Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
1.
Cell ; 184(20): 5163-5178.e24, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34559985

ABSTRACT

Rift Valley fever virus (RVFV) is a zoonotic pathogen with pandemic potential. RVFV entry is mediated by the viral glycoprotein (Gn), but host entry factors remain poorly defined. Our genome-wide CRISPR screen identified low-density lipoprotein receptor-related protein 1 (mouse Lrp1/human LRP1), heat shock protein (Grp94), and receptor-associated protein (RAP) as critical host factors for RVFV infection. RVFV Gn directly binds to specific Lrp1 clusters and is glycosylation independent. Exogenous addition of murine RAP domain 3 (mRAPD3) and anti-Lrp1 antibodies neutralizes RVFV infection in taxonomically diverse cell lines. Mice treated with mRAPD3 and infected with pathogenic RVFV are protected from disease and death. A mutant mRAPD3 that binds Lrp1 weakly failed to protect from RVFV infection. Together, these data support Lrp1 as a host entry factor for RVFV infection and define a new target to limit RVFV infections.


Subject(s)
Host-Pathogen Interactions , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Rift Valley fever virus/physiology , Virus Internalization , Animals , Antibody Specificity/immunology , Base Sequence , Brain/pathology , Brain/virology , CRISPR-Cas Systems/genetics , Cell Membrane/metabolism , Cells, Cultured , Glycoproteins/metabolism , Glycosaminoglycans/metabolism , Glycosylation , Humans , LDL-Receptor Related Protein-Associated Protein/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/deficiency , Membrane Glycoproteins/metabolism , Mice , Protein Binding , Protein Denaturation , Rift Valley Fever/pathology , Rift Valley Fever/prevention & control , Rift Valley Fever/virology , Rift Valley fever virus/immunology
2.
Blood ; 137(23): 3251-3258, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33513604

ABSTRACT

Low-density lipoprotein (LDL) receptor-related protein-associated protein 1 (LRPAP1) had been identified by B-cell receptor (BCR) expression cloning and subsequent protein array screening as a frequent and proliferation-inducing autoantigen of mantle cell lymphoma (MCL). Of interest, high-titered and light chain-restricted LRPAP1 autoantibodies were detected in 8 of 28 patients with MCL. In the present study, LRPAP1 autoantibodies in sera of patients treated within the Younger and Elderly trials of the European MCL Network were analyzed regarding frequency, association with disease characteristics, and prognostic impact. LRPAP1 autoantibodies were detected in 41 (13%) of 312 evaluable patients with MCL. These LRPAP1 autoantibodies belonged predominantly to the immunoglobulin G (IgG) class and were clonally light chain restricted (27 with κ light chains, 14 patients with λ light chains). Titers ranged between 1:400 and 1:3200. The presence of LRPAP1 autoantibodies was not significantly associated with any baseline clinical characteristic, however, it was associated with a superior 5-year probability for failure-free survival (FFS) of 70% (95% confidence interval [CI], 57% to 87%) vs 51% (95% CI, 44% to 58%), P = .0052; and for overall survival (OS) of 93% (95% CI, 85% to 100%) vs 68% (95% CI, 62% to 74%), P = .0142. LRPAP1-seropositive patients had a Mantle Cell Lymphoma International Prognostic Index-adjusted hazard ratio for FFS of 0.48 (95% CI 0.27-0.83, P = .0083) and for OS of 0.47 (95% CI 0.24-0.94, P = .032). LRPAP1 autoantibodies were frequently detected in a large cohort of MCL patients treated within prospective multicenter clinical trials. Our results suggest better outcomes for LRPAP1-autoantibody seropositive patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Autoantibodies/immunology , Immunoglobulin G/immunology , LDL-Receptor Related Protein-Associated Protein/immunology , Lymphoma, Mantle-Cell , Neoplasm Proteins/immunology , Adult , Aged , Aged, 80 and over , Cyclophosphamide/administration & dosage , Disease-Free Survival , Doxorubicin/administration & dosage , Female , Humans , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Prednisone/administration & dosage , Rituximab/administration & dosage , Survival Rate , Vincristine/administration & dosage
3.
J Biol Chem ; 297(1): 100842, 2021 07.
Article in English | MEDLINE | ID: mdl-34058195

ABSTRACT

The low-density lipoprotein receptor (LDLR) family of receptors are cell-surface receptors that internalize numerous ligands and play crucial role in various processes, such as lipoprotein metabolism, hemostasis, fetal development, etc. Previously, receptor-associated protein (RAP) was described as a molecular chaperone for LDLR-related protein 1 (LRP1), a prominent member of the LDLR family. We aimed to verify this role of RAP for LRP1 and two other LDLR family receptors, LDLR and vLDLR, and to investigate the mechanisms of respective interactions using a cell culture model system, purified system, and in silico modelling. Upon coexpression of RAP with clusters of the ligand-binding complement repeats (CRs) of the receptors in secreted form in insect cells culture, the isolated proteins had increased yield, enhanced folding, and improved binding properties compared with proteins expressed without RAP, as determined by circular dichroism and surface plasmon resonance. Within LRP1 CR-clusters II and IV, we identified multiple sites comprised of adjacent CR doublets, which provide alternative bivalent binding combinations with specific pairs of lysines on RAP. Mutational analysis of these lysines within each of isolated RAP D1/D2 and D3 domains having high affinity to LRP1 and of conserved tryptophans on selected CR-doublets of LRP1, as well as in silico docking of a model LRP1 CR-triplet with RAP, indicated a universal role for these residues in interaction of RAP and LRP1. Consequently, we propose a new model of RAP interaction with LDLR family receptors based on switching of the bivalent contacts between molecules over time in a dynamic mode.


Subject(s)
LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Protein Folding , Receptors, LDL/metabolism , DNA Mutational Analysis , Humans , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Molecular Docking Simulation , Protein Binding , Repetitive Sequences, Amino Acid
4.
Biochem Biophys Res Commun ; 590: 14-19, 2022 01 29.
Article in English | MEDLINE | ID: mdl-34968779

ABSTRACT

BACKGROUND: Alzheimer's disease (AD), has caused a mass of disability and mortality in elder populations, which increases global health burden. There are still limited effective disease-modifying drugs. Alleviating microglia-evoked neuroinflammation has become a promising treatment strategy for AD. Ginsenoside Compound K has been demonstrated to exhibit anti-inflammatory and neuroprotective benefits. Here we measured the effects of Ginsenoside Compound K in inhibiting amyloid-induced microglia inflammation and the possible molecular mechanisms and target of action in vitro. METHODS: The cytotoxicity of all chemical reagents on BV2 cells were evaluated using the MTT assay. qRT-PCR and ELISA were carried out to detect the inflammatory cytokines levels. Western blot was utilized to determine the effect of Ginsenoside Compound K on the nuclear factor-κB (NF-κB) p65 nuclear translocation. Antagonist Receptor Associated Protein (RAP) was used to verify the engagement of low-density lipoprotein receptor-related protein 1(LRP1). RESULTS: Ginsenoside Compound K diminished inflammatory cytokine production and reversed NF-κB p65 nuclear translocation induced by Aß42 oligomers. LRP1 expression was up-regulated by Ginsenoside Compound K. When LRP1 was blocked by antagonist RAP, the protective effect of Ginsenoside Compound K was massively eliminated. CONCLUSION: These observations provide evidence for anti-inflammatory effect of Ginsenoside Compound K through NF-κB pathway via LRP1 activation, and support further evaluation of Ginsenoside Compound K as a potential effective modulator for AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/pathology , Ginsenosides/pharmacology , Inflammation/pathology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Microglia/pathology , NF-kappa B/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/biosynthesis , LDL-Receptor Related Protein-Associated Protein , Mice , Microglia/drug effects , Microglia/metabolism , Transcription Factor RelA/metabolism
5.
Cancer Immunol Immunother ; 71(2): 289-300, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34142235

ABSTRACT

Cancer cells frequently display defects in their antigen-processing pathway and thereby evade CD8 T cell immunity. We described a novel category of cancer antigens, named TEIPP, that emerge on cancers with functional loss of the peptide pump TAP. TEIPPs are non-mutated neoantigens despite their 'self' origin by virtue of their absence on normal tissues. Here, we describe the development of a synthetic long peptide (SLP) vaccine for the most immunogenic TEIPP antigen identified thus far, derived from the TAP-independent LRPAP1 signal sequence. LRPAP121-30-specific CD8 T cells were present in blood of all tested healthy donors as well as patients with non-small cell lung adenocarcinoma. SLPs with natural flanking, however, failed to be cross-presented by monocyte-derived dendritic cells. Since the C-terminus of LRPAP121-30 is an unconventional and weakly binding serine (S), we investigated if replacement of this anchor would result in efficient cross-presentation. Exchange into a valine (V) resulted in higher HLA-A2 binding affinity and enhanced T cell stimulation. Importantly, CD8 T cells isolated using the V-variant were able to bind tetramers with the natural S-variant and respond to TAP-deficient cancer cells. A functional screen with an array of N-terminal and C-terminal extended SLPs pointed at the 24-mer V-SLP, elongated at the N-terminus, as most optimal vaccine candidate. This SLP was efficiently cross-presented and consistently induced a strong polyclonal LRPAP121-30-specific CD8 T cells from the endogenous T cell repertoire. Thus, we designed a TEIPP SLP vaccine from the LRPAP1 signal sequence ready for validation in clinical trials.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 2/metabolism , Adenocarcinoma of Lung/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Lung Neoplasms/immunology , Peptide Fragments/pharmacology , T-Lymphocytes, Cytotoxic/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Antigen Presentation/immunology , Antigens, Neoplasm , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Humans , LDL-Receptor Related Protein-Associated Protein/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Sorting Signals , Tumor Cells, Cultured , Tumor Escape
6.
Cell Commun Signal ; 20(1): 162, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261846

ABSTRACT

BACKGROUND: Frameshift mutations in LRPAP1 are responsible for autosomal recessive high myopia in human beings but its underlying mechanism remains elusive. This study aims to investigate the effect of LRPAP1 defect on ocular refractive development and its involved mechanism. METHODS: A lrpap1 mutant zebrafish line with homozygous frameshift mutation was generated by CRISPR/Cas9 technology and confirmed by Sanger sequencing. The ocular refractive phenotype was analyzed by calculating the relative refractive error (RRE) with vivo photography and histological analysis at different development stages, together with examining ocular structure change via transmission electron microscopy. Further, RNA sequencing and bioinformatics analysis were performed. The potentially involved signaling pathway as well as the interacted protein were investigated in vivo. RESULTS: The lrpap1 homozygous mutant zebrafish line showed myopic phenotype. Specifically, the mutant lines showed larger eye axial length-to-body length in one-month old individuals and a myopic shift with an RRE that changed after two months. Collagen fibers became thinning and disordered in the sclera. Further, RNA sequencing and bioinformatics analysis indicated that apoptosis signaling was activated in mutant line; this was further confirmed by acridine orange and TUNEL staining. Moreover, the expression of TGF-ß protein was elevated in the mutant lines. Finally, the treatment of wild-type embryos with a TGF-ß agonist aggravated the degree of eyeball apoptosis; conversely, the use of a TGF-ß inhibitor mitigated apoptosis in mutant embryos. CONCLUSION: The study provides functional evidence of a link between lrpap1 and myopia, suggesting that lrpap1 deficiency could lead to myopia through TGF-ß-induced apoptosis signaling. Video abstract.


Subject(s)
LDL-Receptor Related Protein-Associated Protein , Myopia , Zebrafish Proteins , Zebrafish , Animals , Humans , Acridine Orange/metabolism , Apoptosis , Collagen/metabolism , Myopia/genetics , Myopia/pathology , Sclera/metabolism , Sclera/pathology , Transforming Growth Factor beta/metabolism , Zebrafish/metabolism
7.
Clin Exp Rheumatol ; 40(11): 2052-2059, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34936547

ABSTRACT

OBJECTIVES: The role of epigenetic mechanisms in the pathogenesis and course of RA as well as response to treatment is increasingly being emphasised. The aim of our study was to determine the ADAMTSL2 and LRPAP1 gene methylation levels in RA patients' serum divided according to disease activity and in comparison with the results with the control group. METHODS: Quantitative real-time methylation-specific PCR was used to analyse the methylation status of the investigated genes. RESULTS: We observed a significant difference in the methylation levels of both the ADAMTSL2 and the LRPAP1 genes in patients with high RA activity compared to patients in remission. CONCLUSIONS: ADAMTSL2 methylation status was inversely correlated with DAS28. High disease activity was associated with lower methylation levels than in remission as well as in the control group. Different results were obtained for the methylation levels of the LRPAP1 gene. High disease activity and the control group were characterised by a higher level of LRPAP1 gene methylation compared to patients in remission. We have proven that methylation may play an important role in the course and severity of RA. The level of ADAMTSL2 and LRPAP1 gene methylation might impact the development of disease and reflect the activity of RA.


Subject(s)
ADAMTS Proteins , Arthritis, Rheumatoid , LDL-Receptor Related Protein-Associated Protein , Humans , ADAMTS Proteins/genetics , Epigenesis, Genetic , Methylation , Severity of Illness Index , LDL-Receptor Related Protein-Associated Protein/genetics
8.
Biol Chem ; 402(11): 1427-1440, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34472763

ABSTRACT

Glycosaminoglycans (GAGs) are essential functional components of the extracellular matrix (ECM). Artificial GAGs like sulfated hyaluronan (sHA) exhibit pro-osteogenic properties and boost healing processes. Hence, they are of high interest for supporting bone regeneration and wound healing. Although sulfated GAGs (sGAGs) appear intracellularly, the knowledge about intracellular effects and putative interaction partners is scarce. Here we used an affinity-purification mass spectrometry-based (AP-MS) approach to identify novel and particularly intracellular sGAG-interacting proteins in human bone marrow stromal cells (hBMSC). Overall, 477 proteins were found interacting with at least one of four distinct sGAGs. Enrichment analysis for protein localization showed that mainly intracellular and cell-associated interacting proteins were identified. The interaction of sGAG with α2-macroglobulin receptor-associated protein (LRPAP1), exportin-1 (XPO1), and serine protease HTRA1 (HTRA1) was confirmed in reverse assays. Consecutive pathway and cluster analysis led to the identification of biological processes, namely processes involving binding and processing of nucleic acids, LRP1-dependent endocytosis, and exosome formation. Respecting the preferentially intracellular localization of sGAG in vesicle-like structures, also the interaction data indicate sGAG-specific modulation of vesicle-based transport processes. By identifying many sGAG-specific interacting proteins, our data provide a resource for upcoming studies aimed at molecular mechanisms and understanding of sGAG cellular effects.


Subject(s)
Glycosaminoglycans/metabolism , High-Temperature Requirement A Serine Peptidase 1/metabolism , Karyopherins/metabolism , LDL-Receptor Related Protein-Associated Protein/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Cells, Cultured , Chromatography, Liquid , Glycosaminoglycans/chemistry , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/isolation & purification , Humans , Karyopherins/chemistry , Karyopherins/isolation & purification , LDL-Receptor Related Protein-Associated Protein/chemistry , LDL-Receptor Related Protein-Associated Protein/isolation & purification , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/isolation & purification , Tandem Mass Spectrometry , Exportin 1 Protein
9.
Exp Dermatol ; 30(1): 112-120, 2021 01.
Article in English | MEDLINE | ID: mdl-32813921

ABSTRACT

Hypertrophic scars are a common complication of burn injuries and represent a major challenge in terms of prevention and treatment. These scars are characterized by a supraphysiological vascular density and by the presence of pathological myofibroblasts (Hmyos) displaying a low apoptosis propensity. However, the nature of the association between these two hallmarks of hypertrophic scarring remains largely unexplored. Here, we show that Hmyos produce signalling entities known as microvesicles that significantly increase the three cellular processes underlying blood vessel formation: endothelial cell proliferation, migration and assembly into capillary-like structures. The release of microvesicles from Hmyos was dose-dependently induced by the serum protein α-2-macroglobulin. Using flow cytometry, we revealed the presence of the α-2-macroglobulin receptor-low-density lipoprotein receptor-related protein 1-on the surface of Hmyos. The inhibition of the binding of α-2-macroglobulin to its receptor abolished the shedding of proangiogenic microvesicles from Hmyos. These findings suggest that the production of microvesicles by Hmyos contributes to the excessive vascularization of hypertrophic scars. α-2-Macroglobulin modulates the release of these microvesicles through interaction with low-density lipoprotein receptor-related protein 1.


Subject(s)
Cell-Derived Microparticles/metabolism , Cicatrix, Hypertrophic/metabolism , Myofibroblasts , Neovascularization, Pathologic/metabolism , alpha-Macroglobulins/metabolism , Adult , Cell Movement , Cell Proliferation , Cells, Cultured , Cicatrix, Hypertrophic/pathology , Endothelial Cells/physiology , Humans , LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Myofibroblasts/metabolism , Neovascularization, Pathologic/pathology , Young Adult , alpha-Macroglobulins/pharmacology
10.
Cancer Sci ; 111(12): 4453-4464, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32939876

ABSTRACT

Some cancers are related to atherosclerotic diseases; therefore, these two types of disease may share some antibody biomarkers in common. To investigate this, a first screening of sera was performed from patients with esophageal squamous cell carcinoma (ESCC) or acute ischemic stroke (AIS) for serological identification of antigens using recombinant cDNA expression cloning (SEREX). The amplified luminescent proximity homogeneous assay-linked immunosorbent assay (AlphaLISA) method, which incorporates glutathione donor beads and anti-human IgG acceptor beads, was used to evaluate serum antibody levels. SEREX screening identified low-density lipoprotein receptor-related protein-associated protein 1 (LRPAP1) as a target antigen of serum IgG antibodies in the sera of patients with ESCC or AIS. Antigens, including recombinant glutathione S-transferase-fused LRPAP1 protein, were prepared to examine serum antibody levels. AlphaLISA revealed significantly higher antibody levels against the LRPAP1 protein in patients with solid cancers such as ESCC and colorectal carcinoma and some atherosclerosis-related diseases such as AIS and diabetes mellitus compared with healthy donors. Correlation analysis revealed that the elevated serum antibody levels against LRPAP1 were associated with smoking, a well-known risk factor for both cancer and atherosclerosis. Serum LRPAP1 antibody is therefore a common marker for the early diagnosis of some cancers and atherosclerotic diseases and may reflect diseases caused by habitual smoking.


Subject(s)
Esophageal Neoplasms/blood , Esophageal Squamous Cell Carcinoma/blood , Immunoglobulin G/blood , Ischemic Stroke/blood , LDL-Receptor Related Protein-Associated Protein/immunology , Acute Disease , Biomarkers/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/immunology , DNA, Complementary , Esophageal Neoplasms/immunology , Esophageal Squamous Cell Carcinoma/immunology , Humans , Immunoenzyme Techniques , Ischemic Stroke/immunology , Neoplasm Proteins/immunology
11.
Med Sci Monit ; 25: 7813-7825, 2019 Oct 18.
Article in English | MEDLINE | ID: mdl-31625533

ABSTRACT

BACKGROUND The aim of this study was to investigate the protective mechanism of neurovascular unit of Buyang Huanwu decoction (BYHWD) in an Alzheimer's disease (AD) cell model via RAGE/LRP1 pathway and find a reliable target for Alzheimer's disease treatment. MATERIAL AND METHODS Rat brain microvessel endothelial cells (BMECs) were cultured in 10% FBS and 1% penicillin/streptomycin. The AD model was established by administration of 24 µmol/L amyloid-ß peptides 25~35. Different concentrations of BYHWD (0.1 mg/mL, 1 mg/mL, and 10 mg/mL) were added as the drug intervention. The morphology of the cells was observed by light microscopy and the ultrastructure of the cells was observed by microscopy. The inflammatory factors IL-1ß, IL-6, TNF-alpha, and Aß25-35 were detected by ELISA. Flow cytometry was used to assess the apoptosis rate. The expressions of RAGE, LRP1, ICAM-1, VCAM-1, Apo J, Apo E, and NF-kappaBp65 were detected by Western blotting. RESULTS The structure of cells in BYHWDM and BYHWDH gradually recovered with increasing dose. BYHWD decreased the apoptotic rate of BMECs induced by Aß25-35. The cells treated with different concentrations of BYHWD had significant difference in terms of anti-apoptotic effect. The therapeutic effect of BYHWD on AD was via the RAGE/LRP1 and NF-kappaBp65 pathways. CONCLUSIONS BYHWD regulates Aß metabolism via the RAGE/LRP1 pathway, inhibits vascular endothelial inflammation induced by ICAM-1 and VCAM-1 via the NF-kappaBP65 pathway, and promotes morphological changes induced by Aß-induced brain microvascular endothelial cell damage.


Subject(s)
Alzheimer Disease/drug therapy , Drugs, Chinese Herbal/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Cell Proliferation/drug effects , Drugs, Chinese Herbal/metabolism , Endothelial Cells/metabolism , Inflammation/drug therapy , Inflammation/metabolism , LDL-Receptor Related Protein-Associated Protein/drug effects , LDL-Receptor Related Protein-Associated Protein/metabolism , Models, Biological , Primary Cell Culture , Rats , Receptor for Advanced Glycation End Products/drug effects , Signal Transduction/drug effects
12.
J Biol Chem ; 292(45): 18699-18712, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28972143

ABSTRACT

In the CNS, microglia are activated in response to injury or infection and in neurodegenerative diseases. The endocytic and cell signaling receptor, LDL receptor-related protein-1 (LRP1), is reported to suppress innate immunity in macrophages and oppose microglial activation. The goal of this study was to identify novel mechanisms by which LRP1 may regulate microglial activation. Using primary cultures of microglia isolated from mouse brains, we demonstrated that LRP1 gene silencing increases expression of proinflammatory mediators; however, the observed response was modest. By contrast, the LRP1 ligand, receptor-associated protein (RAP), robustly activated microglia, and its activity was attenuated in LRP1-deficient cells. An important element of the mechanism by which RAP activated microglia was its ability to cause LRP1 shedding from the plasma membrane. This process eliminated cellular LRP1, which is anti-inflammatory, and generated a soluble product, shed LRP1 (sLRP1), which is potently proinflammatory. Purified sLRP1 induced expression of multiple proinflammatory cytokines and the mRNA encoding inducible nitric-oxide synthase in both LRP1-expressing and -deficient microglia. LPS also stimulated LRP1 shedding, as did the heat-shock protein and LRP1 ligand, calreticulin. Other LRP1 ligands, including α2-macroglobulin and tissue-type plasminogen activator, failed to cause LRP1 shedding. Treatment of microglia with a metalloproteinase inhibitor inhibited LRP1 shedding and significantly attenuated RAP-induced cytokine expression. RAP and sLRP1 both caused neuroinflammation in vivo when administered by stereotaxic injection into mouse spinal cords. Collectively, these results suggest that LRP1 shedding from microglia may amplify and sustain neuroinflammation in response to proinflammatory stimuli.


Subject(s)
Cell-Derived Microparticles/metabolism , Cerebral Cortex/metabolism , Inflammation Mediators/agonists , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Animals, Newborn , Calreticulin/genetics , Calreticulin/metabolism , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/immunology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/immunology , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , LDL-Receptor Related Protein-Associated Protein/metabolism , Ligands , Lipopolysaccharides/toxicity , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , Microglia/immunology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RNA Interference , Receptors, LDL/agonists , Receptors, LDL/antagonists & inhibitors , Receptors, LDL/genetics , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/agonists , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
13.
J Neuroinflammation ; 15(1): 91, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29558961

ABSTRACT

BACKGROUND: Apolipoprotein E (ApoE) is the major carrier protein that mediates the transport and delivery of cholesterol and other lipids in the brain. Three isoforms of ApoE (ApoE2, ApoE3, ApoE4) exist in humans, and their relative expression levels impact HIV-1 infection, HIV-1/AIDS disease progression, and cognitive decline associated with HIV-1-associated neurocognitive disorder. Because HIV-1 Tat, a viral protein essential for HIV-1 replication, can bind to low-density lipoprotein receptor-related protein 1 (LRP1) that controls ApoE uptake in the brain, we determined the extent to which different isoforms of ApoE affected Tat-mediated HIV-1 LTR transactivation. METHODS: Using U87MG glioblastoma cells expressing LTR-driven luciferase, we determined the extent to which LRP1 as well as ApoE2, ApoE3, and ApoE4 affected Tat-mediated HIV-1 LTR transactivation. RESULTS: A specific LRP1 antagonist and siRNA knockdown of LRP1 both restricted significantly Tat-mediated LTR transactivation. Of the three ApoEs, ApoE4 was the least potent and effective at preventing HIV-1 Tat internalization and at decreasing Tat-mediated HIV-1 LTR transactivation. Further, Tat-mediated LTR transactivation was attenuated by an ApoE mimetic peptide, and ApoE4-induced restriction of Tat-mediated LTR transactivation was potentiated by an ApoE4 structure modulator that changes ApoE4 into an ApoE3-like phenotype. CONCLUSIONS: These findings help explain observed differential effects of ApoEs on HIV-1 infectivity and the prevalence of HAND in people living with HIV-1 infection and suggest that ApoE mimetic peptides and ApoE4 structure modulator might be used as a therapeutic strategy against HIV-1 infection and associated neurocognitive disorders.


Subject(s)
Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , HIV Long Terminal Repeat/physiology , Transcriptional Activation/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E3/pharmacology , Apolipoprotein E4/genetics , Apolipoprotein E4/pharmacology , Cell Line, Tumor , Cholesterol, HDL/metabolism , Dose-Response Relationship, Drug , HIV Long Terminal Repeat/genetics , Humans , LDL-Receptor Related Protein-Associated Protein/pharmacology , Neuroblastoma/pathology , Protein Transport/drug effects , Protein Transport/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcriptional Activation/drug effects , Transfection
14.
J Biol Chem ; 291(35): 18430-9, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27402839

ABSTRACT

The LDL receptor-related protein 1 (LRP1) is a large endocytic receptor that binds and mediates the endocytosis of numerous structurally diverse ligands. Currently, the basis for ligand recognition by LRP1 is not well understood. LRP1 requires a molecular chaperone, termed the receptor-associated protein (RAP), to escort the newly synthesized receptor from the endoplasmic reticulum to the Golgi. RAP is a three-domain protein that contains the following two high affinity binding sites for LRP1: one is located within domains 1 and 2, and one is located in its third domain. Studies on the interaction of the RAP third domain with LRP1 reveal critical contributions by lysine 256 and lysine 270 for this interaction. From these studies, a model for ligand recognition by this class of receptors has been proposed. Here, we employed surface plasmon resonance to investigate the binding of RAP D1D2 to LRP1. Our results reveal that the high affinity of D1D2 for LRP1 results from avidity effects mediated by the simultaneous interactions of lysine 60 in D1 and lysine 191 in D2 with sites on LRP1 to form a bivalent D1D2-LRP1 complex. When lysine 60 and 191 are both mutated to alanine, the binding of D1D2 to LRP1 is ablated. Our data also reveal that D1D2 is able to bind to a second distinct site on LRP1 to form a monovalent complex. The studies confirm the canonical model for ligand recognition by this class of receptors, which is initiated by pairs of lysine residues that dock into acidic pockets on the receptor.


Subject(s)
LDL-Receptor Related Protein-Associated Protein/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Humans , LDL-Receptor Related Protein-Associated Protein/genetics , LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , Protein Domains
15.
Biochem Biophys Res Commun ; 493(1): 509-513, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28867180

ABSTRACT

Lactoferrin (LF) is a multifunctional, iron-binding glycoprotein in mammalian secretions, such as breast milk, and has several beneficial effects for human health. However, how these effects are exerted at the cellular level is still largely unknown. In this study, we investigated the effects of LF on autophagy activity in NIH/3T3 mouse fibroblasts. LF from bovine milk was found to increase LC3-I to LC3-II conversion and LC3-positive cytosolic punctate structures because of increased autophagy flux. Knockdown of the putative LF receptor low-density receptor-related protein 1 (LRP1) completely abolished LC3 conversion in cells by LF treatment. Moreover, exposure to LF increased the phosphorylation levels of AMPK in cells, and treatment of dorsomorphin, a pharmacological inhibitor of AMPK signaling, attenuated LC3 conversion by LF. Therefore, we concluded that the beneficial effects of LF might be due to an increase of autophagy activity via AMPK signaling through the LRP1 receptor. These findings provide a novel insight into the physiological role of LF for the maintenance of cellular and tissue homeostasis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Autophagy/physiology , LDL-Receptor Related Protein-Associated Protein/metabolism , Lactoferrin/administration & dosage , Animals , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Mice , NIH 3T3 Cells , Phosphorylation
16.
J Endocrinol Invest ; 40(9): 991-997, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28382504

ABSTRACT

OBJECTIVE: Sortilin, a Vps10p family member, is expressed by thyroid epithelial cells (TEC), where it binds to internalized thyroglobulin (Tg) molecules. Premature binding of Tg to sortilin during biosynthesis may cause intracellular retention of Tg. Such a premature interaction may be prevented by one or more inhibitor/s. Because both sortilin and Tg bind to the low-density lipoprotein receptor-associated protein (RAP), we investigated whether RAP serves such a function. METHODS: Immunofluorescence staining for sortilin, Tg, and RAP was performed in FRTL-5 cells. Co-immunoprecipitation experiments were performed in extracts from FRTL-5 or COS-7 cells, the former co-transfected with Tg and/or RAP and/or sortilin, or in thyroid extracts from RAP KO mice. RESULTS: Tg and sortilin did not co-localize in FRTL-5 cells following inhibition of protein synthesis, suggesting that newly synthesized, endogenous sortilin and Tg do not interact, in confirmation of which an anti-sortilin antibody did not co-precipitate Tg in FRTL-5 cells. In contrast, Tg co-localized with RAP in FRTL-5 cells. Co-immunoprecipitation of Tg with an anti-sortilin antibody in COS-7 cells transfected with sortilin and Tg was abolished when cells were co-transfected with RAP, indicating that RAP prevents binding of Tg to sortilin during biosynthesis, in confirmation of which an anti-sortilin antibody co-precipitated Tg in thyroid extracts from RAP KO mice to a greater extent than in thyroid extracts from WT mice. CONCLUSIONS: Tg does not bind prematurely to sortilin because of its interaction with RAP during protein biosynthesis. These findings add new information to the knowledge of thyroid physiology.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , LDL-Receptor Related Protein-Associated Protein/metabolism , Thyroglobulin/metabolism , Adaptor Proteins, Vesicular Transport/analysis , Animals , Biosynthetic Pathways/physiology , COS Cells , Cattle , Cell Line , Chlorocebus aethiops , Female , LDL-Receptor Related Protein-Associated Protein/analysis , Mice , Mice, Knockout , Protein Binding/physiology , Rats , Thyroglobulin/analysis
17.
J Biol Chem ; 290(28): 17262-8, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26013822

ABSTRACT

The low density lipoprotein receptor-related protein 1 (LRP1) is a member of the low density lipoprotein receptor family and plays important roles in a number of physiological and pathological processes. Expression of LRP1 requires the receptor-associated protein (RAP), a molecular chaperone that binds LRP1 and other low density lipoprotein receptor family members in the endoplasmic reticulum and traffics with them to the Golgi where the acidic environment causes its dissociation. Exogenously added RAP is a potent LRP1 antagonist and binds to LRP1 on the cell surface, preventing ligands from binding. Following endocytosis, RAP dissociates in the acidic endosome, allowing LRP1 to recycle back to the cell surface. The acid-induced dissociation of RAP is mediated by its D3 domain, a relatively unstable three-helical bundle that denatures at pH <6.2 due to protonation of key histidine residues on helices 2 and 3. To develop an LRP1 inhibitor that does not dissociate at low pH, we introduced a disulfide bond between the second and third helices in the RAP D3 domain. By combining this disulfide bond with elimination of key histidine residues, we generated a stable RAP molecule that is resistant to both pH- and heat-induced denaturation. This molecule bound to LRP1 with high affinity at both neutral and acidic pH and proved to be a potent inhibitor of LRP1 function both in vitro and in vivo, suggesting that our stable RAP molecule may be useful in multiple pathological settings where LRP1 blockade has been shown to be effective.


Subject(s)
LDL-Receptor Related Protein-Associated Protein/chemistry , LDL-Receptor Related Protein-Associated Protein/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/antagonists & inhibitors , Receptors, LDL/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Animals , Cell Line , Humans , Hydrogen-Ion Concentration , LDL-Receptor Related Protein-Associated Protein/genetics , Mice , Mice, Inbred C57BL , Models, Molecular , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Protein Binding , Protein Denaturation , Protein Engineering , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary
18.
Circulation ; 131(6): 536-49, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25533967

ABSTRACT

BACKGROUND: Cardiovascular disease (CVD) reflects a highly coordinated complex of traits. Although genome-wide association studies have reported numerous single nucleotide polymorphisms (SNPs) to be associated with CVD, the role of most of these variants in disease processes remains unknown. METHODS AND RESULTS: We built a CVD network using 1512 SNPs associated with 21 CVD traits in genome-wide association studies (at P≤5×10(-8)) and cross-linked different traits by virtue of their shared SNP associations. We then explored whole blood gene expression in relation to these SNPs in 5257 participants in the Framingham Heart Study. At a false discovery rate <0.05, we identified 370 cis-expression quantitative trait loci (eQTLs; SNPs associated with altered expression of nearby genes) and 44 trans-eQTLs (SNPs associated with altered expression of remote genes). The eQTL network revealed 13 CVD-related modules. Searching for association of eQTL genes with CVD risk factors (lipids, blood pressure, fasting blood glucose, and body mass index) in the same individuals, we found examples in which the expression of eQTL genes was significantly associated with these CVD phenotypes. In addition, mediation tests suggested that a subset of SNPs previously associated with CVD phenotypes in genome-wide association studies may exert their function by altering expression of eQTL genes (eg, LDLR and PCSK7), which in turn may promote interindividual variation in phenotypes. CONCLUSIONS: Using a network approach to analyze CVD traits, we identified complex networks of SNP-phenotype and SNP-transcript connections. Integrating the CVD network with phenotypic data, we identified biological pathways that may provide insights into potential drug targets for treatment or prevention of CVD.


Subject(s)
Cardiovascular Diseases/genetics , Gene Regulatory Networks/genetics , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Adult , Chromosome Mapping , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 1/genetics , Female , Gene Expression , Genetic Variation , Genome-Wide Association Study , Humans , LDL-Receptor Related Protein-Associated Protein/genetics , Lipoproteins, HDL/genetics , Lipoproteins, LDL/genetics , Male , Phenotype , Risk Factors , Smoking/genetics
19.
Am J Hum Genet ; 93(2): 313-20, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23830514

ABSTRACT

Myopia is an extremely common eye disorder but the pathogenesis of its isolated form, which accounts for the overwhelming majority of cases, remains poorly understood. There is strong evidence for genetic predisposition to myopia, but determining myopia genetic risk factors has been difficult to achieve. We have identified Mendelian forms of myopia in four consanguineous families and implemented exome/autozygome analysis to identify homozygous truncating variants in LRPAP1 and CTSH as the likely causal mutations. LRPAP1 encodes a chaperone of LRP1, which is known to influence TGF-ß activity. Interestingly, we observed marked deficiency of LRP1 and upregulation of TGF-ß in cells from affected individuals, the latter being consistent with available data on the role of TGF-ß in the remodeling of the sclera in myopia and the high frequency of myopia in individuals with Marfan syndrome who characteristically have upregulation of TGF-ß signaling. CTSH, on the other hand, encodes a protease and we show that deficiency of the murine ortholog results in markedly abnormal globes consistent with the observed human phenotype. Our data highlight a role for LRPAP1 and CTSH in myopia genetics and demonstrate the power of Mendelian forms in illuminating new molecular mechanisms that may be relevant to common phenotypes.


Subject(s)
Cathepsin H/genetics , LDL-Receptor Related Protein-Associated Protein/genetics , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Marfan Syndrome/genetics , Mutation , Myopia/genetics , Transforming Growth Factor beta/genetics , Adolescent , Animals , Cathepsin H/metabolism , Child , Child, Preschool , Female , Gene Expression , Genetic Predisposition to Disease , Homozygote , Humans , Infant , LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Male , Marfan Syndrome/metabolism , Marfan Syndrome/pathology , Mice , Myopia/metabolism , Myopia/pathology , Pedigree , Phenotype , Sclera/metabolism , Sclera/pathology , Severity of Illness Index , Transforming Growth Factor beta/metabolism
20.
J Neuroinflammation ; 13(1): 304, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27931217

ABSTRACT

BACKGROUND: Neuroinflammation is characterized by microglial activation and the increased levels of cytokines and chemokines in the central nervous system (CNS). Recent evidence has implicated both beneficial and toxic roles of microglia when over-activated upon nerve injury or in neurodegenerative diseases, including Alzheimer's disease (AD). The low-density lipoprotein receptor-related protein 1 (LRP1) is a major receptor for apolipoprotein E (apoE) and amyloid-ß (Aß), which play critical roles in AD pathogenesis. LRP1 regulates inflammatory responses in peripheral tissues by modulating the release of inflammatory cytokines and phagocytosis. However, the roles of LRP1 in brain innate immunity and neuroinflammation remain unclear. METHODS: In this study, we determined whether LRP1 modulates microglial activation by knocking down Lrp1 in mouse primary microglia. LRP1-related functions in microglia were also assessed in the presence of LRP1 antagonist, the receptor-associated protein (RAP). The effects on the production of inflammatory cytokines were measured by quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). Potential involvement of specific signaling pathways in LRP1-regulated functions including mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB) were assessed using specific inhibitors. RESULTS: We found that knocking down of Lrp1 in mouse primary microglia led to the activation of both c-Jun N-terminal kinase (JNK) and NF-κB pathways with corresponding enhanced sensitivity to lipopolysaccharide (LPS) in the production of pro-inflammatory cytokines. Similar effects were observed when microglia were treated with LRP1 antagonist RAP. In addition, treatment with pro-inflammatory stimuli suppressed Lrp1 expression in microglia. Interestingly, NF-κB inhibitor not only suppressed the production of cytokines induced by the knockdown of Lrp1 but also restored the down-regulated expression of Lrp1 by LPS. CONCLUSIONS: Our study uncovers that LRP1 suppresses microglial activation by modulating JNK and NF-κB signaling pathways. Given that dysregulation of LRP1 has been associated with AD pathogenesis, our work reveals a critical regulatory mechanism of microglial activation by LRP1 that could be associated with other AD-related pathways thus further nominating LRP1 as a potential disease-modifying target for the treatment of AD.


Subject(s)
MAP Kinase Kinase 4/metabolism , Microglia/immunology , NF-kappa B/metabolism , Receptors, LDL/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , LDL-Receptor Related Protein-Associated Protein/pharmacology , Lipopolysaccharides/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Inbred C57BL , Microglia/drug effects , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, LDL/genetics , Signal Transduction/drug effects , Transfection , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL