1
|
Majou D, Dermenghem AL. Effects of DHA (omega-3 fatty acid) and estradiol on amyloid β-peptide regulation in the brain. Brain Res 2024; 1823:148681. [PMID: 37992797 DOI: 10.1016/j.brainres.2023.148681] [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: 09/15/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
In the early stages of sporadic Alzheimer's disease (SAD), there is a strong correlation between memory impairment and cortical levels of soluble amyloid-β peptide oligomers (Aβ). It has become clear that Aβ disrupt glutamatergic synaptic function, which can in turn lead to the characteristic cognitive deficits of SAD, but the actual pathways are still not well understood. This opinion article describes the pathogenic mechanisms underlying cerebral amyloidosis. These mechanisms are dependent on the amyloid precursor protein and concern the synthesis of Aβ peptides with competition between the non-amyloidogenic pathway and the amyloidogenic pathway (i.e. a competition between the ADAM10 and BACE1 enzymes), on the one hand, and the various processes of Aβ residue clearance, on the other hand. This clearance mobilizes both endopeptidases (NEP, and IDE) and removal transporters across the blood-brain barrier (LRP1, ABCB1, and RAGE). Lipidated ApoE also plays a major role in all processes. The disturbance of these pathways induces an accumulation of Aβ. The description of the mechanisms reveals two key molecules in particular: (i) free estradiol, which has genomic and non-genomic action, and (ii) free DHA as a preferential ligand of PPARα-RXRα and PPARɣ-RXRα heterodimers. DHA and free estradiol are also self-regulating, and act in synergy. When a certain level of chronic DHA and free estradiol deficiency is reached, a permanent imbalance is established in the central nervous system. The consequences of these deficits are revealed in particular by the presence of Aβ peptide deposits, as well as other markers of the etiology of SAD.
Collapse
Affiliation(s)
- Didier Majou
- ACTIA, 149, rue de Bercy, 75595 Paris Cedex 12, France.
| | | |
Collapse
|
2
|
Jakovac H, Stašić N, Krašević M, Jonjić N, Radošević-Stašić B. Expression profiles of metallothionein-I/II and megalin/LRP-2 in uterine cervical squamous lesions. Virchows Arch 2020; 478:735-746. [PMID: 33084977 PMCID: PMC7990851 DOI: 10.1007/s00428-020-02947-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/18/2020] [Accepted: 10/09/2020] [Indexed: 12/29/2022]
Abstract
Metallothioneins (MTs) are phylogenetically old cysteine-rich proteins, which are implicated in a variety of physiological and pathological processes. Their growth-regulating, anti-apoptotic and anti-inflammatory functions have been attributed not only to intracellular free radical scavenging and to zinc and copper regulation but also to the ability of secreted MT to bind on surface lipoprotein receptor-megalin/LRP2, which enables the endocytosis of MT-I/II and a wide range of other functionally distinct ligands. In the present study, we analysed the expression pattern of both proteins in 55 cases of premalignant transformation of cervical squamous cells, i.e. in low- and high-grade squamous intraepithelial lesion (LSIL and HSIL). The data showed that in LSIL (cervical intraepithelial neoplasia CIN1; N = 25) MTs were present only in basal and parabasal cells and that megalin was only weakly expressed. In HSIL (CIN2; N = 15 and CIN 3/carcinoma in situ; N = 15), however, overexpression and co-localization of MT with megalin were found in the entire hyperplastic epithelium. Moreover, megalin immunoreactivity appeared on the glandular epithelium and vascular endothelium, as well as on lymphatic cells in stroma. Besides, multiple megalin-positive cells expressed phosphorylated Akt1, implying that MT- and/or megalin-dependent prosurvival signal transduction pathways might contribute to the development of severe cervical dysplasia. The data emphasize the diagnostic power of combined MT/megalin analysis in pre-cancer screening.
Collapse
Affiliation(s)
- Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Nikola Stašić
- Teaching Institute of Public Health, Primorsko-goranska County, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - Maja Krašević
- Department of Pathology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - Nives Jonjić
- Department of Pathology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - Biserka Radošević-Stašić
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| |
Collapse
|
3
|
Jakovac H, Grubić Kezele T, Radošević-Stašić B. Expression Profiles of Metallothionein I/II and Megalin in Cuprizone Model of De- and Remyelination. Neuroscience 2018; 388:69-86. [PMID: 30025861 DOI: 10.1016/j.neuroscience.2018.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/20/2018] [Accepted: 07/05/2018] [Indexed: 11/15/2022]
Abstract
Copper chelator cuprizone (CPZ) is neurotoxicant, which selectively disrupts oligodendroglial respiratory chain, leading to oxidative stress and subsequent apoptosis. Demyelination is, however, followed by spontaneous remyelination owing to the activation of intrinsic CNS repair mechanisms. To explore the participation of metallothioneins (MTs) in these processes, in this study we analyzed the expression profiles of MT-I/II and their receptor megalin (low-density lipoprotein receptor related protein-2) in the brain of mice subjected to different protocols of CPZ feeding. Experiments were performed in female C57BL/6 mice fed with 0.25% CPZ during 1, 3 and 5 weeks. They were sacrificed immediately after feeding with CPZ or 2 weeks after the withdrawal of CPZ. The data showed that CPZ-induced demyelination was followed by high astrogliosis and enhanced expression of MTs and megalin in white (corpus callosum and internal capsule) and gray matter of the brain (cortex, hippocampus, and cerebellum). Moreover, in numerous cortical neurons and progenitor cells the signs of MT/megalin interactions and Akt1 phosphorylation was found supporting the hypothesis that MTs secreted from the astrocytes might directly affect the neuronal differentiation and survival. Furthermore, in mice treated with CPZ for 5 weeks the prominent MTs and megalin immunoreactivities were found on several neural stem cells and oligodendrocyte progenitors in subgranular zone of dentate gyrus and subventricular zone of lateral ventricles pointing to high modulatory effect of MTs on adult neuro- and oligodendrogenesis. The data show that MT I/II perform important cytoprotective and growth-regulating functions in remyelinating processes activated after toxic demyelinating insults.
Collapse
Affiliation(s)
- Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 RIJEKA, Croatia
| | - Tanja Grubić Kezele
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 RIJEKA, Croatia
| | - Biserka Radošević-Stašić
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 RIJEKA, Croatia.
| |
Collapse
|
4
|
Wei J, Fan S, Liu B, Zhang B, Su J, Yu D. Transcriptome analysis of the immune reaction of the pearl oyster Pinctada fucata to xenograft from Pinctada maxima. FISH & SHELLFISH IMMUNOLOGY 2017; 67:331-345. [PMID: 28606863 DOI: 10.1016/j.fsi.2017.06.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/13/2017] [Accepted: 06/08/2017] [Indexed: 06/07/2023]
Abstract
The pearl oyster Pinctada maxima exhibits great difficulty to culture pearls through nuclear insertion with an allograft, but it is easy for P. fucata to culture pearls after allografting. If P. fucata could be used as a surrogate mother to culture P. maxima pearls, it would benefit the pearl culture industry of P. maxima. However, this is blocked by the immune rejection of P. fucata against P. maxima mantle grafts. In this study, the immune responses of P. fucata hemocyte to allograft and xenograft were investigated after transplantation by transcriptome analysis. In total, 107.93 Gb clean reads were produced and assembled using the reference genome of P. fucata. Gene Ontology Term enrichment and KEGG enrichment analyses indicated that apoptosis, hippo signaling pathway, oxidation-reduction, MAPK signaling pathway, ribosome, protein processing in endoplasmic reticulum, purine metabolism, NF-kappa B signaling pathway, oxidative phosphorylation, Ras signaling pathway, and ubiquitin mediated proteolysis were involved in response to transplantation. Many genes related to oxidation-reduction reactions, the MAPK signaling pathway, and apoptosis were identified by comparison of the allograft group and the xenograft group at 0 h, 6 h, 12 h, 24 h, 48 h, 72 h, and 96 h post-transplantation. Among them, the expression levels of NADH dehydrogenase, succinate dehydrogenase and other dehydrogenases were increased significantly in the xenograft groups compared with allograft groups at 0 h post transplantation, indicating that a respiratory burst of neutrophils occurred immediately after xenograft transplantation. Additionally, HSP70 was highly expressed from 0 h to 96 h in the xenograft groups, indicating an oyster immune response to the xenograft. The genes enriched in the ribosome and hippo-signaling pathways were also identified, and expression patterns of these DEGs were different as compared between transplantation and control groups. Finally, altered expression levels of 10 randomly selected immune-related DEGs were confirmed by quantitative real-time PCR. These results indicated that oxidation-reduction is likely the key factor responsible for immune rejection to transplantation. The findings should provide some new insight into the molecular mechanism of immune rejection of the host against xenograft, and thus benefit to development of immunosuppressive reagents to facilitate effective xenograft pearling.
Collapse
Affiliation(s)
- Jinfen Wei
- Qinzhou University, Qinzhou 535011, Guangxi, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Sigang Fan
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Baosuo Liu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Jiaqi Su
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Dahui Yu
- Qinzhou University, Qinzhou 535011, Guangxi, China.
| |
Collapse
|
5
|
Lockyer P, Mao H, Fan Q, Li L, Yu-Lee LY, Eissa NT, Patterson C, Xie L, Pi X. LRP1-Dependent BMPER Signaling Regulates Lipopolysaccharide-Induced Vascular Inflammation. Arterioscler Thromb Vasc Biol 2017; 37:1524-1535. [PMID: 28596374 DOI: 10.1161/atvbaha.117.309521] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 05/30/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Bacterial endotoxin (lipopolysaccharide)-mediated sepsis involves dysregulated systemic inflammation, which injures the lung and other organs, often fatally. Vascular endothelial cells act as both targets and mediators of lipopolysaccharide-induced inflammatory responses. Dysfunction of endothelium results in increases of proinflammatory cytokine production and permeability leakage. BMPER (bone morphogenetic protein-binding endothelial regulator), an extracellular modulator of bone morphogenetic protein signaling, has been identified as a vital component in chronic endothelial inflammatory responses and atherosclerosis. However, it is unclear whether BMPER also regulates inflammatory response in an acute setting such as sepsis. To address this question, we investigated the role of BMPER during lipopolysaccharide-induced acute lung injury. APPROACH AND RESULTS Mice missing 1 allele of BMPER (BMPER+/- mice used in the place of BMPER-/- mice that die at birth) were used for lipopolysaccharide challenge. Lipopolysaccharide-induced pulmonary inflammation and injury was reduced in BMPER+/- mice as shown by several measures, including survival rate, infiltration of inflammatory cells, edema, and production of proinflammatory cytokines. Mechanistically, we have demonstrated that BMPER is required and sufficient for the activation of nuclear factor of activated T cells c1. This BMPER-induced nuclear factor of activated T cells activation is coordinated by multiple signaling pathways, including bone morphogenetic protein-independent low-density lipoprotein receptor-related protein 1-extracellular signal-regulated kinase activation, calcineurin signaling, and low-density lipoprotein receptor-related protein 1β-mediated nuclear factor 45 nuclear export in response to BMPER treatment. CONCLUSIONS We conclude that BMPER plays a pivotal role in pulmonary inflammatory response, which provides new therapeutic options against sepsis shock. The new signaling pathway initiated by BMPER/low-density lipoprotein receptor-related protein 1 axis broadens our understanding about BMPER's role in vascular homeostasis.
Collapse
Affiliation(s)
- Pamela Lockyer
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Hua Mao
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Qiying Fan
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Luge Li
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Li-Yuan Yu-Lee
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - N Tony Eissa
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Cam Patterson
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Liang Xie
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.)
| | - Xinchun Pi
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.); Department of Medicine, Section of Athero & Lipo, Cardiovascular Research Institute (H.M., Q.F., L.L., L.X., X.P.), Departments of Molecular and Cellular Biology and Medicine, Section of Immunology Allergy and Rheumatology, Integrative Molecular and Biomedical Sciences (L.Y.Y.L.), and Departments of Medicine and Pathology and Immunology (N.T.E.), Baylor College of Medicine, Houston, TX; and New York-Presbyterian Hospital, New York (C.P.).
| |
Collapse
|
6
|
Chen Q, Ma JX. Canonical Wnt signaling in diabetic retinopathy. Vision Res 2017; 139:47-58. [PMID: 28545982 DOI: 10.1016/j.visres.2017.02.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 11/16/2022]
Abstract
Diabetic retinopathy (DR) is a common eye complication of diabetes, and the pathogenic mechanism of DR is still under investigation. The canonical Wnt signaling pathway is an evolutionarily conserved pathway that plays fundamental roles in embryogenesis and adult tissue homeostasis. Wnt signaling regulates expression of multiple genes that control retinal development and eye organogenesis, and dysregulated Wnt signaling plays pathophysiological roles in many ocular diseases, including DR. This review highlights recent progress in studies of Wnt signaling in DR. We discuss Wnt signaling regulation in the retina and dysregulation of Wnt signaling associated with ocular diseases with an emphasis on DR. We also discuss the therapeutic potential of modulating Wnt signaling in DR. Continued studies in this field will advance our current understanding on DR and contribute to the development of new treatments.
Collapse
Affiliation(s)
- Qian Chen
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Jian-Xing Ma
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States.
| |
Collapse
|
7
|
Endocytic receptor LRP2/megalin—of holoprosencephaly and renal Fanconi syndrome. Pflugers Arch 2017; 469:907-916. [DOI: 10.1007/s00424-017-1992-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 12/31/2022]
|
8
|
Dissmore T, Seye CI, Medeiros DM, Weisman GA, Bradford B, Mamedova L. The P2Y2 receptor mediates uptake of matrix-retained and aggregated low density lipoprotein in primary vascular smooth muscle cells. Atherosclerosis 2016; 252:128-135. [PMID: 27522265 PMCID: PMC5060008 DOI: 10.1016/j.atherosclerosis.2016.07.927] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS The internalization of aggregated low-density lipoproteins (agLDL) mediated by low-density lipoprotein receptor related protein (LRP1) may involve the actin cytoskeleton in ways that differ from the endocytosis of soluble LDL by the LDL receptor (LDLR). This study aims to define novel mechanisms of agLDL uptake through modulation of the actin cytoskeleton, to identify molecular targets involved in foam cell formation in vascular smooth muscle cells (VSMCs). The critical observation that formed the basis for these studies is that under pathophysiological conditions, nucleotide release from blood-derived and vascular cells activates SMC P2Y2 receptors (P2Y2Rs) leading to rearrangement of the actin cytoskeleton and cell motility. Therefore, we tested the hypothesis that P2Y2R activation mediates agLDL uptake by VSMCs. METHODS Primary VSMCs were isolated from aortas of wild type (WT) C57BL/6 and.P2Y2R-/- mice to investigate whether P2Y2R activation modulates LRP1 expression. Cells were transiently transfected with cDNA encoding a hemagglutinin-tagged (HA-tagged) WT P2Y2R, or a mutant P2Y2R that unlike the WT P2Y2R does not bind the cytoskeletal actin-binding protein filamin-A (FLN-A). RESULTS P2Y2R activation significantly increased agLDL uptake, and LRP1 mRNA expression decreased in P2Y2R-/- VSMCs versus WT. SMCs, expressing P2Y2R defective in FLN-A binding, exhibit 3-fold lower LDLR expression levels than SMCs expressing WT P2Y2R, while cells transfected with WT P2Y2R show greater agLDL uptake in both WT and P2Y2R-/- VSMCs versus cells transfected with the mutant P2Y2R. CONCLUSIONS Together, these results show that both LRP1 and LDLR expression and agLDL uptake are regulated by P2Y2R in VSMCs, and that agLDL uptake due to P2Y2R activation is dependent upon cytoskeletal reorganization mediated by P2Y2R binding to FLN-A.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Aorta/metabolism
- Cell Movement
- Cells, Cultured
- Cytoskeleton/metabolism
- Dose-Response Relationship, Drug
- Endocytosis
- Filamins/metabolism
- Foam Cells/metabolism
- Humans
- Lipoproteins, LDL/blood
- Low Density Lipoprotein Receptor-Related Protein-1
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microfilament Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Receptors, LDL/metabolism
- Receptors, Purinergic P2Y2/metabolism
- Signal Transduction
- Tumor Suppressor Proteins/metabolism
- Uridine Triphosphate/chemistry
Collapse
Affiliation(s)
| | - Cheikh I Seye
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Denis M Medeiros
- School of Graduate Studies, University of Missouri, Kansas City, MO, United States
| | - Gary A Weisman
- Department of Biochemistry and Bond Life Sciences Center, University of Missouri, Columbia, United States
| | - Barry Bradford
- Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States
| | - Laman Mamedova
- Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States.
| |
Collapse
|
9
|
Beck BH, Fuller SA, Li C, Green BW, Zhao H, Rawles SD, Webster CD, Peatman E. Hepatic transcriptomic and metabolic responses of hybrid striped bass (Morone saxatilis×Morone chrysops) to acute and chronic hypoxic insult. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2016; 18:1-9. [DOI: 10.1016/j.cbd.2016.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 01/05/2016] [Accepted: 01/17/2016] [Indexed: 02/06/2023]
|
10
|
Low Density Lipoprotein Receptor Related Proteins as Regulators of Neural Stem and Progenitor Cell Function. Stem Cells Int 2016; 2016:2108495. [PMID: 26949399 PMCID: PMC4754494 DOI: 10.1155/2016/2108495] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/24/2015] [Accepted: 01/06/2016] [Indexed: 12/20/2022] Open
Abstract
The central nervous system (CNS) is a highly organised structure. Many signalling systems work in concert to ensure that neural stem cells are appropriately directed to generate progenitor cells, which in turn mature into functional cell types including projection neurons, interneurons, astrocytes, and oligodendrocytes. Herein we explore the role of the low density lipoprotein (LDL) receptor family, in particular family members LRP1 and LRP2, in regulating the behaviour of neural stem and progenitor cells during development and adulthood. The ability of LRP1 and LRP2 to bind a diverse and extensive range of ligands, regulate ligand endocytosis, recruit nonreceptor tyrosine kinases for direct signal transduction and signal in conjunction with other receptors, enables them to modulate many crucial neural cell functions.
Collapse
|
11
|
Ramanathan A, Nelson AR, Sagare AP, Zlokovic BV. Impaired vascular-mediated clearance of brain amyloid beta in Alzheimer's disease: the role, regulation and restoration of LRP1. Front Aging Neurosci 2015; 7:136. [PMID: 26236233 PMCID: PMC4502358 DOI: 10.3389/fnagi.2015.00136] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/02/2015] [Indexed: 12/12/2022] Open
Abstract
Amyloid beta (Aβ) homeostasis in the brain is governed by its production and clearance mechanisms. An imbalance in this homeostasis results in pathological accumulations of cerebral Aβ, a characteristic of Alzheimer’s disease (AD). While Aβ may be cleared by several physiological mechanisms, a major route of Aβ clearance is the vascular-mediated removal of Aβ from the brain across the blood-brain barrier (BBB). Here, we discuss the role of the predominant Aβ clearance protein—low-density lipoprotein receptor-related protein 1 (LRP1)—in the efflux of Aβ from the brain. We also outline the multiple factors that influence the function of LRP1-mediated Aβ clearance, such as its expression, shedding, structural modification and transcriptional regulation by other genes. Finally, we summarize approaches aimed at restoring LRP1-mediated Aβ clearance from the brain.
Collapse
Affiliation(s)
- Anita Ramanathan
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - Amy R Nelson
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - Abhay P Sagare
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
12
|
Cai Y, Nagel DJ, Zhou Q, Cygnar KD, Zhao H, Li F, Pi X, Knight PA, Yan C. Role of cAMP-phosphodiesterase 1C signaling in regulating growth factor receptor stability, vascular smooth muscle cell growth, migration, and neointimal hyperplasia. Circ Res 2015; 116:1120-32. [PMID: 25608528 DOI: 10.1161/circresaha.116.304408] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RATIONALE Neointimal hyperplasia characterized by abnormal accumulation of vascular smooth muscle cells (SMCs) is a hallmark of occlusive disorders such as atherosclerosis, postangioplasty restenosis, vein graft stenosis, and allograft vasculopathy. Cyclic nucleotides are vital in SMC proliferation and migration, which are regulated by cyclic nucleotide phosphodiesterases (PDEs). OBJECTIVE Our goal is to understand the regulation and function of PDEs in SMC pathogenesis of vascular diseases. METHODS AND RESULTS We performed screening for genes differentially expressed in normal contractile versus proliferating synthetic SMCs. We observed that PDE1C expression was low in contractile SMCs but drastically elevated in synthetic SMCs in vitro and in various mouse vascular injury models in vivo. In addition, PDE1C was highly induced in neointimal SMCs of human coronary arteries. More importantly, injury-induced neointimal formation was significantly attenuated by PDE1C deficiency or PDE1 inhibition in vivo. PDE1 inhibition suppressed vascular remodeling of human saphenous vein explants ex vivo. In cultured SMCs, PDE1C deficiency or PDE1 inhibition attenuated SMC proliferation and migration. Mechanistic studies revealed that PDE1C plays a critical role in regulating the stability of growth factor receptors, such as PDGF receptor β (PDGFRβ) known to be important in pathological vascular remodeling. PDE1C interacts with low-density lipoprotein receptor-related protein-1 and PDGFRβ, thus regulating PDGFRβ endocytosis and lysosome-dependent degradation in an low-density lipoprotein receptor-related protein-1-dependent manner. A transmembrane adenylyl cyclase cAMP-dependent protein kinase cascade modulated by PDE1C is critical in regulating PDGFRβ degradation. CONCLUSIONS These findings demonstrated that PDE1C is an important regulator of SMC proliferation, migration, and neointimal hyperplasia, in part through modulating endosome/lysosome-dependent PDGFRβ protein degradation via low-density lipoprotein receptor-related protein-1.
Collapse
Affiliation(s)
- Yujun Cai
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - David J Nagel
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - Qian Zhou
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - Katherine D Cygnar
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - Haiqing Zhao
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - Faqian Li
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - Xinchun Pi
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - Peter A Knight
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.)
| | - Chen Yan
- From the Department of Medicine, Aab Cardiovascular Research Institute (Y.C., D.J.N., Q.Z., C.Y.), Department of Pathology and Laboratory Medicine (F.L.), and Department of Surgery (P.A.K.), School of Medicine and Dentistry, University of Rochester, NY; Department of Biology, Johns Hopkins University, Baltimore, MD (K.D.C., H.Z.); and Department of Medicine, Athero and Lipo Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P.).
| |
Collapse
|
13
|
Gan M, Jiang P, McLean P, Kanekiyo T, Bu G. Low-density lipoprotein receptor-related protein 1 (LRP1) regulates the stability and function of GluA1 α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor in neurons. PLoS One 2014; 9:e113237. [PMID: 25500815 PMCID: PMC4264746 DOI: 10.1371/journal.pone.0113237] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 10/23/2014] [Indexed: 11/18/2022] Open
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional endocytic receptor abundantly expressed in neurons. Increasing evidence demonstrates that LRP1 regulates synaptic integrity and function at the post synapses, at least partially by regulating glutamate receptors. The α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) are critical ionotropic glutamate receptors consisting of homotetramer or heterotetramer of GluA1-4 subunits and play an essential role in synaptic transmission and synaptic plasticity. Our previous work has shown that neuronal deletion of the Lrp1 gene in mice leads to decreased level of GluA1 and reduced long-term potentiation. To understand the underlying mechanism, we investigated the cellular and functional consequences of LRP1 deletion in primary neurons. Here, we show that LRP1 interacts with and regulates the cellular distribution and turnover of GluA1. LRP1 knockdown in mouse primary neurons led to accelerated turnover and decreased cell surface distribution of GluA1, which correspond to decreased phosphorylation of GluA1 at S845 and S831 sites. Decreased LRP1 expression also attenuated AMPA-evoked calcium influx and reduced GluA1-regulated neurite outgrowth and filopodia density. Our results reveal a novel mechanism by which LRP1 controls synaptic integrity and function, specifically by regulating GluA1 trafficking, phosphorylation and turnover. They further demonstrate that LRP1-GluA1 pathway may hold promises as a therapeutic target for restoring synaptic functions in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming Gan
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Peizhou Jiang
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Pamela McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America; Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
14
|
Emonard H, Théret L, Bennasroune AH, Dedieu S. Regulation of LRP-1 expression: make the point. ACTA ACUST UNITED AC 2014; 62:84-90. [PMID: 24661974 DOI: 10.1016/j.patbio.2014.02.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/14/2014] [Indexed: 12/14/2022]
Abstract
The low-density lipoprotein receptor-related protein-1 (LRP-1) is a membrane receptor displaying both scavenging and signaling functions. The wide variety of extracellular ligands and of cytoplasmic scaffolding and signaling proteins interacting with LRP-1 gives it a major role not only in physiological processes, such as embryogenesis and development, but also in critical pathological situations, including cancer and neurological disorders. In this review, we describe the molecular mechanisms involved at distinct levels in the regulation of LRP-1, from its expression to the proper location and stability at the cell surface.
Collapse
Affiliation(s)
- H Emonard
- UMR CNRS 7369, unité MEDyC (matrice extracellulaire et dynamique cellulaire), université de Reims-Champagne-Ardenne (URCA), UFR sciences exactes et naturelles, campus Moulin-de-la-Housse, BP 1039, 51687 Reims cedex 2, France
| | - L Théret
- UMR CNRS 7369, unité MEDyC (matrice extracellulaire et dynamique cellulaire), université de Reims-Champagne-Ardenne (URCA), UFR sciences exactes et naturelles, campus Moulin-de-la-Housse, BP 1039, 51687 Reims cedex 2, France
| | - A H Bennasroune
- UMR CNRS 7369, unité MEDyC (matrice extracellulaire et dynamique cellulaire), université de Reims-Champagne-Ardenne (URCA), UFR sciences exactes et naturelles, campus Moulin-de-la-Housse, BP 1039, 51687 Reims cedex 2, France
| | - S Dedieu
- UMR CNRS 7369, unité MEDyC (matrice extracellulaire et dynamique cellulaire), université de Reims-Champagne-Ardenne (URCA), UFR sciences exactes et naturelles, campus Moulin-de-la-Housse, BP 1039, 51687 Reims cedex 2, France.
| |
Collapse
|
15
|
Liu R, Wang L, Sun Y, Wang L, Zhang H, Song L. A low-density lipoprotein receptor-related protein (LRP)-like molecule identified from Chlamys farreri participated in immune response against bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2014; 36:336-343. [PMID: 24345370 DOI: 10.1016/j.fsi.2013.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/22/2013] [Accepted: 11/25/2013] [Indexed: 06/03/2023]
Abstract
Low-density lipoprotein receptor-related protein (LRP) is a group of important endocytic receptors contributing to binding ligands and maintaining internal environment. In the present study, an LRP-like molecule was identified from Zhikong scallop Chlamys farreri (CfLPR), and its mRNA expression profiles, tissue location, and immunology activities were analyzed to explore its possible function in the innate immune system. The ORF of CfLRP was of 1971 bp encoding a polypeptide of 656 amino acids with ten low-density lipoprotein-receptor YWTD (LY) domains and one scavenger receptor cysteine-rich (SRCR) domain. It shared similar structure with out-membrane domains of LRP family members in mammalian. The mRNA transcripts of CfLRP were dominantly expressed in hepatopancreas and mantle (P < 0.01), and its mRNA level in hemocytes was up-regulated (P < 0.01) significantly after the stimulations of lipopolysaccharides (LPS), peptidoglycan (PGN) and β-glucan. Western blotting assay using polyclonal antibody specific for CfLRP revealed that CfLRP was localized in the plasma. The recombinant protein of CfLRP (rCfLRP) could bind acetylated low density lipoprotein (Ac-LDL), metalloprotease SPF1 of Vibrio splendidus and mannan, but could not bind other typical PAMPs such as LPS, PGN, β-glucan and zymosan. Meanwhile, rCfLRP also exhibited strong bacteriostatic activity to Gram-negative bacteria Vibrio anguillarum and V. splendidus. These results indicated that CfLRP could serve as a receptor to recognize and eliminate the invading pathogens, which provided a new implication in the function of LRP-like molecules in invertebrate immunity.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Bacterial Physiological Phenomena
- Base Sequence
- Blotting, Western
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- Gene Expression Regulation
- Immunity, Innate
- Molecular Sequence Data
- Pectinidae/classification
- Pectinidae/genetics
- Pectinidae/immunology
- Pectinidae/microbiology
- Phylogeny
- Protein Structure, Tertiary
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Random Allocation
- Real-Time Polymerase Chain Reaction
- Receptors, LDL/chemistry
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
Collapse
Affiliation(s)
- Rui Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Lingling Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Ying Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Leilei Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Linsheng Song
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
16
|
Pi X, Schmitt CE, Xie L, Portbury AL, Wu Y, Lockyer P, Dyer LA, Moser M, Bu G, Flynn EJ, Jin SW, Patterson C. LRP1-dependent endocytic mechanism governs the signaling output of the bmp system in endothelial cells and in angiogenesis. Circ Res 2012; 111:564-74. [PMID: 22777006 DOI: 10.1161/circresaha.112.274597] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE Among the extracellular modulators of Bmp (bone morphogenetic protein) signaling, Bmper (Bmp endothelial cell precursor-derived regulator) both enhances and inhibits Bmp signaling. Recently we found that Bmper modulates Bmp4 activity via a concentration-dependent, endocytic trap-and-sink mechanism. OBJECTIVE To investigate the molecular mechanisms required for endocytosis of the Bmper/Bmp4 and signaling complex and determine the mechanism of Bmper's differential effects on Bmp4 signaling. METHODS AND RESULTS Using an array of biochemical and cell biology techniques, we report that LRP1 (LDL receptor-related protein 1), a member of the LDL receptor family, acts as an endocytic receptor for Bmper and a coreceptor of Bmp4 to mediate the endocytosis of the Bmper/Bmp4 signaling complex. Furthermore, we demonstrate that LRP1-dependent Bmper/Bmp4 endocytosis is essential for Bmp4 signaling, as evidenced by the phenotype of lrp1-deficient zebrafish, which have abnormal cardiovascular development and decreased Smad1/5/8 activity in key vasculogenic structures. CONCLUSIONS Together, these data reveal a novel role for LRP1 in the regulation of Bmp4 signaling by regulating receptor complex endocytosis. In addition, these data introduce LRP1 as a critical regulator of vascular development. These observations demonstrate Bmper's ability to fine-tune Bmp4 signaling at the single-cell level, unlike the spatial regulatory mechanisms applied by other Bmp modulators.
Collapse
Affiliation(s)
- Xinchun Pi
- Health Care Entrepreneurship, UNC Center for Heart and Vascular Care, Division of Cardiology, 8200 Medical Biomolecular Research Building, Chapel Hill, NC 27599-7126, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Makarova AM, Lebedeva TV, Nassar T, Higazi AAR, Xue J, Carinato ME, Bdeir K, Cines DB, Stepanova V. Urokinase-type plasminogen activator (uPA) induces pulmonary microvascular endothelial permeability through low density lipoprotein receptor-related protein (LRP)-dependent activation of endothelial nitric-oxide synthase. J Biol Chem 2011; 286:23044-53. [PMID: 21540184 PMCID: PMC3123072 DOI: 10.1074/jbc.m110.210195] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Revised: 04/19/2011] [Indexed: 01/11/2023] Open
Abstract
Urokinase plasminogen activator (uPA) and PA inhibitor type 1 (PAI-1) are elevated in acute lung injury, which is characterized by a loss of endothelial barrier function and the development of pulmonary edema. Two-chain uPA and uPA-PAI-1 complexes (1-20 nM) increased the permeability of monolayers of human pulmonary microvascular endothelial cells (PMVECs) in vitro and lung permeability in vivo. The effects of uPA-PAI-1 were abrogated by the nitric-oxide synthase (NOS) inhibitor L-NAME (N(D)-nitro-L-arginine methyl ester). Two-chain uPA (1-20 nM) and uPA-PAI-1 induced phosphorylation of endothelial NOS-Ser(1177) in PMVECs, which was followed by generation of NO and the nitrosylation and dissociation of β-catenin from VE-cadherin. uPA-induced phosphorylation of eNOS was decreased by anti-low density lipoprotein receptor-related protein-1 (LRP) antibody and an LRP antagonist, receptor-associated protein (RAP), and when binding to the uPA receptor was blocked by the isolated growth factor-like domain of uPA. uPA-induced phosphorylation of eNOS was also inhibited by the protein kinase A (PKA) inhibitor, myristoylated PKI, but was not dependent on PI3K-Akt signaling. LRP blockade and inhibition of PKA prevented uPA- and uPA-PAI-1-induced permeability of PMVEC monolayers in vitro and uPA-induced lung permeability in vivo. These studies identify a novel pathway involved in regulating PMVEC permeability and suggest the utility of uPA-based approaches that attenuate untoward permeability following acute lung injury while preserving its salutary effects on fibrinolysis and airway remodeling.
Collapse
Affiliation(s)
- Anastasia M. Makarova
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Tatiana V. Lebedeva
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Taher Nassar
- the Department of Clinical Biochemistry, Hebrew University-Hadassah Medical Center, Jerusalem 91120, Israel, and
| | - Abd Al-Roof Higazi
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- the Department of Clinical Biochemistry, Hebrew University-Hadassah Medical Center, Jerusalem 91120, Israel, and
| | - Jing Xue
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- the Department of Laboratory Medicine, Tianjin Huanhu Hospital, Tianjin 300060, China
| | - Maria E. Carinato
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Khalil Bdeir
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Douglas B. Cines
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Victoria Stepanova
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
18
|
Reekmans SM, Pflanzner T, Gordts PLSM, Isbert S, Zimmermann P, Annaert W, Weggen S, Roebroek AJM, Pietrzik CU. Inactivation of the proximal NPXY motif impairs early steps in LRP1 biosynthesis. Cell Mol Life Sci 2010; 67:135-45. [PMID: 19856143 PMCID: PMC11115674 DOI: 10.1007/s00018-009-0171-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 09/24/2009] [Accepted: 10/05/2009] [Indexed: 11/25/2022]
Abstract
The proximal NPXY and distal NPXYXXL motifs in the intracellular domain of LRP1 play an important role in regulation of the function of the receptor. The impact of single and double inactivating knock-in mutations of these motifs on receptor maturation, cell surface expression, and ligand internalization was analyzed in mutant and control wild-type mice and MEFs. Single inactivation of the proximal NPXY or in combination with inactivation of the distal NPXYXXL motif are both shown to be associated with an impaired maturation and premature proteasomal degradation of full-length LRP1. Therefore, only a small mature LRP1 pool is able to reach the cell surface resulting indirectly in severe impairment of ligand internalization. Single inactivation of the NPXYXXL motif revealed normal maturation, but direct impairment of ligand internalization. In conclusion, the proximal NPXY motif proves to be essential for early steps in the LRP1 biosynthesis, whereas NPXYXXL appears rather relevant for internalization.
Collapse
Affiliation(s)
- Sara M. Reekmans
- Laboratory for Experimental Mouse Genetics, Center for Human Genetics, KU Leuven, Herestraat 49, bus 602, 3000 Leuven, Belgium
- Laboratory for Experimental Mouse Genetics, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Thorsten Pflanzner
- Molecular Neurodegeneration, Department of Physiological Chemistry and Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Philip L. S. M. Gordts
- Laboratory for Experimental Mouse Genetics, Center for Human Genetics, KU Leuven, Herestraat 49, bus 602, 3000 Leuven, Belgium
- Laboratory for Experimental Mouse Genetics, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Simone Isbert
- Molecular Neurodegeneration, Department of Physiological Chemistry and Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Pascale Zimmermann
- Laboratory for Signal Integration in Cell Fate Decision, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Wim Annaert
- Laboratory of Membrane Trafficking, Center for Human Genetics, KU Leuven, Leuven, Belgium
- Laboratory of Membrane Trafficking, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Sascha Weggen
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anton J. M. Roebroek
- Laboratory for Experimental Mouse Genetics, Center for Human Genetics, KU Leuven, Herestraat 49, bus 602, 3000 Leuven, Belgium
- Laboratory for Experimental Mouse Genetics, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Claus U. Pietrzik
- Molecular Neurodegeneration, Department of Physiological Chemistry and Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| |
Collapse
|
19
|
Gordts PLSM, Reekmans S, Lauwers A, Van Dongen A, Verbeek L, Roebroek AJM. Inactivation of the LRP1 intracellular NPxYxxL motif in LDLR-deficient mice enhances postprandial dyslipidemia and atherosclerosis. Arterioscler Thromb Vasc Biol 2009; 29:1258-64. [PMID: 19667105 DOI: 10.1161/atvbaha.109.192211] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The purpose of this study was to determine the significance of the intracellular NPxYxxL motif of LRP1 for the atheroprotective role of this multifunctional receptor. METHODS AND RESULTS LRP1 knock-in mice carrying an inactivating mutation in the NPxYxxL motif were crossed with LDLR-deficient mice, a model for atherosclerosis. In this LDLR(-/-) background the mutated mice showed a more atherogenic lipoprotein profile, which was associated with a decreased clearance of postprandial lipids because of a compromised endocytosis rate and reduced lipase activity. On an atherogenic diet LRP1 mutant mice revealed a 50% increased development of atherosclerosis. This aggravation was accompanied by an increase in smooth muscle cell (SMC) and collagen content and apoptotic cells in the lesions. The mutation showed, however, a limited impact on basal PDGFR-beta expression and signaling and the antimigratory property of apoE on PDGF-BB-stimulated SMCs. Additionally, levels of LRP1 atherogenic ligands, like MMP2, t-PA, FVIII, and the inflammatory ligand TNF-alpha showed to be significantly elevated. CONCLUSIONS These findings demonstrate that the NPxYxxL motif is essential for the atheroprotective role of LRP1. This motif is relevant for normal control of lipid metabolism and of atherogenic and inflammatory ligands, but has no pronounced effect on regulating PDGF-BB/PDGFR-beta signaling in SMCs.
Collapse
|
20
|
Ay N, Irmler K, Fischer A, Uhlemann R, Reuter G, Humbeck K. Epigenetic programming via histone methylation at WRKY53 controls leaf senescence in Arabidopsis thaliana. Traffic 2009; 58:333-46. [PMID: 19143996 DOI: 10.1111/j.1365-313x.2008.03782.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Leaf senescence, the final step of leaf development, involves extensive reprogramming of gene expression. Here, we show that these processes include discrete changes of epigenetic indexing, as well as global alterations in chromatin organization. During leaf senescence, the interphase nuclei show a decondensation of chromocenter heterochromatin, and changes in the nuclear distribution of the H3K4me2, H3K4me3, and the H3K27me2 and H3K27me3 histone modification marks that index active and inactive chromatin, respectively. Locus-specific epigenetic indexing was studied at the WRKY53 key regulator of leaf senescence. During senescence, when the locus becomes activated, H3K4me2 and H3K4me3 are significantly increased at the 5' end and at coding regions. Impairment of these processes is observed in plants overexpressing the SUVH2 histone methyltransferase, which causes ectopic heterochromatization. In these plants the transcriptional initiation of WRKY53 and of the senescence-associated genes SIRK, SAG101, ANAC083, SAG12 and SAG24 is inhibited, resulting in a delay of leaf senescence. In SUVH2 overexpression plants, significant levels of H3K27me2 and H3K27me3 are detected at the 5'-end region of WRKY53, resulting in its transcriptional repression. Furthermore, SUVH2 overexpression inhibits senescence-associated global changes in chromatin organization. Our data suggest that complex epigenetic processes control the senescence-specific gene expression pattern.
Collapse
Affiliation(s)
- Nicole Ay
- Department of Plant Physiology, Institute of Biology, Martin-Luther University Halle-Wittenberg, Weinbergweg 10, D-06120 Halle, Germany
| | | | | | | | | | | |
Collapse
|
21
|
Tufail M, Takeda M. Insect vitellogenin/lipophorin receptors: molecular structures, role in oogenesis, and regulatory mechanisms. JOURNAL OF INSECT PHYSIOLOGY 2009; 55:87-103. [PMID: 19071131 DOI: 10.1016/j.jinsphys.2008.11.007] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 11/10/2008] [Accepted: 11/13/2008] [Indexed: 05/27/2023]
Abstract
Insect vitellogenin and lipophorin receptors (VgRs/LpRs) belong to the low-density lipoprotein receptor (LDLR) gene superfamily and play a critical role in oocyte development by mediating endocytosis of the major yolk protein precursors Vg and Lp, respectively. Precursor Vg and Lp are synthesized, in the majority of insects, extraovarially in the fat body and are internalized by competent oocytes through membrane-bound receptors (i.e., VgRs and LpRs, respectively). Structural analysis reveals that insect VgRs/LpRs and all other LDLR family receptors share a group of five structural domains: clusters of cysteine-rich repeats constituting the ligand-binding domain (LBD), epidermal growth factor (EGF)-precursor homology domain that mediates the acid-dependent dissociation of ligands, an O-linked sugar domain of unknown function, a transmembrane domain anchoring the receptor in the plasma membrane, and a cytoplasmic domain that mediates the clustering of the receptor into the coated pits. The sequence analysis indicates that insect VgRs harbor two LBDs with five repeats in the first and eight repeats in the second domain as compared to LpRs which have a single 8-repeat LBD. Moreover, the cytoplasmic domain of all insect VgRs contains a LI internalization signal instead of the NPXY motif found in LpRs and in the majority of other LDLR family receptors. The exception is that of Solenopsis invicta VgR, which also contains an NPXY motif in addition to LI signal. Cockroach VgRs still harbor another motif, NPTF, which is also believed to be a functional internalization signal. The expression studies clearly demonstrate that insect VgRs are ovary-bound receptors of the LDLR family as compared to LpRs, which are transcribed in a wide range of tissues including ovary, fat body, midgut, brain, testis, Malpighian tubules, and muscles. VgR/LpR mRNA and the protein were detected in the germarium, suggesting that the genes involved in receptor-endocytotic machinery are specifically expressed long before they are functionally required.
Collapse
Affiliation(s)
- Muhammad Tufail
- Graduate School of Science and Technology, Kobe University, Nada, Kobe 657-8501, Japan.
| | | |
Collapse
|
22
|
Costa-Junior HM, Suetsugu MJ, Krieger JE, Schechtman D. Specific modulation of protein kinase activity via small peptides. ACTA ACUST UNITED AC 2009; 153:11-8. [DOI: 10.1016/j.regpep.2008.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Revised: 12/01/2008] [Accepted: 12/06/2008] [Indexed: 11/15/2022]
|
23
|
Lin S, Cheng M, Dailey W, Drenser K, Chintala S. Norrin attenuates protease-mediated death of transformed retinal ganglion cells. Mol Vis 2009; 15:26-37. [PMID: 19137075 PMCID: PMC2615462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 12/26/2008] [Indexed: 11/17/2022] Open
Abstract
PURPOSE To investigate the effects of norrin, a nonconventional ligand for Wingless-Int (Wnt)-beta-catenin signaling pathway, on protease-mediated death of transformed rat retinal ganglion cells (RGC-5). METHODS Transformed RGC-5 cells were treated with 2.0 microM staurosporine (SS), a broad-spectrum protein kinase-C inhibitor, to induce growth arrest, differentiation, and elevated levels of tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA). RGC-5 cells were also treated with 2.0 microM SS and varying doses of recombinant norrin (3.125 to 100 ng/ml). Activation of Wnt pathway was assessed by nuclear translocation of beta-catenin. Proteolytic activity of tPA and uPA was determined by zymography assays and cell viability was determined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assays. Expression and phosphorylation of the low-density lipoprotein-related receptor-1 (LRP-1), a cell surface receptor for tPA and uPA, was determined by immunoprecipitation and western blot analysis. RESULTS Compared to RGC-5 cells left untreated, cells treated with either SS alone or SS and norrin secreted elevated levels of tPA and uPA. A significant number of RGC-5 cells treated with only SS underwent cell death, whereas cells treated with SS and norrin did not, even though RGC-5 cells secreted elevated levels of tPA and uPA under both treatment conditions. Although norrin activated the Wnt pathway, Dickkopf related protein 1 (Dkk1), an inhibitor of Wnt/beta-catenin pathway, failed to completely block norrin's neuroprotective effects. Assays for expression and phosphorylation of LRP-1 indicated that tPA and uPA cause RGC-5 cell death, in part, by reducing phosphorylation of LRP-1, whereas norrin attenuated tPA and uPA-mediated RGC cell death, in part, by restoring phosphorylation of LRP-1. CONCLUSIONS Our results suggest that norrin attenuates tPA- and uPA-mediated death of RGC-5 cells by activating Wnt/beta-catenin pathway and by regulating phosphorylation of LRP-1.
Collapse
Affiliation(s)
- Song Lin
- Eye Research Institute of Oakland University, Rochester, MI
| | - Mei Cheng
- Eye Research Institute of Oakland University, Rochester, MI
| | - Wendelin Dailey
- Department of Ophthalmology, William Beaumont Hospital, Royal Oak, MI
| | - Kimberly Drenser
- Department of Ophthalmology, William Beaumont Hospital, Royal Oak, MI
| | | |
Collapse
|
24
|
Shiroshima T, Oka C, Kawaichi M. Identification of LRP1B-interacting proteins and inhibition of protein kinase Calpha-phosphorylation of LRP1B by association with PICK1. FEBS Lett 2008; 583:43-8. [PMID: 19071120 DOI: 10.1016/j.febslet.2008.11.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 11/09/2008] [Accepted: 11/18/2008] [Indexed: 11/30/2022]
Abstract
Recent studies show LDL receptor-related protein 1B, LRP1B as a transducer of extracellular signals. Here, we identify six interacting partners of the LRP1B cytoplasmic region by yeast two-hybrid screen and confirmed their in vivo binding by immunoprecipitation. One of the partners, PICK1 recognizes the C-terminus of LRP1B and LRP1. The cytoplasmic domains of LRP1B are phosphorylated by PKCalpha about 100 times more efficiently than LRP1. Binding of PICK1 inhibits phosphorylation of LRP1B, but does not affect LRP1 phosphorylation. This study presents the possibility that LRP1B participates in signal transduction which PICK1 may regulate by inhibiting PKCalpha phosphorylation of LRP1B.
Collapse
Affiliation(s)
- Tomoko Shiroshima
- Division of Gene Function in Animals, Nara Institute of Science and Technology, Takayama, Ikoma, Nara, Japan
| | | | | |
Collapse
|
25
|
Donoso M, Cancino J, Lee J, van Kerkhof P, Retamal C, Bu G, Gonzalez A, Cáceres A, Marzolo MP. Polarized traffic of LRP1 involves AP1B and SNX17 operating on Y-dependent sorting motifs in different pathways. Mol Biol Cell 2008; 20:481-97. [PMID: 19005208 DOI: 10.1091/mbc.e08-08-0805] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is an endocytic recycling receptor with two cytoplasmic tyrosine-based basolateral sorting signals. Here we show that during biosynthetic trafficking LRP1 uses AP1B adaptor complex to move from a post-TGN recycling endosome (RE) to the basolateral membrane. Then it recycles basolaterally from the basolateral sorting endosome (BSE) involving recognition by sorting nexin 17 (SNX17). In the biosynthetic pathway, Y(29) but not N(26) from a proximal NPXY directs LRP1 basolateral sorting from the TGN. A N(26)A mutant revealed that this NPXY motif recognized by SNX17 is required for the receptor's exit from BSE. An endocytic Y(63)ATL(66) motif also functions in basolateral recycling, in concert with an additional endocytic motif (LL(86,87)), by preventing LRP1 entry into the transcytotic apical pathway. All this sorting information operates similarly in hippocampal neurons to mediate LRP1 somatodendritic distribution regardless of the absence of AP1B in neurons. LRP1 basolateral distribution results then from spatially and temporally segregation steps mediated by recognition of distinct tyrosine-based motifs. We also demonstrate a novel function of SNX17 in basolateral/somatodendritic recycling from a different compartment than AP1B endosomes.
Collapse
Affiliation(s)
- Maribel Donoso
- Centro de Regulación Celular y Patología , Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile and the Millenium Institute for Fundamental and Applied Biology, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wojtal KA, Hoekstra D, van Ijzendoorn SCD. cAMP-dependent protein kinase A and the dynamics of epithelial cell surface domains: moving membranes to keep in shape. Bioessays 2008; 30:146-55. [PMID: 18200529 DOI: 10.1002/bies.20705] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) and cAMP-dependent protein kinase A (PKA) are evolutionary conserved molecules with a well-established position in the complex network of signal transduction pathways. cAMP/PKA-mediated signaling pathways are implicated in many biological processes that cooperate in organ development including the motility, survival, proliferation and differentiation of epithelial cells. Cell surface polarity, here defined as the anisotropic organisation of cellular membranes, is a critical parameter for most of these processes. Changes in the activity of cAMP/PKA elicit a variety of effects on intracellular membrane dynamics, including membrane sorting and trafficking. One of the most intriguing aspects of cAMP/PKA signaling is its evolutionary conserved abundance on the one hand and its precise spatial-temporal actions on the other. Here, we review recent developments with regard to the role of cAMP/PKA in the regulation of intracellular membrane trafficking in relation to the dynamics of epithelial surface domains.
Collapse
Affiliation(s)
- Kacper A Wojtal
- Division of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
27
|
Amos S, Mut M, diPierro CG, Carpenter JE, Xiao A, Kohutek ZA, Redpath GT, Zhao Y, Wang J, Shaffrey ME, Hussaini IM. Protein kinase C-alpha-mediated regulation of low-density lipoprotein receptor related protein and urokinase increases astrocytoma invasion. Cancer Res 2007; 67:10241-51. [PMID: 17974965 DOI: 10.1158/0008-5472.can-07-0030] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Aggressive and infiltrative invasion is one of the hallmarks of glioblastoma. Low-density lipoprotein receptor-related protein (LRP) is expressed by glioblastoma, but the role of this receptor in astrocytic tumor invasion remains poorly understood. We show that activation of protein kinase C-alpha (PKC-alpha) phosphorylated and down-regulated LRP expression. Pretreatment of tumor cells with PKC inhibitors, phosphoinositide 3-kinase (PI3K) inhibitor, PKC-alpha small interfering RNA (siRNA), and short hairpin RNA abrogated phorbol 12-myristate 13-acetate-induced down-regulation of LRP and inhibited astrocytic tumor invasion in vitro. In xenograft glioblastoma mouse model and in vitro transmembrane invasion assay, LRP-deficient cells, which secreted high levels of urokinase-type plasminogen activator (uPA), invaded extensively the surrounding normal brain tissue, whereas the LRP-overexpressing and uPA-deficient cells did not invade into the surrounding normal brain. siRNA, targeted against uPA in LRP-deficient clones, attenuated their invasive potential. Taken together, our results strongly suggest the involvement of PKC-alpha/PI3K signaling pathways in the regulation of LRP-mediated astrocytoma invasion. Thus, a strategy of combining small molecule inhibitors of PKC-alpha and PI3K could provide a new treatment paradigm for glioblastomas.
Collapse
Affiliation(s)
- Samson Amos
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Usynin I, Klotz C, Frevert U. Malaria circumsporozoite protein inhibits the respiratory burst in Kupffer cells. Cell Microbiol 2007; 9:2610-28. [PMID: 17573905 DOI: 10.1111/j.1462-5822.2007.00982.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
After transmission by infected mosquitoes, malaria sporozoites rapidly travel to the liver. To infect hepatocytes, sporozoites traverse Kupffer cells, but surprisingly, the parasites are not killed by these resident macrophages of the liver. Here we show that Plasmodium sporozoites and recombinant circumsporozoite protein (CSP) suppress the respiratory burst in Kupffer cells. Sporozoites and CSP increased the intracellular concentration of cyclic adenosyl mono-phosphate (cAMP) and inositol 1,4,5-triphosphate in Kupffer cells, but not in hepatocytes or liver endothelia. Preincubation with cAMP analogues or inhibition of phosphodiesterase also inhibited the respiratory burst. By contrast, adenylyl cyclase inhibition abrogated the suppressive effect of sporozoites. Selective protein kinase A (PKA) inhibitors failed to reverse the CSP-mediated blockage and stimulation of the exchange protein directly activated by cAMP (EPAC), but not PKA inhibited the respiratory burst. Both blockage of the low-density lipoprotein receptor-related protein (LRP-1) with receptor-associated protein and elimination of cell surface proteoglycans inhibited the cAMP increase in Kupffer cells. We propose that by binding of CSP to LRP-1 and cell surface proteoglycans, malaria sporozoites induce a cAMP/EPAC-dependent, but PKA-independent signal transduction pathway that suppresses defence mechanisms in Kupffer cells. This allows the sporozoites to safely pass through these professional phagocytes and to develop inside neighbouring hepatocytes.
Collapse
Affiliation(s)
- Ivan Usynin
- Department of Medical Parasitology, New York University School of Medicine, 341 E 25 St, New York, NY 10010, USA
| | | | | |
Collapse
|
29
|
Yuseff MI, Farfan P, Bu G, Marzolo MP. A cytoplasmic PPPSP motif determines megalin's phosphorylation and regulates receptor's recycling and surface expression. Traffic 2007; 8:1215-30. [PMID: 17555532 DOI: 10.1111/j.1600-0854.2007.00601.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Megalin is a large endocytic receptor expressed at the apical surface of several absorptive epithelia. It binds multiple ligands including apolipoproteins, vitamin and hormone carrier proteins and signaling molecules such as parathyroid hormone and the morphogen sonic hedgehog. An important characteristic of megalin is its high endocytic activity, which is mediated by tyrosine-based endocytic motifs within the receptor's cytoplasmic tail. This domain also harbors several putative consensus phosphorylation motifs for protein kinase (PK) C and casein kinase-II and one consensus motif for PKA and glycogen synthase kinase-3 (GSK3). Here we report that the cytoplasmic domain of megalin is constitutively phosphorylated depending on the integrity of a PPPSP motif, a putative GSK3 site, with a minor participation of the other phosphorylation motifs. Mutation of the serine residue within the PPPSP motif as well as blocking GSK3 activity, with two different inhibitors, significantly decreased the phosphorylation levels of the receptor. Both the megalin PPPAP mutant and the underphosphorylated wild-type receptor, by inhibition of GSK3 activity, were more expressed at the cell surface and more efficiently recycled, but they were not inhibited in their initial endocytosis rates. Altogether, these results show that the PPPSP motif and the GSK3 activity are critical to allow megalin phosphorylation and also negatively regulate the receptor's recycling.
Collapse
Affiliation(s)
- María Isabel Yuseff
- FONDAP Center for Cell Regulation and Pathology (CRCP), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile and MIFAB, Santiago, Chile
| | | | | | | |
Collapse
|
30
|
Neuberger G, Schneider G, Eisenhaber F. pkaPS: prediction of protein kinase A phosphorylation sites with the simplified kinase-substrate binding model. Biol Direct 2007; 2:1. [PMID: 17222345 PMCID: PMC1783638 DOI: 10.1186/1745-6150-2-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Accepted: 01/12/2007] [Indexed: 11/23/2022] Open
Abstract
Background Protein kinase A (cAMP-dependent kinase, PKA) is a serine/threonine kinase, for which ca. 150 substrate proteins are known. Based on a refinement of the recognition motif using the available experimental data, we wished to apply the simplified substrate protein binding model for accurate prediction of PKA phosphorylation sites, an approach that was previously successful for the prediction of lipid posttranslational modifications and of the PTS1 peroxisomal translocation signal. Results Approximately 20 sequence positions flanking the phosphorylated residue on both sides have been found to be restricted in their sequence variability (region -18...+23 with the site at position 0). The conserved physical pattern can be rationalized in terms of a qualitative binding model with the catalytic cleft of the protein kinase A. Positions -6...+4 surrounding the phosphorylation site are influenced by direct interaction with the kinase in a varying degree. This sequence stretch is embedded in an intrinsically disordered region composed preferentially of hydrophilic residues with flexible backbone and small side chain. This knowledge has been incorporated into a simplified analytical model of productive binding of substrate proteins with PKA. Conclusion The scoring function of the pkaPS predictor can confidently discriminate PKA phosphorylation sites from serines/threonines with non-permissive sequence environments (sensitivity of ~96% at a specificity of ~94%). The tool "pkaPS" has been applied on the whole human proteome. Among new predicted PKA targets, there are entirely uncharacterized protein groups as well as apparently well-known families such as those of the ribosomal proteins L21e, L22 and L6. Availability The supplementary data as well as the prediction tool as WWW server are available at . Reviewers Erik van Nimwegen (Biozentrum, University of Basel, Switzerland), Sandor Pongor (International Centre for Genetic Engineering and Biotechnology, Trieste, Italy), Igor Zhulin (University of Tennessee, Oak Ridge National Laboratory, USA).
Collapse
Affiliation(s)
- Georg Neuberger
- IMP – Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | - Georg Schneider
- IMP – Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | - Frank Eisenhaber
- IMP – Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| |
Collapse
|
31
|
Roebroek AJM, Reekmans S, Lauwers A, Feyaerts N, Smeijers L, Hartmann D. Mutant Lrp1 knock-in mice generated by recombinase-mediated cassette exchange reveal differential importance of the NPXY motifs in the intracellular domain of LRP1 for normal fetal development. Mol Cell Biol 2006; 26:605-16. [PMID: 16382151 PMCID: PMC1346909 DOI: 10.1128/mcb.26.2.605-616.2006] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lrp1 knock-in mice carrying either a wild-type allele or three different mutated alleles encoding the multifunctional endocytic receptor LRP1 were generated by recombinase-mediated cassette exchange (RMCE). Reinsertion by RMCE of a wild-type allele led to a normal pattern and level of gene expression and a completely normal phenotype, indicating that the RMCE procedure itself is neutral with respect to the function of the gene locus. In contrast, reinsertion of mutated LRP1 alleles carrying either inactivating mutations in the proximal NPXY motif (NPTY-->AATA) of the cytoplasmic domain or in the furin cleavage site (RHRR-->AHAA) caused distinctive liver phenotypes: respectively, either a late fetal destruction of the organ causing perinatal death or a selective enlargement of von-Kupffer cell lysosomes reminiscent of a mild lysosomal storage without an apparent negative effect on animal survival. Notably, mutation of the distal NPXY motif overlapping with an YXXL motif (NPVYATL-->AAVAATL) did not cause any obvious pathological effect. The mutations showed no effect on the LRP1 expression level; however, as expected, the proteolytic maturation of LRP1 into its two subunits was significantly impaired, although not completely abolished, in the furin cleavage mutant. These data demonstrate that RMCE is a reliable and efficient approach to generate multiple mutant knock-in alleles for in vivo functional analysis of individual domains or motifs of large multidomain proteins. Its application in Lrp1 reveals dramatically variant phenotypes, of which further characterization will definitively contribute to our understanding of the biology of this multifunctional receptor.
Collapse
Affiliation(s)
- Anton J M Roebroek
- Experimental Mouse Genetics, Center for Human Genetics, KU Leuven and Flanders Interuniversity Institute for Biotechnology, Herestraat 49, bus 602, B-3000 Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
32
|
Li X, Herz J, Monard D. Activation of ERK signaling upon alternative protease nexin-1 internalization mediated by syndecan-1. J Cell Biochem 2006; 99:936-51. [PMID: 16741952 DOI: 10.1002/jcb.20881] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Protease nexin-1 (PN-1), an inhibitor of serine proteases, contributes to tissue homeostasis and influences the behavior of some tumor cells. The internalization of PN-1 protease complexes is considered to be mediated by the low-density lipoprotein receptor related protein 1 (LRP1). In this study, both wild-type and LRP1-/- mouse embryonic fibroblasts (MEF) were shown to internalize PN-1. Receptor associated protein (RAP) interfered with PN-1 uptake only in wild-type MEF cells, indicating that another receptor mediates PN-1 uptake in the absence of LRP1. In LRP1-/- MEF cells, inhibitor sensitivity and kinetic values (t(1/2) at 45 min) of PN-1 uptake showed a similarity to syndecan-1-mediated endocytosis. In these cells, PN-1 uptake was increased by overexpression of full-length syndecan-1 and decreased by RNA interference targeting this proteoglycan. Most important, in contrast to PKA activation known to be triggered by LRP1-mediated internalization, our study shows that syndecan-1-mediated internalization of PN-1 stimulated the Ras-ERK signaling pathway.
Collapse
Affiliation(s)
- Xiaobiao Li
- Friedrich Miescher Institute for Biomedical Research, CH-4058, Basel, Switzerland
| | | | | |
Collapse
|
33
|
van Kerkhof P, Lee J, McCormick L, Tetrault E, Lu W, Schoenfish M, Oorschot V, Strous GJ, Klumperman J, Bu G. Sorting nexin 17 facilitates LRP recycling in the early endosome. EMBO J 2005; 24:2851-61. [PMID: 16052210 PMCID: PMC1187941 DOI: 10.1038/sj.emboj.7600756] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Accepted: 07/05/2005] [Indexed: 11/08/2022] Open
Abstract
The low-density lipoprotein (LDL) receptor-related protein (LRP) is a multiligand endocytic receptor and a member of the LDL receptor family. Here we show that sorting nexin 17 (Snx 17) is part of the cellular sorting machinery that regulates cell surface levels of LRP by promoting its recycling. While the phox (PX) domain of Snx 17 interacts with phosphatidylinositol-3-phosphate for membrane association, the FERM domain and the carboxyl-terminal region participate in LRP binding. Immunoelectron microscopy shows that the membrane-bound fraction of Snx 17 is localized to the limiting membrane and recycling tubules of early endosomes. The NPxY motif, proximal to the plasma membrane in the LRP cytoplasmic tail, is identified as the Snx 17-binding motif. Functional mutation of this motif did not interfere with LRP endocytosis, but decreased LRP recycling from endosomes, resulting in increased lysosomal degradation. Similar effects are found after knockdown of endogenous Snx 17 expression by short interfering RNA. We conclude that Snx 17 binds to a motif in the LRP tail distinct from the endocytosis signals and promotes LRP sorting to the recycling pathway in the early endosomes.
Collapse
Affiliation(s)
- Peter van Kerkhof
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Institute of Biomembranes, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jiyeon Lee
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Lynn McCormick
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Elena Tetrault
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Wenyan Lu
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Marissa Schoenfish
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Viola Oorschot
- Department of Cell Biology and Institute of Biomembranes, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ger J Strous
- Department of Cell Biology and Institute of Biomembranes, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Judith Klumperman
- Department of Cell Biology and Institute of Biomembranes, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Guojun Bu
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, CB 8208, 660 South Euclid Ave, St Louis, MO 63110, USA. Tel.: +1 314 286 2860; Fax: +1 314 286 2894; E-mail:
| |
Collapse
|
34
|
Abstract
The low-density lipoprotein receptor (LDLR)-related protein, LRP, is a unique member of the LDLR family. Frequently referred to as a scavenger receptor, LRP is a large transmembrane endocytic receptor that can bind and internalize many functionally distinct ligands. Besides its role as a cargo-receptor, LRP has also been implicated in many signaling pathways. LRP knockout mice die at early embryonic age, which strongly suggests that LRP's functions are essential for normal development. Within the CNS, LRP is highly expressed in neuronal cell bodies and dendritic processes. In vitro, neurite outgrowth is stimulated by apolipoprotein E (apoE)-containing lipoprotein particles via binding to LRP. ApoE is the major cholesterol transporter in the brain and human carriers of one or two copies of the e4 allele of apoE are at a higher risk of developing Alzheimer's disease (AD). LRP also binds the amyloid precursor protein (APP) and its proteolytic fragment, the amyloid-beta peptide (Abeta), which are major players in the pathogenesis of AD. Finally, LRP has been linked to AD by genetic evidence. In this review we discuss the potential mechanisms by which LRP can affect APP and Abeta metabolism, and therefore contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Celina V Zerbinatti
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
35
|
May P, Rohlmann A, Bock HH, Zurhove K, Marth JD, Schomburg ED, Noebels JL, Beffert U, Sweatt JD, Weeber EJ, Herz J. Neuronal LRP1 functionally associates with postsynaptic proteins and is required for normal motor function in mice. Mol Cell Biol 2004; 24:8872-83. [PMID: 15456862 PMCID: PMC517900 DOI: 10.1128/mcb.24.20.8872-8883.2004] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The LDL receptor-related protein 1 (LRP1) is a multifunctional cell surface receptor that is highly expressed on neurons. Neuronal LRP1 in vitro can mediate ligand endocytosis, as well as modulate signal transduction processes. However, little is known about its role in the intact nervous system. Here, we report that mice that lack LRP1 selectively in differentiated neurons develop severe behavioral and motor abnormalities, including hyperactivity, tremor, and dystonia. Since their central nervous systems appear histoanatomically normal, we suggest that this phenotype is likely attributable to abnormal neurotransmission. This conclusion is supported by studies of primary cultured neurons that show that LRP1 is present in close proximity to the N-methyl-D-aspartate (NMDA) receptor in dendritic synapses and can be coprecipitated with NMDA receptor subunits and the postsynaptic density protein PSD-95 from neuronal cell lysates. Moreover, treatment with NMDA, but not dopamine, reduces the interaction of LRP1 with PSD-95, indicating that LRP1 participates in transmitter-dependent postsynaptic responses. Together, these findings suggest that LRP1, like other ApoE receptors, can modulate synaptic transmission in the brain.
Collapse
Affiliation(s)
- Petra May
- Zentrum für Neurowissenschaften, University of Freiburg, Albertstrabetae 23, 79104 Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- David Y Hui
- Department of Pathology and the Genome Research Institute, University of Cincinnati College of Medicine, 2120 E. Galbraith Road, Cincinnati, OH 45237, USA.
| |
Collapse
|
37
|
Pan W, Kastin AJ. Why study transport of peptides and proteins at the neurovascular interface. ACTA ACUST UNITED AC 2004; 46:32-43. [PMID: 15297153 DOI: 10.1016/j.brainresrev.2004.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2004] [Indexed: 01/17/2023]
Abstract
The blood-brain barrier (BBB) is an immense neurovascular interface. In neurodegenerative, ischemic, and traumatic disorders of the central nervous system (CNS), the BBB may hinder the delivery of many therapeutic peptides and proteins to the brain and spinal cord. Fortunately, the mistaken dogma that peptides and proteins do not cross the BBB has been corrected during the past two decades by the accumulating evidence that peptides and proteins in the periphery exert potent effects in the CNS. Not only can peptides and proteins serve as carriers for selective therapeutic agents, but they themselves may directly cross the BBB after delivery into the bloodstream. Their passage may be mediated by simple diffusion or specific transport, both of which can be affected by interactions in the blood compartment (outside the BBB) and within the endothelial cells (at the BBB level). Although the majority of current delivery strategies focuses on modification of the molecule to be delivered, understanding the mechanisms of transport will eventually facilitate regulation of the BBB directly. We review the different aspects of interactions and discuss recent advances in the cell biology of peptide/protein transport across the BBB. Better understanding of the nature and regulation of the transport systems at the BBB will provide a new direction to enhance the interactions of peripheral peptides and proteins with the CNS.
Collapse
Affiliation(s)
- Weihong Pan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | | |
Collapse
|
38
|
Huang SS, Leal SM, Chen CL, Liu IH, Huang JS. Identification of insulin receptor substrate proteins as key molecules for the TβR‐V/LRP‐1‐mediated growth inhibitory signaling cascade in epithelial and myeloid cells. FASEB J 2004; 18:1719-21. [PMID: 15371331 DOI: 10.1096/fj.04-1872fje] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The type V TGF-beta receptor (TbetaR-V) mediates IGF-independent growth inhibition by IGFBP-3 and mediates growth inhibition by TGF-beta1 in concert with the other TGF-beta receptor types. TbetaR-V was recently found to be identical to LRP-1. Here we find that insulin and (Q3A4Y15L16) IGF-I (an IGF-I analog that has a low affinity for IGFBP-3) antagonize growth inhibition by IGFBP-3 in mink lung epithelial cells (Mv1Lu cells) stimulated by serum. In these cells, IGFBP-3 induces serine-specific dephosphorylation of IRS-1 and IRS-2. The IGFBP-3-induced dephosphorylation of IRS-2 is prevented by cotreatment of cells with insulin, (Q3A4Y15L16) IGF-I, or TbetaR-V/LRP-1 antagonists. The magnitude of the IRS-2 dephosphorylation induced by IGFBP-3 positively correlates with the degree of growth inhibition by IGFBP-3 in Mv1Lu cells and mutant cells derived from Mv1Lu cells. Stable transfection of murine 32D myeloid cells (which lack endogenous IRS proteins and are insensitive to growth inhibition by IGFBP-3) with IRS-1 or IRS-2 cDNA confers sensitivity to growth inhibition by IGFBP-3; this IRS-mediated growth inhibition can be completely reversed by insulin in 32D cells stably expressing IRS-2 and the insulin receptor. These results suggest that IRS-1 and IRS-2 are key molecules for the TbetaR-V/LRP-1-mediated growth inhibitory signaling cascade.
Collapse
Affiliation(s)
- Shuan Shian Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1402 South Grand Boulevard St. Louis, MO 63104, USA.
| | | | | | | | | |
Collapse
|
39
|
Ranganathan S, Liu CX, Migliorini MM, Von Arnim CAF, Peltan ID, Mikhailenko I, Hyman BT, Strickland DK. Serine and threonine phosphorylation of the low density lipoprotein receptor-related protein by protein kinase Calpha regulates endocytosis and association with adaptor molecules. J Biol Chem 2004; 279:40536-44. [PMID: 15272003 DOI: 10.1074/jbc.m407592200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The low density lipoprotein receptor-related protein (LRP) is a large receptor that participates in endocytosis, signaling pathways, and phagocytosis of necrotic cells. Mechanisms that direct LRP to function in these distinct pathways likely involve its association with distinct cytoplasmic adaptor proteins. We tested the hypothesis that the association of various adaptor proteins with the LRP cytoplasmic domain is modulated by its phosphorylation state. Phosphoamino acid analysis of metabolically labeled LRP revealed that this receptor is phosphorylated at serine, threonine, and tyrosine residues within its cytoplasmic domain, whereas inhibitor studies identified protein kinase Calpha (PKCalpha) as a kinase capable of phosphorylating LRP. Mutational analysis identified critical threonine and serine residues within the LRP cytoplasmic domain that are necessary for phosphorylation mediated by PKCalpha. Mutating these threonine and serine residues to alanines generated a receptor that was not phosphorylated and that was internalized more rapidly than wild-type LRP, revealing that phosphorylation reduces the association of LRP with adaptor molecules of the endocytic machinery. In contrast, serine and threonine phosphorylation was necessary for the interaction of LRP with Shc, an adaptor protein that participates in signaling events. Furthermore, serine and threonine phosphorylation increased the interaction of LRP with other adaptor proteins such as Dab-1 and CED-6/GULP. These results indicate that phosphorylation of LRP by PKCalpha modulates the endocytic and signaling function of LRP by modifying its association with adaptor proteins.
Collapse
Affiliation(s)
- Sripriya Ranganathan
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Tseng WF, Huang SS, Huang JS. LRP-1/TbetaR-V mediates TGF-beta1-induced growth inhibition in CHO cells. FEBS Lett 2004; 562:71-8. [PMID: 15044004 DOI: 10.1016/s0014-5793(04)00185-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Accepted: 02/06/2004] [Indexed: 11/17/2022]
Abstract
The type V transforming growth factor-beta (TGF-beta) receptor (TbetaR-V) is hypothesized to be involved in cellular growth inhibition by TGF-beta(1). Recently, TbetaR-V was found to be identical to low density lipoprotein receptor-related protein-1 (LRP-1). Here we demonstrate that TGF-beta(1) inhibits growth of wild-type CHO cells but not LRP-1-deficient mutant cells (CHO-LRP-1(-) cells). Stable transfection of CHO-LRP-1(-) cells with LRP-1 cDNA restores the wild-type morphology and the sensitivity to growth inhibition by TGF-beta(1). In addition, overexpression of LRP-1 minireceptors exerts a dominant negative effect and attenuates the growth inhibitory response to TGF-beta(1) in wild-type CHO cells. These results suggest that LRP-1/TbetaR-V is critical for TGF-beta(1)-mediated growth inhibition in CHO cells.
Collapse
Affiliation(s)
- Wen-Fang Tseng
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St. Louis, MO 63104, USA
| | | | | |
Collapse
|
41
|
Huang SS, Leal SM, Chen CL, Liu IH, Huang JS. Cellular growth inhibition by TGF-β1involves IRS proteins. FEBS Lett 2004; 565:117-21. [PMID: 15135063 DOI: 10.1016/j.febslet.2004.03.082] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2004] [Revised: 03/24/2004] [Accepted: 03/24/2004] [Indexed: 11/17/2022]
Abstract
In Mv1Lu cells, insulin partially reverses transforming growth factor-beta1 (TGF-beta1) growth inhibition in the presence of alpha5beta1 integrin antagonists. TGF-beta1 appears to induce phosphorylation of IRS-2 in these cells; this is inhibited by a TGF-beta antagonist known to reverse TGF-beta growth inhibition. Stable transfection of 32D myeloid cells (which lack endogenous IRS proteins and are insensitive to growth inhibition by TGF-beta1) with IRS-1 or IRS-2 cDNA confers sensitivity to growth inhibition by TGF-beta1; this IRS-mediated growth inhibition can be partially reversed by insulin in 32D cells stably expressing IRS-2 and the insulin receptor (IR). These results suggest that growth inhibition by TGF-beta1 involves IRS proteins.
Collapse
Affiliation(s)
- Shuan Shian Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St. Louis, MO 63104, USA.
| | | | | | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW We will discuss the diverse roles of lipoprotein receptors that contribute to the maintenance and integrity of the vascular wall. RECENT FINDINGS Lipoprotein receptors function not only as transporters for cholesterol and other lipids. They also act as sensors and signal transducers through which the endothelium, macrophages and smooth muscle cells communicate with their environment. SUMMARY Traditionally, lipoprotein receptors were thought of merely as transporters of cholesterol and triglycerides to specific target cells, either for the purpose of delivery and redistribution of nutrients, or for the destruction or clearance of modified (oxidized) lipids by macrophages. Only recently have we begun to appreciate that the same receptors engage in a much more sophisticated and multi-faceted interaction with their environment. Inasmuch, they not only act as mere transporters, but as surprisingly versatile and adaptive signal transducers and modulators throughout the vessel wall. These recent findings now begin to reshape our thinking of how such structurally different and evolutionarily unrelated lipoprotein receptors orchestrate the response of the vessel wall to mechanical or metabolic damage.
Collapse
Affiliation(s)
- Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9046, USA.
| | | |
Collapse
|
43
|
Zilberberg A, Yaniv A, Gazit A. The low density lipoprotein receptor-1, LRP1, interacts with the human frizzled-1 (HFz1) and down-regulates the canonical Wnt signaling pathway. J Biol Chem 2004; 279:17535-42. [PMID: 14739301 DOI: 10.1074/jbc.m311292200] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Members of the low density lipoprotein receptor family (LDLR), LRP5/6, were shown to interact with the Frizzled (Fz) receptors and to function as Wnt coreceptors. Here we show that mLRP4T100, a minireceptor of LRP1, another member of the LDLR family, interacts with the human Fz-1 (HFz1), previously shown to serve as a receptor transmitting the canonical Wnt-3a-induced signaling cascade. However, in contrast to LRP5/6, mLRP4T100, as well as the full-length LRP1, did not cooperate with HFz1 in transmitting the Wnt-3a signaling but rather repressed it. mLRP4T100 inhibitory effect was displayed also by endocytosis-defective mLRP4T100 mutants, suggesting that LRP1 repressive effect is not attributable to LRP1-mediated enhanced HFz1 internalization and subsequent degradation. Enforced expression of mLRP4T100 decreased the capacity of HFz1 cysteine-rich domain (CRD) to interact with LRP6, in contrast to HFz1-CRD/Wnt-3a interaction that was not disrupted by overexpressing mLRP4T100. These data suggest that LRP1, by sequestering HFz1, disrupts the receptor/coreceptor complex formation, leading to the repression of the canonical Wnt signaling. Thus, this study implies that the ability to interact with Fz receptors is shared by several members of the LDLR family. However, whereas some members of the LDLR family, such as LRP5/6, interact with Fz and serve as Wnt coreceptors, others negatively regulate Wnt signaling, presumably by sequestering Fz.
Collapse
Affiliation(s)
- Alona Zilberberg
- Department of Human Microbiology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | |
Collapse
|
44
|
Affiliation(s)
- Olav M Andersen
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | | |
Collapse
|
45
|
Strickland DK, Ranganathan S. Diverse role of LDL receptor-related protein in the clearance of proteases and in signaling. J Thromb Haemost 2003; 1:1663-70. [PMID: 12871303 DOI: 10.1046/j.1538-7836.2003.00330.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The low density lipoprotein receptor-related protein (LRP) is a large endocytic receptor that participates in several biological pathways and plays prominent roles in lipoprotein metabolism and in the catabolism of proteinases involved in coagulation and fibrinolysis. LRP also mediates the cellular entry of certain viruses and toxins and facilitates the activation of various lysosomal enzymes. Deletion of the LRP gene in mice is lethal, confirming an important role for this receptor in development, although its exact function in development is still not known. In addition to its role in the endocytosis of numerous ligands, recent studies are emerging that describe a signaling role for this receptor as well.
Collapse
Affiliation(s)
- D K Strickland
- Department of Vascular Biology, Jerome H. Holland Laboratory for the Biomedical Sciences, American Red Cross, Rockville, MD, USA.
| | | |
Collapse
|
46
|
Choi EK, Miller JS, Zaidi NF, Salih E, Buxbaum JD, Wasco W. Phosphorylation of calsenilin at Ser63 regulates its cleavage by caspase-3. Mol Cell Neurosci 2003; 23:495-506. [PMID: 12837631 DOI: 10.1016/s1044-7431(03)00072-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Calsenilin is a member of the neuronal calcium sensor (NCS) family of proteins that interacts with the presenilins. Calsenilin has been found to act as a Kv4alpha channel interactor and as a transcriptional repressor. We have recently shown that calsenilin can be cleaved by caspase-3 and that its cleavage separates the conserved calcium-binding domain from the variable N-terminal domain. Here, we demonstrate that calsenilin can be phosphorylated by casein kinase I and that its phosphorylation can be regulated by intracellular calcium. In addition, phosphorylated calsenilin is a substrate for serine/threonine protein phosphatase (PP) 1 and/or 2A. Phosphorylation within the N-terminal domain at Ser63, the major phosphorylation site of calsenilin, inhibits cleavage of the molecule by caspase-3. Given that the N-terminal domain of calsenilin is not conserved in the larger NCS family including other KChIP/CALP proteins, phosphorylation of calsenilin may regulate a functional role that is unique to this member of the superfamily.
Collapse
Affiliation(s)
- E K Choi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Marzolo MP, Yuseff MI, Retamal C, Donoso M, Ezquer F, Farfán P, Li Y, Bu G. Differential distribution of low-density lipoprotein-receptor-related protein (LRP) and megalin in polarized epithelial cells is determined by their cytoplasmic domains. Traffic 2003; 4:273-88. [PMID: 12694565 DOI: 10.1034/j.1600-0854.2003.00081.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Megalin and the low-density lipoprotein (LDL) receptor-related protein (LRP) are two large members of the LDL receptor family that bind and endocytose multiple ligands. The molecular and cellular determinants that dictate the sorting behavior of these receptors in polarized epithelial cells are largely unknown. Megalin is found apically distributed, whereas the limited information on LRP indicates its polarity. We show here that in Madin-Darby canine kidney cells, both endogenous LRP and a minireceptor containing the fourth ligand-binding, transmembrane and LRP cytosolic domains were basolaterally sorted. In contrast, minireceptors that either lacked the cytoplasmic domain or had the tyrosine in the NPTY motif mutated to alanine showed a preferential apical distribution. In LLC-PK1 cells, endogenous megalin was found exclusively in the apical membrane. Studies were also done using chimeric proteins harboring the cytosolic tail of megalin, one with the fourth ligand-binding domain of LRP and the other two containing the green fluorescent protein as the ectodomain and transmembrane domains of either megalin or LRP. Findings from these experiments showed that the cytosolic domain of megalin is sufficient for apical sorting, and that the megalin transmembrane domain promotes association with lipid rafts. In conclusion, we show that LRP and megalin both contain sorting information in their cytosolic domains that directs opposite polarity, basolateral for LRP and apical for megalin. Additionally, we show that the NPTY motif in LRP is important for basolateral sorting and the megalin transmembrane domain directs association with lipid rafts.
Collapse
Affiliation(s)
- María-Paz Marzolo
- Centro de Regulación Celular y Patología, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Li Y, Knisely JM, Lu W, McCormick LM, Wang J, Henkin J, Schwartz AL, Bu G. Low density lipoprotein (LDL) receptor-related protein 1B impairs urokinase receptor regeneration on the cell surface and inhibits cell migration. J Biol Chem 2002; 277:42366-71. [PMID: 12194987 DOI: 10.1074/jbc.m207705200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The low density lipoprotein (LDL) receptor-related protein 1B (LRP1B) is a newly identified member of the LDL receptor family and is closely related to LRP. It was discovered as a putative tumor suppressor and is frequently inactivated in lung cancer cells. In the present study, we used an LRP1B minireceptor (mLRP1B4), which mimics the function and trafficking of LRP1B, to explore the roles of LRP1B on the plasminogen activation system. We found that mLRP1B4 and urokinase plasminogen activator receptor (uPAR) form immunoprecipitable complexes on the cell surface in the presence of complexes of uPA and its inhibitor, plasminogen activator inhibitor type-1 (PAI-1). However, compared with cells expressing the analogous LRP minireceptor (mLRP4), cells expressing mLRP1B4 display a substantially slower rate of uPA.PAI-1 complex internalization. Expression of mLRP1B4, or an mLRP4 mutant deficient in endocytosis, leads to an accumulation of uPAR at the cell surface and increased cell-associated uPA and PAI-1 when compared with cells expressing mLRP4. In addition, we found that expression of mLRP1B or the mLRP4 endocytosis mutant impairs the regeneration of unoccupied uPAR on the cell surface and that this correlates with a diminished rate of cell migration. Taken together, these results demonstrate that LRP1B can function as a negative regulator of uPAR regeneration and cell migration.
Collapse
Affiliation(s)
- Yonghe Li
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Green PS, Bu G. Genetics and molecular biology: phosphorylation of low-density lipoprotein receptor-related protein by platelet-derived growth factor. Curr Opin Lipidol 2002; 13:569-72. [PMID: 12352020 DOI: 10.1097/00041433-200210000-00013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|