Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
J Fish Dis ; 44(9): 1385-1397, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33999444

ABSTRACT

The definition of scavenger endothelial cells (SEC) is exclusively based on functional and structural characteristics. The following characteristics are common hallmarks for the vertebrate SEC: (a) All vertebrates examined are furnished with a population of special SEC that plays a role in the catabolism of physiologic and non-physiologic soluble waste macromolecules. (b) From the ligands that are endocytosed, SEC in all seven vertebrate classes appear to express the collagen α-chain receptor and the scavenger receptors. In addition, the hyaluronan and the mannose receptors are present on SEC of mammalia (several species) and osteichthyes (e.g., salmon and cod). It is likely that all four receptor types are present in all vertebrate classes. (c) Like liver endothelial cells (LEC) in mammals, SEC in all vertebrate classes are geared to endocytosis of soluble macromolecules, but phagocytic uptake of particles is taken care of mainly by macrophages. (d) The most primitive vertebrates (hagfish, lamprey and ray) carry their SEC in gill vessels, whereas phylogenetically younger fishes (salmon, carp, cod and plaice) carry their SEC in either kidney or heart and in all terrestrial vertebrates-SEC are found exclusively in the liver. (e) SEC of all vertebrates are localized in blood sinusoids or trabeculae that carry large amounts of slowly flowing and O2 poor blood. (f) SEC differs functionally and structurally from what is normally associated with "conventional vascular endothelium."


Subject(s)
Endothelium, Vascular/physiology , Fishes/physiology , Receptors, Scavenger/physiology , Animals , Endocytosis/physiology , Endothelial Cells/physiology
2.
Biochem Biophys Res Commun ; 533(3): 493-500, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32977946

ABSTRACT

Silica crystals (silica), which are a major mineral component of volcanic ash and desert dust, contribute to the pathogenesis of pulmonary disorders such as asthma and fibrosis. Although administration of silica or sand dust to rodents exacerbates development of ovalbumin-induced or house dust mite-induced asthma-like airway inflammation, the detailed mechanisms remain unclear. Here, using murine models, we found that silica can induce IL-33 expression in pulmonary epithelial cells. IL-33, but not IL-25 or TSLP, and type 2 cytokines such as IL-5 and IL-13 were critically involved in silica's exacerbation of OVA-induced airway eosinophilia in mice. Innate lymphoid cells (ILCs), but not T, B or NKT cells, were also involved in the setting. Moreover, a scavenger receptor that recognized silica was important for silica's exacerbating effect. These observations suggest that IL-33 induced in epithelial cells by silica activates ILCs to produce IL-5 and/or IL-13, contributing to silica's exacerbation of OVA-induced airway eosinophilia in mice. Our findings provide new insight into the underlying mechanisms of exacerbation of pulmonary disorders such as asthma following inhalation of silica-containing materials such as volcanic ash and desert dust.


Subject(s)
Interleukin-33/physiology , Pulmonary Eosinophilia/immunology , Silicon Dioxide/toxicity , Animals , Asthma/immunology , Cytokines/physiology , Interleukin-13/physiology , Interleukin-33/biosynthesis , Interleukin-5/physiology , Interleukins/physiology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Eosinophilia/chemically induced , Receptors, Scavenger/physiology , Thymic Stromal Lymphopoietin
3.
Curr Biol ; 30(14): R790-R795, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32693066

ABSTRACT

Scavenger receptors (SRs) are a large family of cell-surface receptors that are diverse in their structure and biological function and are divided into different classes. SRs can bind to a range of ligands and enhance the elimination of altered-self or non-self targets. The functional mechanisms that lead to their clearance of harmful substances involve phagocytosis, endocytosis, adhesion, and signaling.


Subject(s)
Receptors, Scavenger/metabolism , Receptors, Scavenger/physiology , Alarmins , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/therapy , Endocytosis , Humans , Immunity, Innate , Inflammation , Ligands , Macrophages/metabolism , Molecular Targeted Therapy , Neurodegenerative Diseases/etiology , Pathogen-Associated Molecular Pattern Molecules , Phagocytosis , Receptors, Scavenger/chemistry , Receptors, Scavenger/classification , Signal Transduction , Toll-Like Receptors
4.
J Am Soc Nephrol ; 31(9): 2013-2024, 2020 09.
Article in English | MEDLINE | ID: mdl-32611589

ABSTRACT

BACKGROUND: IgA nephropathy (IgAN) begins with aberrant IgA deposition in glomeruli, progresses to IgM/IgG/complement codeposition, and results in chronic inflammation and glomerular damage. However, the mechanism that drives such phlogogenic cascade has been unclear. Recently, apoptosis inhibitor of macrophage (AIM) protein was shown to modulate macrophages' function in various pathologic conditions, thereby profoundly affecting the progression of renal disorders, including AKI. A spontaneous IgAN model, grouped ddY (gddY) mouse, revealed the requirement of AIM for the overall inflammatory glomerular injury following IgA deposition. METHODS: We established an AIM-deficient IgAN model (AIM-/- gddY) using CRISPR/Cas9 and compared its phenotype with that of wild-type gddY with or without recombinant AIM administration. An IgA-deficient IgAN model (IgA-/- gddY) was also generated to further determine the role of AIM. RESULTS: In both human and murine IgAN, AIM colocalized with IgA/IgM/IgG in glomeruli, whereas control kidneys did not exhibit AIM deposition. Although AIM-/- gddY showed IgA deposition at levels comparable with those of wild-type gddY, they did not exhibit glomerular accumulation of IgM/IgG complements, CD45+ leukocyte infiltration, and upregulation of inflammatory/fibrogenic genes, indicating protection from glomerular lesions and proteinuria/hematuria. Recombinant AIM administration reconstituted the IgAN phenotype, resulting in IgM/IgG/complement IgA codeposition. Neither spontaneous IgM/IgG codeposition nor disease was observed in IgA-/- gddY mice. CONCLUSIONS: AIM may contribute to stable immune complex formation in glomeruli, thereby facilitating IgAN progression. Therefore, AIM deposition blockage or disassociation from IgM/IgG may present a new therapeutic target on the basis of its role in IgAN inflammation initiation.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Glomerulonephritis, IGA/complications , Inflammation/etiology , Kidney Glomerulus/pathology , Receptors, Scavenger/physiology , Animals , Complement Activation , Glomerulonephritis, IGA/pathology , Humans , Immunoglobulin A/metabolism , Kidney Glomerulus/immunology , Mice , Mice, Inbred BALB C
5.
Front Immunol ; 11: 268, 2020.
Article in English | MEDLINE | ID: mdl-32194551

ABSTRACT

Phagocytosis is an ancient, highly conserved process in all multicellular organisms, through which the host can protect itself against invading microorganisms and environmental particles, as well as remove self-apoptotic cells/cell debris to maintain tissue homeostasis. In crustacean, phagocytosis by hemocyte has also been well-recognized as a crucial defense mechanism for the host against infectious agents such as bacteria and viruses. In this review, we summarized the current knowledge of hemocyte-mediated phagocytosis, in particular focusing on the related receptors for recognition and internalization of pathogens as well as the downstream signal pathways and intracellular regulators involved in the process of hemocyte phagocytosis. We attempted to gain a deeper understanding of the phagocytic mechanism of different hemocytes and their contribution to the host defense immunity in crustaceans.


Subject(s)
Crustacea/immunology , Hemocytes/physiology , Phagocytes/physiology , Phagocytosis , Animals , Cell Adhesion Molecules/physiology , Crustacea/cytology , Host-Pathogen Interactions/immunology , Immunity, Innate , Lectins/physiology , Opsonin Proteins/physiology , Phagosomes/physiology , Pore Forming Cytotoxic Proteins , Receptors, Pattern Recognition/physiology , Receptors, Scavenger/physiology , Signal Transduction/physiology
6.
Sci Rep ; 10(1): 159, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31932599

ABSTRACT

Enterovirus-A71 (EV-A71) is a common cause of hand-foot-and-mouth disease (HFMD) and, rarely, causes severe neurological disease. This study aimed to elucidate the epidemiological and genetic characteristics and virulence of EV-A71 strains isolated from children diagnosed with HFMD. Rectal and throat swabs were collected from 488 children with HFMD in Hanoi, Vietnam, in 2015-2016. From 391 EV-positive patients, 15 EVs, including coxsackievirus A6 (CV-A6; 47.1%) and EV-A71 (32.5%, n = 127), were identified. Of the 127 EV-A71 strains, 117 (92.1%) were the B5 subgenotype and 10 (7.9%) were the C4 subgenotype. A whole-genome analysis of EV-A71 strains showed that seven of the eight C4a strains isolated in 2016 formed a new lineage, including two possible recombinants between EV-A71 C4 and CV-A8. The proportion of inpatients among C4-infected children was higher than among B5-infected children (80.0% vs. 27.4%; P = 0.002). The virulence of EV-A71 strains was examined in human scavenger receptor class B2 (hSCARB2)-transgenic mice, and EV-A71 C4 strains exhibited higher mortality than B5 strains (80.0% vs. 30.0%, P = 0.0001). Thus, a new EV-A71 C4a-lineage, including two possible recombinants between EV-A71 C4 and CV-A8, appeared in 2016 in Vietnam. The EV-A71 C4 subgenotype may be more virulent than the B5 subgenotype.


Subject(s)
Enterovirus/classification , Enterovirus/isolation & purification , Hand, Foot and Mouth Disease/epidemiology , Hand, Foot and Mouth Disease/mortality , Lysosomal Membrane Proteins/physiology , Receptors, Scavenger/physiology , Virus Replication , Animals , Child , Child, Preschool , Disease Outbreaks , Enterovirus/genetics , Female , Hand, Foot and Mouth Disease/virology , Humans , Infant , Male , Mice , Mice, Transgenic , Phylogeny , Serogroup , Survival Rate , Time Factors , Vietnam/epidemiology
7.
Front Immunol ; 11: 620339, 2020.
Article in English | MEDLINE | ID: mdl-33542723

ABSTRACT

Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.


Subject(s)
Host-Pathogen Interactions/immunology , Macrophages/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/physiology , Bacterial Vaccines , Cations/metabolism , Cell Death , Chemotaxis , Cytokines/metabolism , Extracellular Vesicles/metabolism , Humans , Hydrogen-Ion Concentration , Immune Evasion/immunology , Macrophages/classification , Macrophages/enzymology , Macrophages/microbiology , Mice , Nutrients/metabolism , Phagocytosis , Phagosomes/chemistry , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Receptors, Complement/physiology , Receptors, Fc/immunology , Receptors, Scavenger/physiology , Staphylococcus aureus/drug effects
8.
Front Immunol ; 10: 458, 2019.
Article in English | MEDLINE | ID: mdl-30936871

ABSTRACT

Non-typeable Haemophilus influenzae (NTHi) causes persistent respiratory infections in patients with chronic obstructive pulmonary disease (COPD), probably linked to its capacity to invade and reside within pneumocytes. In the alveolar fluid, NTHi is in contact with pulmonary surfactant, a lipoprotein complex that protects the lung against alveolar collapse and constitutes the front line of defense against inhaled pathogens and toxins. Decreased levels of surfactant phospholipids have been reported in smokers and patients with COPD. The objective of this study was to investigate the effect of surfactant phospholipids on the host-pathogen interaction between NTHi and pneumocytes. For this purpose, we used two types of surfactant lipid vesicles present in the alveolar fluid: (i) multilamellar vesicles (MLVs, > 1 µm diameter), which constitute the tensioactive material of surfactant, and (ii) small unilamellar vesicles (SUVs, 0.1 µm diameter), which are generated after inspiration/expiration cycles, and are endocytosed by pneumocytes for their degradation and/or recycling. Results indicated that extracellular pulmonary surfactant binds to NTHi, preventing NTHi self-aggregation and inhibiting adhesion of NTHi to pneumocytes and, consequently, inhibiting NTHi invasion. In contrast, endocytosed surfactant lipids, mainly via the scavenger receptor SR-BI, did not affect NTHi adhesion but inhibited NTHi invasion by blocking bacterial uptake in pneumocytes. This blockade was made possible by inhibiting Akt phosphorylation and Rac1 GTPase activation, which are signaling pathways involved in NTHi internalization. Administration of the hydrophobic fraction of lung surfactant in vivo accelerated bacterial clearance in a mouse model of NTHi pulmonary infection, supporting the notion that the lipid component of lung surfactant protects against NTHi infection. These results suggest that alterations in surfactant lipid levels in COPD patients may increase susceptibility to infection by this pathogen.


Subject(s)
Alveolar Epithelial Cells/drug effects , Haemophilus Infections/prevention & control , Haemophilus influenzae/drug effects , Pulmonary Surfactants/pharmacology , Alveolar Epithelial Cells/metabolism , Animals , Bacterial Adhesion/drug effects , Endocytosis/drug effects , Enzyme Activation/drug effects , Extracellular Vesicles/physiology , Haemophilus Infections/immunology , Haemophilus influenzae/isolation & purification , Haemophilus influenzae/physiology , Host-Pathogen Interactions/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Liposomes , Male , Mice , Neuropeptides/antagonists & inhibitors , Otitis Media/microbiology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Surfactants/immunology , Rats , Rats, Sprague-Dawley , Receptors, Scavenger/antagonists & inhibitors , Receptors, Scavenger/physiology , Specific Pathogen-Free Organisms , rac1 GTP-Binding Protein/antagonists & inhibitors
9.
J Virol ; 93(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30894476

ABSTRACT

Enterovirus 71 (EV71) infection is generally associated with hand-foot-and-mouth disease (HFMD) and may cause severe neurological disorders and even death. An effective murine oral infection model for studying the pathogenesis of various clinical EV71 isolates is lacking. We developed a transgenic (Tg) mouse that expresses an EV71 receptor, that is, human scavenger receptor class B member 2 (hSCARB2), in a pattern highly similar to that of endogenous murine SCARB2 (mSCARB2) protein. A FLAG-tagged SCARB2 cDNA fragment composed of exons 3 to 12 was inserted into a murine Scarb2 gene-containing bacterial artificial chromosome (BAC) clone, and the resulting transgene was used for establishment of chimeric receptor-expressing Tg mice. Tg mice intragastrically (i.g.) infected with clinical isolates of EV71 showed neurological symptoms, such as ataxia and paralysis, and fatality. There was an age-dependent decrease in susceptibility to viral infection. Pathological characteristics of the infected Tg mice resembled those of encephalomyelitis in human patients. Viral infection was accompanied by microglial activation. Clodronate treatment of the brain slices from Tg mice enhanced viral replication, while lipopolysaccharide treatment significantly inhibited it, suggesting an antiviral role for microglia during EV71 infection. Taken together, this Tg mouse provides a model that closely mimics natural infection for studying EV71 pathogenesis and for evaluating the efficacy of vaccines or other antiviral drugs.IMPORTANCE The availability of a murine model of EV71 infection is beneficial for the understanding of pathogenic mechanisms and the development and assessment of vaccines and antiviral drugs. However, the lack of a murine oral infection model thwarted the study of pathogenesis induced by clinically relevant EV71 strains that are transmitted via the oral-oral or oral-fecal route. Our Tg mice could be intragastrically infected with clinically relevant EV71 strains in an efficient way and developed neurological symptoms and pathological changes strikingly resembling those of human infection. Moreover, these mice showed an age-dependent change in susceptibility that is similar to the human case. This Tg mouse, when combined with the use of other genetically modified mice, potentially contributes to studying the relationship between developmental changes in immunity and susceptibility to virus.


Subject(s)
CD36 Antigens/metabolism , Enterovirus Infections/genetics , Lysosomal Membrane Proteins/metabolism , Receptors, Scavenger/metabolism , Animals , CD36 Antigens/physiology , Cell Line , Cells, Cultured , Disease Models, Animal , Enterovirus/genetics , Enterovirus/metabolism , Enterovirus A, Human/genetics , Enterovirus Infections/immunology , Enterovirus Infections/virology , Humans , Lysosomal Membrane Proteins/physiology , Mice , Mice, Transgenic , Receptors, Scavenger/genetics , Receptors, Scavenger/physiology , Receptors, Virus/metabolism , Virus Replication
10.
Br J Haematol ; 183(2): 185-195, 2018 10.
Article in English | MEDLINE | ID: mdl-30378120

ABSTRACT

The mechanisms involved in regulating von Willebrand factor (VWF) clearance remain poorly understood. However recent studies have shown that macrophages play a critical role in regulating the half-life of VWF, and have identified specific lectin (including asialoglycoprotein, macrophage galactose-type lectin, Sigec-5 and C-type lectin domain family 4 member M) and scavenger receptors (including low-density lipoprotein receptor-related protein-1, scavenger receptor A1 and stabilin-2) that are involved in VWF clearance. Further studies will be required to determine the relative importance of these individual receptors with respect to physiological and pathological VWF clearance. Nevertheless, recent clinical data have highlighted the importance of enhanced VWF clearance in the pathogenesis of type 1 von Willebrand disease (VWD). Moreover, increased clearance also contributes to reduced VWF levels in many patients with type 2 and type 3 VWD. Improved understanding regarding VWF clearance is not only of direct biological relevance, but may also have important implications for the development of novel therapeutic agents with extended plasma half-lives for the treatment of both VWD and haemophilia A.


Subject(s)
von Willebrand Diseases/blood , von Willebrand Factor/metabolism , Deamino Arginine Vasopressin/therapeutic use , Hemostatics/therapeutic use , Humans , Mutation , Receptors, Mitogen/physiology , Receptors, Scavenger/physiology , Structure-Activity Relationship , von Willebrand Diseases/drug therapy , von Willebrand Diseases/etiology , von Willebrand Factor/genetics
11.
Haematologica ; 103(4): 728-737, 2018 04.
Article in English | MEDLINE | ID: mdl-29326120

ABSTRACT

Previously, we found that LDL-receptor related protein-1 on macrophages mediated shear stress-dependent clearance of von Willebrand factor. In control experiments, however, we observed that von Willebrand factor also binds to macrophages independently of this receptor under static conditions, suggesting the existence of additional clearance-receptors. In search for such receptors, we focused on the macrophage-specific scavenger-receptor SR-AI. von Willebrand factor displays efficient binding to SR-AI (half-maximum binding 14±5 nM). Binding is calcium-dependent and is inhibited by 72±4% in the combined presence of antibodies against the A1- and D4-domains. Association with SR-AI was confirmed in cell-binding experiments. In addition, binding to bone marrow-derived murine SR-AI-deficient macrophages was strongly reduced compared to binding to wild-type murine macrophages. Following expression via hydrodynamic gene transfer, we determined ratios for von Willebrand factor-propeptide over von Willebrand factor-antigen, a marker of von Willebrand factor clearance. Propeptide/antigen ratios were significantly reduced in SR-AI-deficient mice compared to wild-type mice (0.6±0.2 versus 1.3±0.3; P<0.0001), compatible with a slower clearance of von Willebrand factor in SR-AI-deficient mice. Interestingly, mutants associated with increased clearance (von Willebrand factor/p.R1205H and von Willebrand factor/p.S2179F) had significantly increased binding to purified SR-AI and SR-AI expressed on macrophages. Accordingly, propeptide/antigen ratios for these mutants were reduced in SR-AI-deficient mice. In conclusion, we have identified SR-AI as a novel macrophage-specific receptor for von Willebrand factor. Enhanced binding of von Willebrand factor mutants to SR-AI may contribute to the increased clearance of these mutants.


Subject(s)
Scavenger Receptors, Class A/physiology , von Willebrand Factor/metabolism , Animals , Cell Line , Cell Line, Tumor , Humans , Macrophages , Mice , Mutant Proteins/metabolism , Protein Binding , Receptors, Scavenger/physiology , von Willebrand Factor/genetics
12.
Am J Chin Med ; 46(1): 87-106, 2018.
Article in English | MEDLINE | ID: mdl-29298513

ABSTRACT

oxLDL is involved in the pathogenesis of atherosclerotic lesions through cholesterol accumulation in macrophage foam cells. Andrographolide, the bioactive component of Andrographis paniculata, possesses several biological activities such as anti-inflammatory, anti-oxidant, and anticancer functions. Scavenger receptors (SRs), including class A SR (SR-A) and CD36, are responsible for the internalization of oxLDL. In contrast, receptors for reverse cholesterol transport, including ABCA1 and ABCG1, mediate the efflux of cholesterol from macrophage foam cells. Transcription factor liver X receptor [Formula: see text] (LXR[Formula: see text] plays a key role in lipid metabolism and inflammation as well as in the regulation of ABCA1 and ABCG1 expression. Because of the contribution of inflammation to macrophage foam cell formation and the potent anti-inflammatory activity of andrographolide, we hypothesized that andrographolide might inhibit oxLDL-induced macrophage foam cell formation. The results showed that andrographolide reduced oxLDL-induced lipid accumulation in macrophage foam cells. Andrographolide decreased the mRNA and protein expression of CD36 by inducing the degradation of CD36 mRNA; however, andrographolide had no effect on SR-A expression. In contrast, andrographolide increased the mRNA and protein expression of ABCA1 and ABCG1, which were dependent on LXR[Formula: see text]. Andrographolide enhanced LXR[Formula: see text] nuclear translocation and DNA binding activity. Treatment with the LXR[Formula: see text] antagonist GGPP and transfection with LXR[Formula: see text] siRNA reversed the ability of andrographolide to stimulate ABCA1 and ABCG1 protein expression. In conclusion, inhibition of CD36-mediated oxLDL uptake and induction of ABCA1- and ABCG1-dependent cholesterol efflux are two working mechanisms by which andrographolide inhibits macrophage foam cell formation, which suggests that andrographolide could be a potential candidate to prevent atherosclerosis.


Subject(s)
Andrographis/chemistry , Cholesterol/metabolism , Diterpenes/pharmacology , Foam Cells/metabolism , Lipoproteins, LDL/adverse effects , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Animals , Anti-Inflammatory Agents , Antineoplastic Agents, Phytogenic , Antioxidants , Atherosclerosis/etiology , Biological Transport/genetics , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Line , Gene Expression/drug effects , Liver X Receptors/physiology , Mice , RNA, Messenger/metabolism , Receptors, Scavenger/physiology
13.
Uirusu ; 68(1): 71-78, 2018.
Article in Japanese | MEDLINE | ID: mdl-31105137

ABSTRACT

Since RNA virus genome encodes only a limited number of viral proteins, replication of RNA virus mostly relies on host cells. Elucidation of host proteins that play important roles in the virus replication cycles contributes not only to fundamental virology research but also to applied research such as development of antiviral drugs. We revealed that Ebola virus matrix protein VP40 utilized host COPII transport machinery for its intracellular transport to the plasma membrane. Second, we demonstrated that enterovirus A71 used Scavenger receptor class B member 2 (SCARB2) as a cellular receptor. Finally, we found that host protein CLUH played an important role in the subnuclear transport of influenza virus ribonucleoprotein (vRNP) complexes. Here, I would like to briefly introduce these findings.


Subject(s)
COP-Coated Vesicles/physiology , Host-Pathogen Interactions/genetics , Lysosomal Membrane Proteins/physiology , RNA Viruses/physiology , RNA-Binding Proteins/physiology , Receptors, Scavenger/physiology , Virus Replication/genetics , Active Transport, Cell Nucleus , Animals , Humans , Mice , RNA Viruses/genetics , Viral Matrix Proteins/metabolism
14.
J Immunol ; 198(10): 3775-3789, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28483986

ABSTRACT

Scavenger receptors constitute a large family of proteins that are structurally diverse and participate in a wide range of biological functions. These receptors are expressed predominantly by myeloid cells and recognize a diverse variety of ligands including endogenous and modified host-derived molecules and microbial pathogens. There are currently eight classes of scavenger receptors, many of which have multiple names, leading to inconsistencies and confusion in the literature. To address this problem, a workshop was organized by the United States National Institute of Allergy and Infectious Diseases, National Institutes of Health, to help develop a clear definition of scavenger receptors and a standardized nomenclature based on that definition. Fifteen experts in the scavenger receptor field attended the workshop and, after extensive discussion, reached a consensus regarding the definition of scavenger receptors and a proposed scavenger receptor nomenclature. Scavenger receptors were defined as cell surface receptors that typically bind multiple ligands and promote the removal of nonself or altered-self targets. They often function by mechanisms that include endocytosis, phagocytosis, adhesion, and signaling that ultimately lead to the elimination of degraded or harmful substances. Based on this definition, nomenclature and classification of these receptors into 10 classes were proposed. This classification was discussed at three national meetings and input from participants at these meetings was requested. The following manuscript is a consensus statement that combines the recommendations of the initial workshop and incorporates the input received from the participants at the three national meetings.


Subject(s)
Receptors, Scavenger/classification , Receptors, Scavenger/physiology , Animals , Endocytosis , Humans , Ligands , Mice , National Institute of Allergy and Infectious Diseases (U.S.)/standards , Phagocytosis , Receptors, Immunologic/physiology , Scavenger Receptors, Class A/physiology , Signal Transduction , Terminology as Topic , United States
15.
Lipids Health Dis ; 16(1): 30, 2017 Feb 06.
Article in English | MEDLINE | ID: mdl-28166809

ABSTRACT

Atherosclerosis is considered as an inflammatory and chronic disorder with an important immunologic component, which underlies the majority of cardiovascular diseases; condition that belongs to a group of noncommunicable diseases that to date and despite of prevention and treatment approaches, they remain as the main cause of death worldwide, with 17.5 million of deaths every year. The impact of lipids in human health and disease is taking center stage in research, due to lipotoxicity explained by elevated concentration of circulating lipids, in addition to altered adipose tissue metabolism, and aberrant intracellular signaling. Immune response and metabolic regulation are highly integrated systems and the proper function of each one is dependent on the other. B lymphocytes express a variety of receptors that can recognize foreign, endogenous or modified self-antigens, among them oxidized low density lipoproteins, which are the main antigens in atherosclerosis. Mechanisms of B cells to recognize, remove and present lipids are not completely clear. However, it has been reported that B cell can recognize/remove lipids through a range of receptors, such as LDLR, CD1d, FcR and SR, which might have an atheroprotector or proatherogenic role during the course of atherosclerotic disease. Pertinent literature related to these receptors was examined to inform the present conclusions.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , Animals , Humans , Immunity, Cellular , Lipoproteins, LDL/immunology , Receptors, Scavenger/physiology
16.
Biochim Biophys Acta ; 1860(6): 1118-28, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26922829

ABSTRACT

BACKGROUND: C-reactive protein (CRP) is a plasma pentraxin family protein that is massively induced as part of the innate immune response to infection and tissue injury. CRP and other pentraxin proteins can activate a complement pathway through C1q, collectins, or on microbe surfaces. It has been found that a lectin-like oxidized LDL receptor 1 (LOX-1), which is an endothelial scavenger receptor (SR) having a C-type lectin-like domain, interacts with CRP to activate the complement pathway using C1q. However it remains elusive whether other lectins or SRs are involved in CRP-mediated complement activation and the downstream effect of the complement activation is also unknown. METHODS: We prepared CHO/ldlA7 cells expressing collectin placenta-1 (CL-P1) and studied the interaction of CRP with cells. We further used ELISA for testing binding between proteins. We tested for C3 fragment deposition and terminal complement complex (TCC) formation on HEK293 cells expressing CL-P1. RESULTS: Here, we demonstrated that CL-P1 bound CRP in a charge dependent manner and the interaction of CRP with CL-P1 mediated a classical complement activation pathway through C1q and additionally drove an amplification pathway using properdin. However, CRP also recruits complement factor H (CFH) on CL-P1 expressing cell surfaces, to inhibit the formation of a terminal complement complex in normal complement serum conditions. GENERAL SIGNIFICANCE: The interaction of collectin CL-P1 with CFH might be key for preventing attack on "self" as a result of complement activation induced by the CL-P1 and CRP interaction.


Subject(s)
C-Reactive Protein/chemistry , Collectins/chemistry , Complement Activation , Receptors, Scavenger/chemistry , Animals , C-Reactive Protein/physiology , CHO Cells , Collectins/physiology , Complement Factor H/chemistry , Cricetulus , HEK293 Cells , Humans , Receptors, Scavenger/physiology
17.
Cardiovasc Res ; 109(1): 24-33, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26472132

ABSTRACT

Scavenger receptors (SRs), which recognize modified low-density lipoprotein (LDL) by oxidation or acetylation, are a group of receptors on plasma membrane of macrophages and other cell types. These receptors by facilitating modified LDL uptake are a primary step in the intracellular accumulation of modified LDL and formation of fatty streak. Non-coding RNAs (ncRNAs) are a group of functional RNA nucleotides that are not translated into protein, and include microRNAs (miRs), snoRNAs, siRNAs, snRNAs, exRNAs, piRNAs, and the long ncRNAs (lncRNAs). Recently, ncRNAs have received much attention due to their effects in a variety of disease states such as atherosclerotic cardiovascular disease and cancers. A host of ncRNAs, such as miRs and lncRNAs, have been found to be involved in the regulation of SRs and the inflammatory cascade and subsequently atherosclerosis. Here, we review this important area to create interest in this growing field among researchers and clinicians alike.


Subject(s)
Atherosclerosis/etiology , RNA, Untranslated/physiology , Receptors, Scavenger/physiology , Animals , Humans , Receptors, Scavenger/classification
18.
Cancer Sci ; 107(3): 290-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26708384

ABSTRACT

Wnt5a-Ror2 signaling has been shown to play important roles in promoting aggressiveness of various cancer cells in a cell-autonomous manner. However, little is known about its function in cancer-associated stromal cells, including mesenchymal stem cells (MSCs). Thus, we examined the role of Wnt5a-Ror2 signaling in bone marrow-derived MSCs in regulating proliferation of undifferentiated gastric cancer cells. Coculture of a gastric cancer cell line, MKN45, with MSCs either directly or indirectly promotes proliferation of MKN45 cells, and suppressed expression of Ror2 in MSCs prior to coculture inhibits enhanced proliferation of MKN45 cells. In addition, conditioned media from MSCs, treated with control siRNA, but not siRNAs against Ror2, can enhance proliferation of MKN45 cells. Interestingly, it was found that expression of CXCL16 in MSCs is augmented by Wnt5a-Ror2 signaling, and that recombinant chemokine (C-X-C motif) ligand (CXCL)16 protein can enhance proliferation of MKN45 cells in the absence of MSCs. In fact, suppressed expression of CXCL16 in MSCs or an addition of a neutralizing antibody against CXCL16 fails to promote proliferation of MKN45 cells in either direct or indirect coculture with MSCs. Importantly, we show that MKN45 cells express chemokine (C-X-C motif) receptor (CXCR)6, a receptor for CXCL16, and that suppressed expression of CXCR6 in MKN45 cells results in a failure of its enhanced proliferation in either direct or indirect coculture with MSCs. These findings indicate that Wnt5a-Ror2 signaling enhances expression of CXCL16 in MSCs and, as a result, enhanced secretion of CXCL16 from MSCs might act on CXCR6 expressed on MKN45, leading to the promotion of its proliferation.


Subject(s)
Chemokines, CXC/physiology , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptors, Chemokine/metabolism , Receptors, Scavenger/physiology , Receptors, Virus/metabolism , Stomach Neoplasms/metabolism , Wnt Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL16 , Coculture Techniques , Gene Expression Regulation, Neoplastic , Humans , Receptors, CXCR6 , Signal Transduction , Stomach Neoplasms/pathology , Transcriptional Activation , Wnt-5a Protein
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(5): 723-7, 2015 Oct.
Article in Chinese | MEDLINE | ID: mdl-26419000

ABSTRACT

SCARB2 (scavenger receptor class B, member 2) is a lysosomal membrane glucoprotein, which is encoded by SCARB2 gene. It takes vital parts in the physiological and pathological processes including the transportation of beta-glucocerebrosidase to the lysosome, infection of EV71 and load-induced cardiac myocyte hypertrophy. This article has reviewed the molecular structure and functions of SCARB2 gene and its protein, as well as their relationship with diseases.


Subject(s)
Lysosomal Membrane Proteins/physiology , Receptors, Scavenger/physiology , Hand, Foot and Mouth Disease/genetics , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Myoclonic Epilepsies, Progressive/genetics , Parkinson Disease/genetics , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics
20.
PLoS One ; 9(4): e94197, 2014.
Article in English | MEDLINE | ID: mdl-24718459

ABSTRACT

Beta-amyloid (Aß) aggregates have a pivotal role in pathological processing of Alzheimer's disease (AD). The clearance of Aß monomer or aggregates is a causal strategy for AD treatment. Microglia and astrocytes are the main macrophages that exert critical neuroprotective roles in the brain. They may effectively clear the toxic accumulation of Aß at the initial stage of AD, however, their functions are attenuated because of glial overactivation. In this study, we first showed that heptapeptide XD4 activates the class A scavenger receptor (SR-A) on the glia by increasing the binding of Aß to SR-A, thereby promoting glial phagocytosis of Aß oligomer in microglia and astrocytes and triggering intracellular mitogen-activated protein kinase (MAPK) signaling cascades. Moreover, XD4 enhances the internalization of Aß monomers to microglia and astrocytes through macropinocytosis or SR-A-mediated phagocytosis. Furthermore, XD4 significantly inhibits Aß oligomer-induced cytotoxicity to glial cells and decreases the production of proinflammatory cytokines, such as TNF-α and IL-1ß, in vitro and in vivo. Our findings may provide a novel strategy for AD treatment by activating SR-A.


Subject(s)
Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Microglia/metabolism , Peptide Fragments/metabolism , Peptides/pharmacology , Receptors, Scavenger/physiology , Scavenger Receptors, Class A/physiology , Animals , Astrocytes/drug effects , Astrocytoma/pathology , Cell Line , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/cytology , Drug Evaluation, Preclinical , Humans , Interleukin-1beta/metabolism , Membrane Proteins/metabolism , Mice , Microglia/drug effects , Phagocytosis/drug effects , Rats , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL