
Collapse
|
21
|
Kruglikov IL, Shah M, Scherer PE. Obesity and diabetes as comorbidities for COVID-19: Underlying mechanisms and the role of viral-bacterial interactions. eLife 2020; 9:e61330. [PMID: 32930095 PMCID: PMC7492082 DOI: 10.7554/elife.61330] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity and diabetes are established comorbidities for COVID-19. Adipose tissue demonstrates high expression of ACE2 which SARS- CoV-2 exploits to enter host cells. This makes adipose tissue a reservoir for SARS-CoV-2 viruses and thus increases the integral viral load. Acute viral infection results in ACE2 downregulation. This relative deficiency can lead to disturbances in other systems controlled by ACE2, including the renin-angiotensin system. This will be further increased in the case of pre-conditions with already compromised functioning of these systems, such as in patients with obesity and diabetes. Here, we propose that interactions of virally-induced ACE2 deficiency with obesity and/or diabetes leads to a synergistic further impairment of endothelial and gut barrier function. The appearance of bacteria and/or their products in the lungs of obese and diabetic patients promotes interactions between viral and bacterial pathogens, resulting in a more severe lung injury in COVID-19.
Collapse
Affiliation(s)
| | - Manasi Shah
- Division of Endocrinology, University of Texas Southwestern Medical CenterDallasUnited States
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical CenterDallasUnited States
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Cell Biology, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
22
|
Anand PK. Lipids, inflammasomes, metabolism, and disease. Immunol Rev 2020; 297:108-122. [PMID: 32562313 DOI: 10.1111/imr.12891] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/19/2020] [Accepted: 05/27/2020] [Indexed: 12/19/2022]
Abstract
Inflammasomes are multi-protein complexes that regulate the cleavage of cysteine protease caspase-1, secretion of inflammatory cytokines, and induction of inflammatory cell death, pyroptosis. Several members of the nod-like receptor family assemble inflammasome in response to specific ligands. An exception to this is the NLRP3 inflammasome which is activated by structurally diverse entities. Recent studies have suggested that NLRP3 might be a sensor of cellular homeostasis, and any perturbation in distinct metabolic pathways results in the activation of this inflammasome. Lipid metabolism is exceedingly important in maintaining cellular homeostasis, and it is recognized that cells and tissues undergo extensive lipid remodeling during activation and disease. Some lipids are involved in instigating chronic inflammatory diseases, and new studies have highlighted critical upstream roles for lipids, particularly cholesterol, in regulating inflammasome activation implying key functions for inflammasomes in diseases with defective lipid metabolism. The focus of this review is to highlight how lipids regulate inflammasome activation and how this leads to the progression of inflammatory diseases. The key roles of cholesterol metabolism in the activation of inflammasomes have been comprehensively discussed. Besides, the roles of oxysterols, fatty acids, phospholipids, and lipid second messengers are also summarized in the context of inflammasomes. The overriding theme is that lipid metabolism has numerous but complex functions in inflammasome activation. A detailed understanding of this area will help us develop therapeutic interventions for diseases where dysregulated lipid metabolism is the underlying cause.
Collapse
Affiliation(s)
- Paras K Anand
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
23
|
Li C, Ge X, Su B, Fu Q, Wang B, Liu X, Ren Y, Song L, Yang N. Characterization of class B scavenger receptor type 1 (SRB1) in turbot (Scophthalmus maximus L.). FISH & SHELLFISH IMMUNOLOGY 2020; 100:358-367. [PMID: 32169665 DOI: 10.1016/j.fsi.2020.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 06/10/2023]
Abstract
Class B scavenger receptor type 1 (SRB1) serves as a high-density lipoprotein (HDL) receptor essential for HDL metabolism, and plays vital roles in innate immunity. In this study, the turbot (Scophthalmus maximus) SRB1 was cloned and characterized. The gene structure consists of a coding region of 1,527 bp nucleotides dividing into 13 exons and 12 introns. Such genome structure is highly conserved among teleost fishes. The deduced SRB1 encodes 508 amino acids that mainly has a CD36 transmembrane domain. Tissue distribution of SRB1 showed the lowest expression in liver, while the highest expression was found in intestine. Significantly down-regulation pattern of SmSRB1 expression in intestine was shared after infection with Vibrio anguillarum and Streptococcus iniae. Brach and site models in CODEML program showed that SmSRB1 underwent a conservative evolutionary and three potential positive selected sites 470K, 496E, and 501Y were detected, which requires further investigation and confirmation using base-editing technologies. Subcellular localization demonstrated that turbot SRB1 was distributed in the membrane and cytoplasm. rSmSRB1 showed binding ability in vitro to bacteria, LPS, PGN, LTA and virus. Protein-protein interaction network agrees the function of SRB1 as lipoprotein receptor. Our results indicated SmSRB1 might act as co-receptors to TLRs and NLRs to modulate the immune response to pathogens. Further studies should pay attention to evaluate the specific co-receptor for SRB1 in recognition of different pathogens and selective mechanisms involved.
Collapse
Affiliation(s)
- Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xuefeng Ge
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Baofeng Su
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Qiang Fu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Beibei Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xiaoli Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yichao Ren
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Lin Song
- College of Marine Science and Biological Engineering, Qingdao University of Science & Technology, Qingdao, 266011, China
| | - Ning Yang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
24
|
Trinder M, Genga KR, Kong HJ, Blauw LL, Lo C, Li X, Cirstea M, Wang Y, Rensen PCN, Russell JA, Walley KR, Boyd JH, Brunham LR. Cholesteryl Ester Transfer Protein Influences High-Density Lipoprotein Levels and Survival in Sepsis. Am J Respir Crit Care Med 2020; 199:854-862. [PMID: 30321485 DOI: 10.1164/rccm.201806-1157oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
RATIONALE High-density lipoprotein (HDL) cholesterol (HDL-C) levels decline during sepsis, and lower levels are associated with worse survival. However, the genetic mechanisms underlying changes in HDL-C during sepsis, and whether the relationship with survival is causative, are largely unknown. OBJECTIVES We hypothesized that variation in genes involved in HDL metabolism would contribute to changes in HDL-C levels and clinical outcomes during sepsis. METHODS We performed targeted resequencing of HDL-related genes in 200 patients admitted to an emergency department with sepsis (Early Infection cohort). We examined the association of genetic variants with HDL-C levels, 28-day survival, 90-day survival, organ dysfunction, and need for vasopressor or ventilatory support. Candidate variants were further assessed in the VASST (Vasopressin versus Norepinephrine Infusion in Patients with Septic Shock Trial) cohort (n = 632) and St. Paul's Hospital Intensive Care Unit 2 (SPHICU2) cohort (n = 203). MEASUREMENTS AND MAIN RESULTS We identified a rare missense variant in CETP (cholesteryl ester transfer protein gene; rs1800777-A) that was associated with significant reductions in HDL-C levels during sepsis. Carriers of the A allele (n = 10) had decreased survival, more organ failure, and greater need for organ support compared with noncarriers. We replicated this finding in the VASST and SPHICU2 cohorts, in which carriers of rs1800777-A (n = 35 and n = 12, respectively) had significantly reduced 28-day survival. Mendelian randomization was consistent with genetically reduced HDL levels being a causal factor for decreased sepsis survival. CONCLUSIONS Our results identify CETP as a critical regulator of HDL levels and clinical outcomes during sepsis. These data point toward a critical role for HDL in sepsis.
Collapse
Affiliation(s)
- Mark Trinder
- 1 Centre for Heart Lung Innovation and.,2 Experimental Medicine Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kelly R Genga
- 1 Centre for Heart Lung Innovation and.,2 Experimental Medicine Program, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Lisanne L Blauw
- 3 Department of Medicine, Division of Endocrinology and.,4 Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Cody Lo
- 1 Centre for Heart Lung Innovation and
| | - Xuan Li
- 1 Centre for Heart Lung Innovation and
| | | | - Yanan Wang
- 3 Department of Medicine, Division of Endocrinology and.,4 Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Patrick C N Rensen
- 3 Department of Medicine, Division of Endocrinology and.,4 Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands; and
| | - James A Russell
- 1 Centre for Heart Lung Innovation and.,5 Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Keith R Walley
- 1 Centre for Heart Lung Innovation and.,5 Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - John H Boyd
- 1 Centre for Heart Lung Innovation and.,2 Experimental Medicine Program, University of British Columbia, Vancouver, British Columbia, Canada.,5 Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liam R Brunham
- 1 Centre for Heart Lung Innovation and.,2 Experimental Medicine Program, University of British Columbia, Vancouver, British Columbia, Canada.,5 Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
El-Tahawy NFG, Abozaid SMM. The possible structural changes in the adrenal gland cortex after induction of hepatic ischemia-reperfusion injury in male albino rats: Light and electron microscopic study. J Cell Physiol 2019; 234:15487-15495. [PMID: 30684267 DOI: 10.1002/jcp.28196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
The adrenal gland is an important endocrine gland in the body that secrets the adrenal hormones. One of the important clinical issues is the hepatic ischemia-reperfusion (IR) injury. Liver IR injury results in many distant organs dysfunctions such as lung, kidney, intestine, pancreas, and myocardium. The aim of the present study was to investigate the possible remote effects of hepatic IR on the structure of the adrenal cortex. Twenty healthy males, Sprague-Dawley albino rats aged 6-8 weeks were randomly divided into two groups (10 rats each): the sham control group (SC-group) and the ischemia-reperfusion group (IR-group). Sera were estimated for the following: aspartate transaminase (AST), alanine transaminase (ALT), lactic dehydrogenase (LDH), and corticosterone levels. Also oxidative markers such as malondialdehyde (MDA) and tumor necrosis factor-α (TNF-α), and the antioxidative enzyme, catalase were measured. Adrenal glands were processed for light and transmission electron microscopic study. The results showed a significant increase in serum liver enzymes (AST, ALT, and LDH), corticosterone, MDA, and TNF-α levels and a significant decrease in serum levels of catalase in IR-group compared with SC-group. Adrenal cortical tissue of IR-group showed the loss of normal appearance. Some cells of zona glomerulosa and most of the zona fasciculata cells appeared swollen and degenerated with highly vacuolated cytoplasm. Other cells were shrunken with deeply acidophilic cytoplasm and pyknotic nuclei. Degenerated mitochondria with disrupted cristae, lipid droplets were confluent and dilated smooth endoplasmic reticulum were seen. Few zona reticularis cells had the dark nucleus and cytoplasmic vacuolations. In the different zones, blood capillaries were markedly congested and some inflammatory cells infiltrations were observed. Liver IR affected the structure of the adrenal cortex.
Collapse
|
26
|
Bakker GJ, Schnitzler JG, Bekkering S, de Clercq NC, Koopen AM, Hartstra AV, Meessen ECE, Scheithauer TP, Winkelmeijer M, Dallinga‐Thie GM, Cani PD, Kemper EM, Soeters MR, Kroon J, Groen AK, van Raalte DH, Herrema H, Nieuwdorp M. Oral vancomycin treatment does not alter markers of postprandial inflammation in lean and obese subjects. Physiol Rep 2019; 7:e14199. [PMID: 31423751 PMCID: PMC6698488 DOI: 10.14814/phy2.14199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 12/30/2022] Open
Abstract
Intake of a high-fat meal induces a systemic inflammatory response in the postprandial which is augmented in obese subjects. However, the underlying mechanisms of this response have not been fully elucidated. We aimed to assess the effect of gut microbiota modulation on postprandial inflammatory response in lean and obese subjects. Ten lean and ten obese subjects with metabolic syndrome received oral vancomycin 500 mg four times per day for 7 days. Oral high-fat meal tests (50 g fat/m2 body surface area) were performed before and after vancomycin intervention. Gut microbiota composition, leukocyte counts, plasma lipopolysaccharides (LPS), LPS-binding protein (LBP), IL-6 and MCP-1 concentrations and monocyte CCR2 and cytokine expression were determined before and after the high-fat meal. Oral vancomycin treatment resulted in profound changes in gut microbiota composition and significantly decreased bacterial diversity in both groups (phylogenetic diversity pre- versus post-intervention: lean, 56.9 ± 7.8 vs. 21.4 ± 6.6, P < 0.001; obese, 53.9 ± 7.8 vs. 21.0 ± 5.9, P < 0.001). After intervention, fasting plasma LPS significantly increased (lean, median [IQR] 0.81 [0.63-1.45] EU/mL vs. 2.23 [1.33-3.83] EU/mL, P = 0.017; obese, median [IQR] 0.76 [0.45-1.03] EU/mL vs. 1.44 [1.11-4.24], P = 0.014). However, postprandial increases in leukocytes and plasma LPS were unaffected by vancomycin in both groups. Moreover, we found no changes in plasma LBP, IL-6 and MCP-1 or in monocyte CCR2 expression. Despite major vancomycin-induced disruption of the gut microbiota and increased fasting plasma LPS, the postprandial inflammatory phenotype in lean and obese subjects was unaffected in this study.
Collapse
Affiliation(s)
- Guido J. Bakker
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Johan G. Schnitzler
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Siroon Bekkering
- Department of Experimental Internal MedicineRadboud University Medical CentreNijmegenThe Netherlands
| | - Nicolien C. de Clercq
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Annefleur M. Koopen
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Annick V. Hartstra
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Emma C. E. Meessen
- Department of Endocrinology and MetabolismAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Torsten P. Scheithauer
- Department of Internal Medicine, Diabetes CenterAmsterdam UMC, Location VUMC at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Maaike Winkelmeijer
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Geesje M. Dallinga‐Thie
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Patrice D. Cani
- WELBIO – Walloon Excellence in Life Sciences and Biotechnology, Metabolism and NutritionLouvain Drug Research Institute, Université Catholique de LouvainBrusselsBelgium
| | - Elles Marleen Kemper
- Department of Clinical PharmacyAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Maarten R. Soeters
- Department of Endocrinology and MetabolismAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Jeffrey Kroon
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Albert K. Groen
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Daniël H. van Raalte
- Department of Internal Medicine, Diabetes CenterAmsterdam UMC, Location VUMC at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam UMC, ICar at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Max Nieuwdorp
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
- Department of Internal Medicine, Diabetes CenterAmsterdam UMC, Location VUMC at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam UMC, ICar at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Molecular and Clinical Medicine, Wallenberg LaboratorySahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
27
|
Abstract
Mast cells (MCs) are found in tissues that are in close contact with external environment, such as skin, lungs, or intestinal mucosa but also in the placenta during pregnancy. If their role in mediating allergic conditions is established, several studies now highlight their importance during infection with extracellular pathogens. This study showed a new and effective antimicrobial mechanism of MCs against Coxiella burnetii, an intracellular bacterium whose infection during pregnancy is associated with abortion, preterm labor, and stillbirth. The data reveal that in response to C. burnetii, MCs release extracellular actin filaments that contain antimicrobial agents and are capable to trap and kill bacteria. We show that this mechanism is dependent on the cooperation of two membrane receptors, CD36 and Toll-like receptor 4, and may occur in the placenta during pregnancy by using ex vivo placental MCs. Overall, this study reports an unexpected role for MCs during infection with intracellular bacteria and suggests that MC response to C. burnetii infection is a protective defense mechanism during pregnancy. Mast cells (MCs) are critical mediators of inflammation; however, their microbicidal activity against invading pathogens remains largely unknown. Here, we describe a nonpreviously reported antibacterial mechanism used by MCs against Coxiella burnetii, the agent of Q fever. We show that C. burnetii interaction with MCs does not result in bacterial uptake but rather induces the formation of extracellular actin filaments named cytonemes. MC cytonemes express cathelicidin and neutrophil elastase and mediate the capture and destruction of entrapped bacteria. We provide evidence that MC cytoneme formation and microbicidal activity are dependent on the cooperation of the scavenger receptor CD36 and Toll-like receptor 4. Taken together, our results suggest that MCs use an extracellular sophisticated mechanism of defense to eliminate intracellular pathogens, such as C. burnetii, before their entry into host cells.
Collapse
|
28
|
Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol 2018; 14:576-590. [PMID: 30046148 DOI: 10.1038/s41574-018-0059-4] [Citation(s) in RCA: 1569] [Impact Index Per Article: 261.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ageing and age-related diseases share some basic mechanistic pillars that largely converge on inflammation. During ageing, chronic, sterile, low-grade inflammation - called inflammaging - develops, which contributes to the pathogenesis of age-related diseases. From an evolutionary perspective, a variety of stimuli sustain inflammaging, including pathogens (non-self), endogenous cell debris and misplaced molecules (self) and nutrients and gut microbiota (quasi-self). A limited number of receptors, whose degeneracy allows them to recognize many signals and to activate the innate immune responses, sense these stimuli. In this situation, metaflammation (the metabolic inflammation accompanying metabolic diseases) is thought to be the form of chronic inflammation that is driven by nutrient excess or overnutrition; metaflammation is characterized by the same mechanisms underpinning inflammaging. The gut microbiota has a central role in both metaflammation and inflammaging owing to its ability to release inflammatory products, contribute to circadian rhythms and crosstalk with other organs and systems. We argue that chronic diseases are not only the result of ageing and inflammaging; these diseases also accelerate the ageing process and can be considered a manifestation of accelerated ageing. Finally, we propose the use of new biomarkers (DNA methylation, glycomics, metabolomics and lipidomics) that are capable of assessing biological versus chronological age in metabolic diseases.
Collapse
Affiliation(s)
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden
- Laboratory of Cell Biology, Rizzoli Orthopaedic Institute, Bologna, Italy
- CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy
| | - Paolo Parini
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden
| | - Cristina Giuliani
- Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences (BiGeA), University of Bologna, Bologna, Italy.
- Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Bologna, Italy.
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Bologna, Italy
| |
Collapse
|
29
|
Haghikia A, Landmesser U. High-Density Lipoproteins: Effects on Vascular Function and Role in the Immune Response. Cardiol Clin 2018; 36:317-327. [PMID: 29609761 DOI: 10.1016/j.ccl.2017.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The focus in studies of high-density lipoproteins was on their capacity to remove excess cholesterol and deliver it to the liver. Other functions and vascular effects have been described. Clinical trials and translational/genetic studies have led to a refined understanding of the role of high-density lipoprotein; it is likely not a causal cardiovascular risk factor. In healthy subjects, it limits lipid oxidation, protects endothelial cell functions/integrity, and exerts antiinflammatory/antiapoptotic effects. In patients with coronary disease or diabetes, it undergoes modifications/remodeling, resulting in dysfunctional high-density lipoprotein. We summarize recent findings about the regulation of its function and discuss the clinical implications.
Collapse
Affiliation(s)
- Arash Haghikia
- Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin 12203, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Postfach 65 21 33, Berlin 13316, Germany.
| | - Ulf Landmesser
- Department of Cardiology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin 12203, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Postfach 65 21 33, Berlin 13316, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, Berlin 10178, Germany
| |
Collapse
|
30
|
Cheng N, Liang Y, Du X, Ye RD. Serum amyloid A promotes LPS clearance and suppresses LPS-induced inflammation and tissue injury. EMBO Rep 2018; 19:embr.201745517. [PMID: 30126923 DOI: 10.15252/embr.201745517] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 07/24/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Lipopolysaccharide (LPS) is a major microbial mediator for tissue injury and sepsis resulting from Gram-negative bacterial infection. LPS is an external factor that induces robust expression of serum amyloid A (SAA), a major constituent of the acute-phase proteins, but the relationship between SAA expression and LPS-induced tissue injury remains unclear. Here, we report that mice with inducible transgenic expression of human SAA1 are partially protected against inflammatory response and lung injury caused by LPS and cecal ligation and puncture (CLP). In comparison, transgenic SAA1 does not attenuate TNFα-induced lung inflammation and injury. The SAA1 expression level correlates inversely with the endotoxin concentrations in serum and lung tissues since SAA1 binds directly to LPS to form a complex that promotes LPS uptake by macrophages. Disruption of the SAA1-LPS interaction with a SAA1-derived peptide partially reduces the protective effect and exacerbates inflammation. These findings demonstrate that acute-phase SAA provides innate feedback protection against LPS-induced inflammation and tissue injury.
Collapse
Affiliation(s)
- Ni Cheng
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Yurong Liang
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Xiaoping Du
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Richard D Ye
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA .,State Key Laboratory for Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region, China
| |
Collapse
|
31
|
Cheng N, Liang Y, Du X, Ye RD. Serum amyloid A promotes
LPS
clearance and suppresses
LPS
‐induced inflammation and tissue injury. EMBO Rep 2018. [DOI: 10.15252/embr.201745517 (e45517):14 pp] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Ni Cheng
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
| | - Yurong Liang
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
| | - Xiaoping Du
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
| | - Richard D Ye
- Department of Pharmacology and Center for Lung and Vascular Biology College of Medicine University of Illinois Chicago IL USA
- State Key Laboratory for Quality Research in Chinese Medicine Institute of Chinese Medical Sciences University of Macau Macau Special Administrative Region China
| |
Collapse
|
32
|
Shen WJ, Asthana S, Kraemer FB, Azhar S. Scavenger receptor B type 1: expression, molecular regulation, and cholesterol transport function. J Lipid Res 2018; 59:1114-1131. [PMID: 29720388 DOI: 10.1194/jlr.r083121] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/26/2018] [Indexed: 12/16/2022] Open
Abstract
Cholesterol is required for maintenance of plasma membrane fluidity and integrity and for many cellular functions. Cellular cholesterol can be obtained from lipoproteins in a selective pathway of HDL-cholesteryl ester (CE) uptake without parallel apolipoprotein uptake. Scavenger receptor B type 1 (SR-B1) is a cell surface HDL receptor that mediates HDL-CE uptake. It is most abundantly expressed in liver, where it provides cholesterol for bile acid synthesis, and in steroidogenic tissues, where it delivers cholesterol needed for storage or steroidogenesis in rodents. SR-B1 transcription is regulated by trophic hormones in the adrenal gland, ovary, and testis; in the liver and elsewhere, SR-B1 is subject to posttranscriptional and posttranslational regulation. SR-B1 operates in several metabolic processes and contributes to pathogenesis of atherosclerosis, inflammation, hepatitis C virus infection, and other conditions. Here, we summarize characteristics of the selective uptake pathway and involvement of microvillar channels as facilitators of selective HDL-CE uptake. We also present the potential mechanisms of SR-B1-mediated selective cholesterol transport; the transcriptional, posttranscriptional, and posttranslational regulation of SR-B1; and the impact of gene variants on expression and function of human SR-B1. A better understanding of this unique pathway and SR-B1's role may yield improved therapies for a wide variety of conditions.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Geriatric Research, Education, and Clinical Research Center (GRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 and Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305
| | - Shailendra Asthana
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - Fredric B Kraemer
- Geriatric Research, Education, and Clinical Research Center (GRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 and Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305
| | - Salman Azhar
- Geriatric Research, Education, and Clinical Research Center (GRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 and Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
33
|
Guo J, Chang G, Zhang K, Xu L, Jin D, Bilal MS, Shen X. Rumen-derived lipopolysaccharide provoked inflammatory injury in the liver of dairy cows fed a high-concentrate diet. Oncotarget 2018; 8:46769-46780. [PMID: 28596485 PMCID: PMC5564522 DOI: 10.18632/oncotarget.18151] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/11/2017] [Indexed: 11/29/2022] Open
Abstract
Rumen-derived lipopolysaccharide (LPS) is translocated from the rumen into the bloodstream when subacute ruminal acidosis (SARA) occurs following long-term feeding with a high-concentrate (HC) diet in dairy cows. The objective of this study was to investigate the mechanism of inflammatory responses in the liver caused by HC diet feeding. We found that SARA was induced in dairy cows when rumen pH below 5.6 lasted for at least 3 h/d with HC diet feeding. Also, the LPS levels in the portal and hepatic veins were increased significantly and hepatocytes were impaired as well as the liver function was inhibited during SARA condition. Meanwhile, the mRNA expression of immune genes including TNF receptor associated factor 6 (TRAF6), nuclear factor-kappa B (NF-κB), p38 mitogen-activated protein kinase (MAPK), extracellular regulated protein kinases (ERK) MAPK, Interleukin-1 (IL-1) and serum amyloid A (SAA) in the liver were significantly increased in SARA cows. Moreover, the phosphorylation level of NF-κB p65 and p38 MAPK proteins in the liver and the concentration of Tumor Necrosis Factor (TNF-α), Interleukin-1β (IL-1β) and Interleukin-6 (IL-6) in peripheral blood were obviously increased under SARA condition. In conclusion, the inflammatory injury in the liver caused by LPS that traveled from the digestive tract to the liver through the portal vein after feeding with a HC diet.
Collapse
Affiliation(s)
- Junfei Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| | - Kai Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| | - Lei Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| | - Di Jin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| | - Muhammad Shahid Bilal
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| |
Collapse
|
34
|
D'Arrigo JS. Targeting Early Dementia: Using Lipid Cubic Phase Nanocarriers to Cross the Blood⁻Brain Barrier. Biomimetics (Basel) 2018; 3:E4. [PMID: 31105226 PMCID: PMC6352688 DOI: 10.3390/biomimetics3010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, a frequent co-morbidity of cerebrovascular pathology and Alzheimer's disease has been observed. Numerous published studies indicate that the preservation of a healthy cerebrovascular endothelium can be an important therapeutic target. By incorporating the appropriate drug(s) into biomimetic (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic, which targets certain cell surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood⁻brain barrier. This targeting allows for various cell types related to Alzheimer's to be simultaneously searched out for localized drug treatment in vivo.
Collapse
|
35
|
Role of microbiota-derived lipopolysaccharide in adipose tissue inflammation, adipocyte size and pyroptosis during obesity. Nutr Res Rev 2018; 31:153-163. [DOI: 10.1017/s0954422417000269] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractIt has been established that ingestion of a high-fat diet increases the blood levels of lipopolysaccharides (LPS) from Gram-negative bacteria in the gut. Obesity is characterised by low-grade systemic and adipose tissue inflammation. This is suggested to be implicated in the metabolic syndrome and obesity. In the present review, we hypothesise that LPS directly and indirectly participates in the inflammatory reaction in adipose tissue during obesity. The experimental evidence shows that LPS is involved in the transition of macrophages from the M2 to the M1 phenotype. In addition, LPS inside adipocytes may activate caspase-4/5/11. This may induce a highly inflammatory type of programmed cell death (i.e. pyroptosis), which also occurs after infection with intracellular pathogens. Lipoproteins with or without LPS are taken up by adipocytes. Large adipocytes are more metabolically active and potentially more exposed to LPS than small adipocytes are. Thus, LPS might be involved in defining the adipocyte death size and the formation of crown-like structures. The adipocyte death size is reached when the intracellular concentration of LPS initiates pyroptosis. The mechanistic details remain to be elucidated, but the observations indicate that adipocytes are stimulated to cell death by processes that involve LPS from the gut microbiota. There is a complex interplay between the composition of the diet and microbiota. This influences the amount of LPS that is translocated from the gut. In particular, the lipid content of a meal may correlate with the amount of LPS built in to chylomicrons.
Collapse
|
36
|
Nanotherapy for Alzheimer's disease and vascular dementia: Targeting senile endothelium. Adv Colloid Interface Sci 2018; 251:44-54. [PMID: 29274774 DOI: 10.1016/j.cis.2017.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Due to the complexity of Alzheimer's disease, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted lipid nanoemulsion) are available. Versatile small molecule drug(s) targeting multiple pathways of Alzheimer's disease pathogenesis are known. By incorporating such drug(s) into the targeted "lipid-coated microbubble" [LCM]/"nanoparticle-derived" [ND] (or LCM/ND) nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly class B type I), or SR-BI, making possible for various Alzheimer's-related cell types to be simultaneously searched out for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble [LCM] subpopulation (i.e., a stable LCM suspension); such film-stabilized microbubbles are well known to substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer's patient.
Collapse
|
37
|
Wei XB, Chen XJ, Li YL, Huang JL, Chen XL, Yu DQ, Tan N, Liu YH, Chen JY, He PC. Apolipoprotein A-I: A favorable prognostic marker in infective endocarditis. J Clin Lipidol 2017; 12:498-505. [PMID: 29339066 DOI: 10.1016/j.jacl.2017.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/10/2017] [Accepted: 12/12/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND Decreased apolipoprotein A-I (apoA-I) and high-density lipoprotein cholesterol (HDL-C) are common in inflammation and sepsis. No study with a large sample size has been performed to investigate the prognostic value of apoA-I or HDL-C in infective endocarditis (IE). OBJECTIVE The present study aimed to explore the prognostic value of apoA-I and HDL-C for adverse outcomes in IE patients. METHODS Patients with a definite diagnosis of IE between January 2009 and July 2015 were enrolled and divided into 3 groups according to their apoA-I tertiles at admission. Univariate and multivariate analyses were performed to evaluate the relationship of apoA-I and HDL-C with clinical outcomes. RESULTS Of the 593 included patients, 40 (6.7%) died in hospital. Patients with lower apoA-I experienced markedly higher rates of in-hospital mortality (10.7%, 7.0%, and 2.5% in tertiles 1-3, respectively; P = .006) and major adverse clinical events (32.5%, 24.1%, and 8.6% in tertiles 1-3, respectively; P < .001). ApoA-I (area under the curve, 0.671; P < .001) and HDL-C (area under the curve, 0.672; P < .001) had predictive values for in-hospital death. Multivariate logistic regression showed that apoA-I <0.90 g/L and HDL-C <0.78 mmol/L were independent risk predictors for in-hospital death. A multivariate Cox proportional hazard analysis revealed that apoA-I (increments of 1 g/L; hazard ratio, 0.36; 95% confidence interval, 0.15-0.87; P = .023) and HDL-C (increments of 1 mmol/L; hazard ratio, 0.38; 95% confidence interval, 0.18-0.83; P = .015) were independently associated with long-term mortality. CONCLUSIONS ApoA-I and HDL-C were inversely associated with adverse IE prognosis.
Collapse
Affiliation(s)
- Xue-Biao Wei
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Xiao-Jin Chen
- Department of Internal Medicine, Longnan Medicine Hospital, Ganzhou, China
| | - Yuan-Ling Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Jie-Leng Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Xiao-Lan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Dan-Qing Yu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Ning Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China
| | - Yuan-Hui Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China.
| | - Ji-Yan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China.
| | - Peng-Cheng He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong General Hospital, Guangdong Academic of Medical Sciences, Guangzhou, China; Department of Cardiology, The Second People's Hospital of Nanhai District, Guangdong General Hospital's Nanhai Hospital, Foshan, China.
| |
Collapse
|
38
|
Alzheimer’s Disease, Brain Injury, and C.N.S. Nanotherapy in Humans: Sonoporation Augmenting Drug Targeting. Med Sci (Basel) 2017. [PMCID: PMC5753658 DOI: 10.3390/medsci5040029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Owing to the complexity of neurodegenerative diseases, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted nanoemulsion) are available. Versatile small-molecule drug(s) targeting multiple pathways of Alzheimer’s disease pathogenesis are known. By incorporating such drug(s) into the targeted lipid-coated microbubble/nanoparticle-derived (LCM/ND) lipid nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly scavenger receptor class B type I (SR-BI)), making it possible for various Alzheimer’s-related cell types to be simultaneously sought for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble (LCM) subpopulation (i.e., a stable LCM suspension); such LCM substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer’s patient.
Collapse
|
39
|
Li D, Wang P, Wang P, Hu X, Chen F. Targeting the gut microbiota by dietary nutrients: A new avenue for human health. Crit Rev Food Sci Nutr 2017; 59:181-195. [DOI: 10.1080/10408398.2017.1363708] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Daotong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture; Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Pan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture; Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Pengpu Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture; Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Xiaosong Hu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture; Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Fang Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture; Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, China
| |
Collapse
|
40
|
Nyúl-Tóth Á, Kozma M, Nagyőszi P, Nagy K, Fazakas C, Haskó J, Molnár K, Farkas AE, Végh AG, Váró G, Galajda P, Wilhelm I, Krizbai IA. Expression of pattern recognition receptors and activation of the non-canonical inflammasome pathway in brain pericytes. Brain Behav Immun 2017; 64:220-231. [PMID: 28432035 DOI: 10.1016/j.bbi.2017.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/31/2017] [Accepted: 04/12/2017] [Indexed: 12/27/2022] Open
Abstract
Cerebral pericytes are mural cells embedded in the basement membrane of capillaries. Increasing evidence suggests that they play important role in controlling neurovascular functions, i.e. cerebral blood flow, angiogenesis and permeability of the blood-brain barrier. These cells can also influence neuroinflammation which is highly regulated by the innate immune system. Therefore, we systematically tested the pattern recognition receptor expression of brain pericytes. We detected expression of NOD1, NOD2, NLRC5, NLRP1-3, NLRP5, NLRP9, NLRP10 and NLRX mRNA in non-treated cells. Among the ten known human TLRs, TLR2, TLR4, TLR5, TLR6 and TLR10 were found to be expressed. Inflammatory mediators induced the expression of NLRA, NLRC4 and TLR9 and increased the levels of NOD2, TLR2, inflammasome-forming caspases and inflammasome-cleaved interleukins. Oxidative stress, on the other hand, upregulated expression of TLR10 and NLRP9. Activation of selected pattern recognition receptors can lead to inflammasome assembly and caspase-dependent secretion of IL-1β. TNF-α and IFN-γ increased the levels of pro-IL-1β and pro-caspase-1 proteins; however, no canonical activation of NLRP1, NLRP2, NLRP3 or NLRC4 inflammasomes could be observed in human brain vascular pericytes. On the other hand, we could demonstrate secretion of active IL-1β in response to non-canonical inflammasome activation, i.e. intracellular LPS or infection with E. coli bacteria. Our in vitro results indicate that pericytes might have an important regulatory role in neuroinflammation.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Mihály Kozma
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Péter Nagyőszi
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Krisztina Nagy
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Csilla Fazakas
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - János Haskó
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Attila E Farkas
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Attila G Végh
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - György Váró
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Péter Galajda
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary.
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary; Institute of Life Sciences, Vasile Goldiş Western University of Arad, Str. Liviu Rebreanu 86, 310414 Arad, Romania.
| | - István A Krizbai
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary; Institute of Life Sciences, Vasile Goldiş Western University of Arad, Str. Liviu Rebreanu 86, 310414 Arad, Romania.
| |
Collapse
|
41
|
Lipid testing in infectious diseases: possible role in diagnosis and prognosis. Infection 2017; 45:575-588. [PMID: 28484991 DOI: 10.1007/s15010-017-1022-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 05/04/2017] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Acute infections lead to significant alterations in metabolic regulation including lipids and lipoproteins, which play a central role in the host immune response. In this regard, several studies have investigated the role of lipid levels as a marker of infection severity and prognosis. SCOPE OF REVIEW We review here the role of lipids in immune response and the potential mechanisms underneath. Moreover, we summarize studies on lipid and lipoprotein alterations in acute bacterial, viral and parasitic infections as well as their diagnostic and prognostic significance. Chronic infections (HIV, HBV, HCV) are also considered. RESULTS All lipid parameters have been found to be significantly dearranged during acute infection. Common lipid alterations in this setting include a decrease of total cholesterol levels and an increase in the concentration of triglyceride-rich lipoproteins, mainly very low-density lipoproteins. Also, low-density lipoprotein cholesterol, apolipoprotein A1, low-density lipoprotein cholesterol and apolipoprotein-B levels decrease. These lipid alterations may have prognostic and diagnostic role in certain infections. CONCLUSION Lipid testing may be of help to assess response to treatment in septic patients and those with various acute infections (such as pneumonia, leptospirosis and others). Diagnostically, new onset of altered lipid levels should prompt the clinician to test for underlying infection (such as leishmaniasis).
Collapse
|
42
|
Baranova IN, Souza ACP, Bocharov AV, Vishnyakova TG, Hu X, Vaisman BL, Amar MJ, Chen Z, Remaley AT, Patterson AP, Yuen PST, Star RA, Eggerman TL. Human SR-BII mediates SAA uptake and contributes to SAA pro-inflammatory signaling in vitro and in vivo. PLoS One 2017; 12:e0175824. [PMID: 28423002 PMCID: PMC5396919 DOI: 10.1371/journal.pone.0175824] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/01/2017] [Indexed: 12/13/2022] Open
Abstract
Serum amyloid A (SAA) is an acute phase protein with cytokine-like and chemotactic properties, that is markedly up-regulated during various inflammatory conditions. Several receptors, including FPRL-1, TLR2, TLR4, RAGE, class B scavenger receptors, SR-BI and CD36, have been identified as SAA receptors. This study provides new evidence that SR-BII, splice variant of SR-BI, could function as an SAA receptor mediating its uptake and pro-inflammatory signaling. The uptake of Alexa Fluor488 SAA was markedly (~3 fold) increased in hSR-BII-expressing HeLa cells when compared with mock-transfected cells. The levels of SAA-induced interleukin-8 secretion by hSR-BII-expressing HEK293 cells were also significantly (~3-3.5 fold) higher than those detected in control cells. Moderately enhanced levels of phosphorylation of all three mitogen-activated protein kinases, ERK1/2, and p38 and JNK, were observed in hSR-BII-expressing cells following SAA stimulation when compared with control wild type cells. Transgenic mice with pLiv-11-directed liver/kidney overexpression of hSR-BI or hSR-BII were used to assess the in vivo role of each receptor in SAA-induced pro-inflammatory response in these organs. Six hours after intraperitoneal SAA injection both groups of transgenic mice demonstrated markedly higher (~2-5-fold) expression levels of inflammatory mediators in the liver and kidney compared to wild type mice. Histological examinations of hepatic and renal tissue from SAA-treated mice revealed moderate level of damage in the liver of both transgenic but not in the wild type mice. Activities of plasma transaminases, biomarkers of liver injury, were also moderately higher in hSR-B transgenic mice when compared to wild type mice. Our findings identify hSR-BII as a functional SAA receptor that mediates SAA uptake and contributes to its pro-inflammatory signaling via the MAPKs-mediated signaling pathways.
Collapse
Affiliation(s)
- Irina N. Baranova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ana C. P. Souza
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alexander V. Bocharov
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tatyana G. Vishnyakova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Boris L. Vaisman
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Marcelo J. Amar
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zhigang Chen
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alan T. Remaley
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy P. Patterson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter S. T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas L. Eggerman
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
43
|
Karathanasis SK, Freeman LA, Gordon SM, Remaley AT. The Changing Face of HDL and the Best Way to Measure It. Clin Chem 2016; 63:196-210. [PMID: 27879324 DOI: 10.1373/clinchem.2016.257725] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/26/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND HDL cholesterol (HDL-C) is a commonly used lipid biomarker for assessing cardiovascular health. While a central focus has been placed on the role of HDL in the reverse cholesterol transport (RCT) process, our appreciation for the other cardioprotective properties of HDL continues to expand with further investigation into the structure and function of HDL and its specific subfractions. The development of novel assays is empowering the research community to assess different aspects of HDL function, which at some point may evolve into new diagnostic tests. CONTENT This review discusses our current understanding of the formation and maturation of HDL particles via RCT, as well as the newly recognized roles of HDL outside RCT. The antioxidative, antiinflammatory, antiapoptotic, antithrombotic, antiinfective, and vasoprotective effects of HDL are all discussed, as are the related methodologies for assessing these different aspects of HDL function. We elaborate on the importance of protein and lipid composition of HDL in health and disease and highlight potential new diagnostic assays based on these parameters. SUMMARY Although multiple epidemiologic studies have confirmed that HDL-C is a strong negative risk marker for cardiovascular disease, several clinical and experimental studies have yielded inconsistent results on the direct role of HDL-C as an antiatherogenic factor. As of yet, our increased understanding of HDL biology has not been translated into successful new therapies, but will undoubtedly depend on the development of alternative ways for measuring HDL besides its cholesterol content.
Collapse
Affiliation(s)
| | - Lita A Freeman
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Scott M Gordon
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD.
| |
Collapse
|
44
|
Simonsen JB. Evaluation of reconstituted high-density lipoprotein (rHDL) as a drug delivery platform – a detailed survey of rHDL particles ranging from biophysical properties to clinical implications. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2161-2179. [DOI: 10.1016/j.nano.2016.05.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 12/15/2022]
|
45
|
Bertino G, Privitera G, Purrello F, Demma S, Crisafulli E, Spadaro L, Koukias N, Tsochatzis EA. Emerging hepatic syndromes: pathophysiology, diagnosis and treatment. Intern Emerg Med 2016; 11:905-16. [PMID: 27273018 DOI: 10.1007/s11739-016-1478-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/25/2016] [Indexed: 12/11/2022]
Abstract
Liver cirrhosis is a major cause of morbidity and mortality worldwide, mainly due to complications of portal hypertension. In this article, we review the current understanding on the pathophysiology, the diagnostic criteria and the available therapeutic options for patients with emerging hepatic syndromes in cirrhosis, namely the hepatorenal, hepato-adrenal and hepatopulmonary syndrome. The hepatorenal syndrome is a well-recognized complication of advanced cirrhosis and is usually associated with an accelerated course to death unless liver transplantation is performed. The hepatopulmonary syndrome is often missed in the evaluation of patients with cirrhosis; however, early recognition is essential for the efficient management of individual patients. The hepato-adrenal syndrome, although not fully characterized, offers an exciting field for research and potential therapeutic interventions.
Collapse
Affiliation(s)
- Gaetano Bertino
- Hepatology Unit, Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Catania, Italy
| | - Graziella Privitera
- Internal Medicine, Department of Clinical and Experimental Medicine, University of Catania, Ospedale Garibaldi-Nesima, Catania, Italy
| | - Francesco Purrello
- Internal Medicine, Department of Clinical and Experimental Medicine, University of Catania, Ospedale Garibaldi-Nesima, Catania, Italy
| | - Shirin Demma
- Hepatology Unit, Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Catania, Italy
| | - Emanuele Crisafulli
- Hepatology Unit, Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Catania, Italy
| | - Luisa Spadaro
- Internal Medicine, Department of Clinical and Experimental Medicine, University of Catania, Ospedale Garibaldi-Nesima, Catania, Italy
| | - Nikolaos Koukias
- UCL Institute for Liver and Digestive Health and Sheila Sherlock Liver Unit, Royal Free Hospital and UCL, London, UK
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health and Sheila Sherlock Liver Unit, Royal Free Hospital and UCL, London, UK.
| |
Collapse
|
46
|
Rajora MA, Zheng G. Targeting SR-BI for Cancer Diagnostics, Imaging and Therapy. Front Pharmacol 2016; 7:326. [PMID: 27729859 PMCID: PMC5037127 DOI: 10.3389/fphar.2016.00326] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/06/2016] [Indexed: 01/13/2023] Open
Abstract
Scavenger receptor class B type I (SR-BI) plays an important role in trafficking cholesteryl esters between the core of high density lipoprotein and the liver. Interestingly, this integral membrane protein receptor is also implicated in the metabolism of cholesterol by cancer cells, whereby overexpression of SR-BI has been observed in a number of tumors and cancer cell lines, including breast and prostate cancers. Consequently, SR-BI has recently gained attention as a cancer biomarker and exciting target for the direct cytosolic delivery of therapeutic agents. This brief review highlights these key developments in SR-BI-targeted cancer therapies and imaging probes. Special attention is given to the exploration of high density lipoprotein nanomimetic platforms that take advantage of upregulated SR-BI expression to facilitate targeted drug-delivery and cancer diagnostics, and promising future directions in the development of these agents.
Collapse
Affiliation(s)
- Maneesha A Rajora
- Princess Margaret Cancer Centre and Techna Institute, University Health NetworkToronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of TorontoToronto, ON, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre and Techna Institute, University Health NetworkToronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of TorontoToronto, ON, Canada; Department of Medical Biophysics, University of TorontoToronto, ON, Canada
| |
Collapse
|
47
|
Guerville M, Boudry G. Gastrointestinal and hepatic mechanisms limiting entry and dissemination of lipopolysaccharide into the systemic circulation. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1-G15. [PMID: 27151941 DOI: 10.1152/ajpgi.00098.2016] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/30/2016] [Indexed: 01/31/2023]
Abstract
The human microbiota consists of 100 trillion microorganisms that provide important metabolic and biological functions benefiting the host. However, the presence in host plasma of a gut-derived bacteria component, the lipopolysaccharide (LPS), has been identified as a causal or complicating factor in multiple serious diseases such as sepsis and septic shock and, more recently, obesity-associated metabolic disorders. Understanding the precise mechanisms by which gut-derived LPS is transported from the gut lumen to the systemic circulation is crucial to advance our knowledge of LPS-associated diseases and elaborate targeted strategies for their prevention. The aim of this review is to synthetize current knowledge on the host mechanisms limiting the entry and dissemination of LPS into the systemic circulation. To prevent bacterial colonization and penetration, the intestinal epithelium harbors multiple defense mechanisms including the secretion of antimicrobial peptides and mucins as well as detoxification enzymes. Despite this first line of defense, LPS can reach the apical site of intestinal epithelial cells (IECs) and, because of its large size, likely crosses IECs via transcellular transport, either lipid raft- or clathrin-mediated endocytosis or goblet cell-associated passage. However, the precise pathway remains poorly described. Finally, if LPS crosses the gut mucosa, it is directed via the portal vein to the liver, where major detoxification processes occur by deacetylation and excretion through the bile. If this disposal process is not sufficient, LPS enters the systemic circulation, where it is handled by numerous transport proteins that clear it back to the liver for further excretion.
Collapse
Affiliation(s)
| | - Gaëlle Boudry
- INRA UR1341 ADNC, Domaine de la Prise, Saint-Gilles, France
| |
Collapse
|
48
|
Foit L, Thaxton CS. Synthetic high-density lipoprotein-like nanoparticles potently inhibit cell signaling and production of inflammatory mediators induced by lipopolysaccharide binding Toll-like receptor 4. Biomaterials 2016; 100:67-75. [PMID: 27244690 DOI: 10.1016/j.biomaterials.2016.05.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/25/2022]
Abstract
Toll-like receptor 4 (TLR4) plays a critical role in the innate immune system. Stimulation of TLR4 occurs upon binding lipopolysaccharide (LPS), a component of Gram-negative bacterial cell walls. Due to the potency of the induced inflammatory response, there is a growing interest in agents that can most proximally modulate this LPS/TLR4 interaction to prevent downstream cell signaling events and the production of inflammatory mediators. Building on the natural ability of human high-density lipoprotein (HDL) to bind LPS, we synthesized a suite of HDL-like nanoparticles (HDL-like NP). We identified one HDL-like NP that was particularly effective at decreasing TLR4 signaling caused by addition of purified LPS or Gram-negative bacteria to model human cell lines or primary human peripheral blood cells. The HDL-like NP functioned to inhibit TLR4-dependent inflammatory response to LPS derived from multiple bacterial species. Mechanistically, data show that the NP mainly functions by scavenging and neutralizing the LPS toxin. Taken together, HDL-like NPs constitute a powerful endotoxin scavenger with the potential to significantly reduce LPS-mediated inflammation.
Collapse
Affiliation(s)
- Linda Foit
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E. Superior St, Chicago, IL 60611, USA
| | - C Shad Thaxton
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E. Superior St, Chicago, IL 60611, USA; International Institute for Nanotechnology (IIN), 2145 Sheridan Road, Evanston, IL 60208, USA; Robert H Lurie Comprehensive Cancer Center (RHLCCC), Northwestern University, Feinberg School of Medicine, 303 E Superior, Chicago, IL 60611, USA.
| |
Collapse
|
49
|
Eckel EF, Ametaj BN. Invited review: Role of bacterial endotoxins in the etiopathogenesis of periparturient diseases of transition dairy cows. J Dairy Sci 2016; 99:5967-5990. [PMID: 27209132 DOI: 10.3168/jds.2015-10727] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 04/02/2016] [Indexed: 12/19/2022]
Abstract
The dairy industry continues to suffer severe economic losses due to the increased disease incidence cows experience during the transition period. It has long been the classical view that the major contributing factor to the development of these periparturient diseases is the considerable increase in nutritional demands for milk production. This classical view, however, fails to account for the substantial correlation between both metabolic and infectious diseases and the detrimental effects that can occur with the provision of high-energy diets to support these nutritional demands. Currently, increasing evidence implicates bacterial endotoxins in the etiopathology of most periparturient diseases. Bacterial endotoxins are components of the outer cell wall of gram-negative and gram-positive bacteria that are highly immunostimulatory and can trigger proinflammatory immune responses. The ability of endotoxins to translocate from the mucosal tissues, including the gastrointestinal tract, mammary gland, and uterus, into the systemic circulation has been observed. Once they have entered the circulation, endotoxins potentially contribute to disease either directly, through eliciting an inflammatory response, or indirectly through other factors such as the overreaction of the natural protective mechanisms of the host. Although the evidence implicating a role of endotoxins in the pathogenesis of transition diseases continues to grow, our current knowledge of the host response to mucosal endotoxin exposure and pathogenic mechanisms remain largely unknown. Developing our understanding of the connection between endotoxemia and dairy cattle disease holds significant potential for the future development of preventative measures that could benefit the productivity of the dairy industry as well as animal welfare.
Collapse
Affiliation(s)
- Emily F Eckel
- Department of Agriculture, Food and Nutritional Science, University of Alberta Edmonton, AB T6G 2P5, Canada
| | - Burim N Ametaj
- Department of Agriculture, Food and Nutritional Science, University of Alberta Edmonton, AB T6G 2P5, Canada.
| |
Collapse
|
50
|
Hersoug LG, Møller P, Loft S. Gut microbiota-derived lipopolysaccharide uptake and trafficking to adipose tissue: implications for inflammation and obesity. Obes Rev 2016; 17:297-312. [PMID: 26712364 DOI: 10.1111/obr.12370] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 12/12/2022]
Abstract
The composition of the gut microbiota and excessive ingestion of high-fat diets (HFD) are considered to be important factors for development of obesity. In this review we describe a coherent mechanism of action for the development of obesity, which involves the composition of gut microbiota, HFD, low-grade inflammation, expression of fat translocase and scavenger receptor CD36, and the scavenger receptor class B type 1 (SR-BI). SR-BI binds to both lipids and lipopolysaccharide (LPS) from Gram-negative bacteria, which may promote incorporation of LPS in chylomicrons (CMs). These CMs are transported via lymph to the circulation, where LPS is transferred to other lipoproteins by translocases, preferentially to HDL. LPS increases the SR-BI binding, transcytosis of lipoproteins over the endothelial barrier,and endocytosis in adipocytes. Especially large size adipocytes with high metabolic activity absorb LPS-rich lipoproteins. In addition, macrophages in adipose tissue internalize LPS-lipoproteins. This may contribute to the polarization from M2 to M1 phenotype, which is a consequence of increased LPS delivery into the tissue during hypertrophy. In conclusion, evidence suggests that LPS is involved in the development of obesity as a direct targeting molecule for lipid delivery and storage in adipose tissue.
Collapse
Affiliation(s)
- L-G Hersoug
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - P Møller
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - S Loft
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|