101
|
Van der Horst DJ, Rodenburg KW. Lipoprotein assembly and function in an evolutionary perspective. Biomol Concepts 2015; 1:165-83. [PMID: 25961995 DOI: 10.1515/bmc.2010.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Circulatory fat transport in animals relies on members of the large lipid transfer protein (LLTP) superfamily, including mammalian apolipoprotein B (apoB) and insect apolipophorin II/I (apoLp-II/I). ApoB and apoLp-II/I, constituting the structural (non-exchangeable) basis for the assembly of various lipoproteins, acquire lipids through microsomal triglyceride-transfer protein, another LLTP family member, and bind them by means of amphipathic α-helical and β-sheet structural motifs. Comparative research reveals that LLTPs evolved from the earliest animals and highlights the structural adaptations in these lipid-binding proteins. Thus, in contrast to apoB, apoLp-II/I is cleaved post-translationally by a furin, resulting in the appearance of two non-exchangeable apolipoproteins in the single circulatory lipoprotein in insects, high-density lipophorin (HDLp). The remarkable structural similarities between mammalian and insect lipoproteins notwithstanding important functional differences relate to the mechanism of lipid delivery. Whereas in mammals, partial delipidation of apoB-containing lipoproteins eventually results in endocytic uptake of their remnants, mediated by members of the low-density lipoprotein receptor (LDLR) family, and degradation in lysosomes, insect HDLp functions as a reusable lipid shuttle capable of alternate unloading and reloading of lipid. Also, during muscular efforts (flight activity), an HDLp-based lipoprotein shuttle provides for the transport of lipid for energy generation. Although a lipophorin receptor - a homolog of LDLR - was identified that mediates endocytic uptake of HDLp during specific developmental periods, the endocytosed lipoprotein appears to be recycled in a transferrin-like manner. These data highlight that the functional adaptations in the lipoprotein lipid carriers in mammals and insects also emerge with regard to the functioning of their cognate receptors.
Collapse
|
102
|
Cholesterol overload impairing cerebellar function: The promise of natural products. Nutrition 2015; 31:621-30. [DOI: 10.1016/j.nut.2014.10.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 10/19/2014] [Accepted: 10/21/2014] [Indexed: 11/20/2022]
|
103
|
Liu JJ, Green P, John Mann J, Rapoport SI, Sublette ME. Pathways of polyunsaturated fatty acid utilization: implications for brain function in neuropsychiatric health and disease. Brain Res 2015; 1597:220-46. [PMID: 25498862 PMCID: PMC4339314 DOI: 10.1016/j.brainres.2014.11.059] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/11/2014] [Accepted: 11/27/2014] [Indexed: 12/28/2022]
Abstract
Essential polyunsaturated fatty acids (PUFAs) have profound effects on brain development and function. Abnormalities of PUFA status have been implicated in neuropsychiatric diseases such as major depression, bipolar disorder, schizophrenia, Alzheimer's disease, and attention deficit hyperactivity disorder. Pathophysiologic mechanisms could involve not only suboptimal PUFA intake, but also metabolic and genetic abnormalities, defective hepatic metabolism, and problems with diffusion and transport. This article provides an overview of physiologic factors regulating PUFA utilization, highlighting their relevance to neuropsychiatric disease.
Collapse
Affiliation(s)
- Joanne J Liu
- Department of Molecular Imaging & Neuropathology, New York State Psychiatric Institute, New York, NY, USA; New York Medical College, Valhalla, NY, USA
| | - Pnina Green
- Laboratory of Metabolic Research, Felsenstein Medical Research Center, Tel Aviv University, Petach Tikva, Israel
| | - J John Mann
- Department of Molecular Imaging & Neuropathology, New York State Psychiatric Institute, New York, NY, USA; Department of Psychiatry, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA
| | - Stanley I Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - M Elizabeth Sublette
- Department of Molecular Imaging & Neuropathology, New York State Psychiatric Institute, New York, NY, USA; Department of Psychiatry, Columbia University, New York, NY, USA.
| |
Collapse
|
104
|
Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer's diseases. Neurobiol Dis 2014; 72 Pt A:3-12. [PMID: 25173806 PMCID: PMC4253862 DOI: 10.1016/j.nbd.2014.08.025] [Citation(s) in RCA: 523] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/06/2014] [Accepted: 08/20/2014] [Indexed: 12/01/2022] Open
Abstract
Apolipoprotein (apo) E is a multifunctional protein with central roles in lipid metabolism, neurobiology, and neurodegenerative diseases. It has three major isoforms (apoE2, apoE3, and apoE4) with different effects on lipid and neuronal homeostasis. A major function of apoE is to mediate the binding of lipoproteins or lipid complexes in the plasma or interstitial fluids to specific cell-surface receptors. These receptors internalize apoE-containing lipoprotein particles; thus, apoE participates in the distribution/redistribution of lipids among various tissues and cells of the body. In addition, intracellular apoE may modulate various cellular processes physiologically or pathophysiologically, including cytoskeletal assembly and stability, mitochondrial integrity and function, and dendritic morphology and function. Elucidation of the functional domains within this protein and of the three-dimensional structure of the major isoforms of apoE has contributed significantly to our understanding of its physiological and pathophysiological roles at a molecular level. It is likely that apoE, with its multiple cellular origins and multiple structural and biophysical properties, is involved widely in processes of lipid metabolism and neurobiology, possibly encompassing a variety of disorders of neuronal repair, remodeling, and degeneration by interacting with different factors through various pathways.
Collapse
Affiliation(s)
- Yadong Huang
- Gladstone Institute of Neurological Disease, University of California, San Francisco 94158, USA; Gladstone Institute of Cardiovascular Disease, University of California, San Francisco 94158, USA; Department of Neurology, University of California, San Francisco 94158, USA; Department of Pathology, University of California, San Francisco 94158, USA.
| | - Robert W Mahley
- Gladstone Institute of Neurological Disease, University of California, San Francisco 94158, USA; Gladstone Institute of Cardiovascular Disease, University of California, San Francisco 94158, USA; Department of Pathology, University of California, San Francisco 94158, USA; Department of Medicine, University of California, San Francisco 94158, USA
| |
Collapse
|
105
|
Wasser CR, Masiulis I, Durakoglugil MS, Lane-Donovan C, Xian X, Beffert U, Agarwala A, Hammer RE, Herz J. Differential splicing and glycosylation of Apoer2 alters synaptic plasticity and fear learning. Sci Signal 2014; 7:ra113. [PMID: 25429077 DOI: 10.1126/scisignal.2005438] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoer2 is an essential receptor in the central nervous system that binds to the apolipoprotein ApoE. Various splice variants of Apoer2 are produced. We showed that Apoer2 lacking exon 16, which encodes the O-linked sugar (OLS) domain, altered the proteolytic processing and abundance of Apoer2 in cells and synapse number and function in mice. In cultured cells expressing this splice variant, extracellular cleavage of OLS-deficient Apoer2 was reduced, consequently preventing γ-secretase-dependent release of the intracellular domain of Apoer2. Mice expressing Apoer2 lacking the OLS domain had increased Apoer2 abundance in the brain, hippocampal spine density, and glutamate receptor abundance, but decreased synaptic efficacy. Mice expressing a form of Apoer2 lacking the OLS domain and containing an alternatively spliced cytoplasmic tail region that promotes glutamate receptor signaling showed enhanced hippocampal long-term potentiation (LTP), a phenomenon associated with learning and memory. However, these mice did not display enhanced spatial learning in the Morris water maze, and cued fear conditioning was reduced. Reducing the expression of the mutant Apoer2 allele so that the abundance of the protein was similar to that of Apoer2 in wild-type mice normalized spine density, hippocampal LTP, and cued fear learning. These findings demonstrated a role for ApoE receptors as regulators of synaptic glutamate receptor activity and established differential receptor glycosylation as a potential regulator of synaptic function and memory.
Collapse
Affiliation(s)
- Catherine R Wasser
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Irene Masiulis
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Murat S Durakoglugil
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Courtney Lane-Donovan
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xunde Xian
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Uwe Beffert
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anandita Agarwala
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert E Hammer
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joachim Herz
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA. Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
106
|
Localization of reelin signaling pathway components in murine midbrain and striatum. Cell Tissue Res 2014; 359:393-407. [PMID: 25418135 PMCID: PMC4320311 DOI: 10.1007/s00441-014-2022-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 10/06/2014] [Indexed: 02/01/2023]
Abstract
We investigated the distribution patterns of the extracellular matrix protein Reelin and of crucial Reelin signaling components in murine midbrain and striatum. The cellular distribution of the Reelin receptors VLDLr and ApoER2, the intracellular downstream mediator Dab1, and the alternative Reelin receptor APP were analyzed at embryonic day 16, at postnatal stage 15 (P15), and in 3-month-old mice. Reelin was expressed intracellularly and extracellularly in midbrain mesencephalic dopaminergic (mDA) neurons of newborns. In the striatum, Calbindin D-28k+ neurons exhibited Reelin intracellularly at E16 and extracellularly at P15 and 3 months. ApoER2 and VLDLr were expressed in mDA neurons at E16 and P15 and in oligodendrocytes at 3 months, whereas Dab1 and APP immunoreactivity was observed in mDA at all stages analyzed. In the striatum, Calbindin D-28k+/GAD67+ inhibitory neurons expressed VLDLr, ApoER2, and Dab1 at P15, but only Dab1 at E16 and 3 months. APP was always expressed in mouse striatum in which it colocalized with Calbindin D-28k. Our data underline the importance of Reelin signalling during embryonic development and early postnatal maturation of the mesostriatal and mesocorticolimbic system, and suggest that the striatum and not the midbrain is the primary source of Reelin for midbrain neurons. The loss of ApoER2 and VLDLr expression in the mature midbrain and striatum implies that Reelin functions are restricted to migratory events and early postnatal maturation and are dispensable for the maintenance of dopaminergic neurons.
Collapse
|
107
|
Valenza M, Marullo M, Di Paolo E, Cesana E, Zuccato C, Biella G, Cattaneo E. Disruption of astrocyte-neuron cholesterol cross talk affects neuronal function in Huntington's disease. Cell Death Differ 2014; 22:690-702. [PMID: 25301063 DOI: 10.1038/cdd.2014.162] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/13/2014] [Accepted: 09/03/2014] [Indexed: 01/22/2023] Open
Abstract
In the adult brain, neurons require local cholesterol production, which is supplied by astrocytes through apoE-containing lipoproteins. In Huntington's disease (HD), such cholesterol biosynthesis in the brain is severely reduced. Here we show that this defect, occurring in astrocytes, is detrimental for HD neurons. Astrocytes bearing the huntingtin protein containing increasing CAG repeats secreted less apoE-lipoprotein-bound cholesterol in the medium. Conditioned media from HD astrocytes and lipoprotein-depleted conditioned media from wild-type (wt) astrocytes were equally detrimental in a neurite outgrowth assay and did not support synaptic activity in HD neurons, compared with conditions of cholesterol supplementation or conditioned media from wt astrocytes. Molecular perturbation of cholesterol biosynthesis and efflux in astrocytes caused similarly altered astrocyte-neuron cross talk, whereas enhancement of glial SREBP2 and ABCA1 function reversed the aspects of neuronal dysfunction in HD. These findings indicate that astrocyte-mediated cholesterol homeostasis could be a potential therapeutic target to ameliorate neuronal dysfunction in HD.
Collapse
Affiliation(s)
- M Valenza
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milano, Italy
| | - M Marullo
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milano, Italy
| | - E Di Paolo
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milano, Italy
| | - E Cesana
- Department of Biology and Biotechnology, Università degli Studi di Pavia, Pavia, Italy
| | - C Zuccato
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milano, Italy
| | - G Biella
- Department of Biology and Biotechnology, Università degli Studi di Pavia, Pavia, Italy
| | - E Cattaneo
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
108
|
Meyer RC, Giddens MM, Coleman BM, Hall RA. The protective role of prosaposin and its receptors in the nervous system. Brain Res 2014; 1585:1-12. [PMID: 25130661 DOI: 10.1016/j.brainres.2014.08.022] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 07/18/2014] [Accepted: 08/10/2014] [Indexed: 12/12/2022]
Abstract
Prosaposin (also known as SGP-1) is an intriguing multifunctional protein that plays roles both intracellularly, as a regulator of lysosomal enzyme function, and extracellularly, as a secreted factor with neuroprotective and glioprotective effects. Following secretion, prosaposin can undergo endocytosis via an interaction with the low-density lipoprotein-related receptor 1 (LRP1). The ability of secreted prosaposin to promote protective effects in the nervous system is known to involve activation of G proteins, and the orphan G protein-coupled receptors GPR37 and GPR37L1 have recently been shown to mediate signaling induced by both prosaposin and a fragment of prosaposin known as prosaptide. In this review, we describe recent advances in our understanding of prosaposin, its receptors and their importance in the nervous system.
Collapse
Affiliation(s)
- Rebecca C Meyer
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Michelle M Giddens
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Brilee M Coleman
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Randy A Hall
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, United States.
| |
Collapse
|
109
|
El-Amouri SS, Dai M, Han JF, Brady RO, Pan D. Normalization and improvement of CNS deficits in mice with Hurler syndrome after long-term peripheral delivery of BBB-targeted iduronidase. Mol Ther 2014; 22:2028-2037. [PMID: 25088464 DOI: 10.1038/mt.2014.152] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/29/2014] [Indexed: 12/11/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is a progressive lysosomal storage disorder with systemic and central nervous system (CNS) involvement due to deficiency of α-L-iduronidase (IDUA). We previously identified a receptor-binding peptide from apolipoprotein E (e) that facilitated a widespread delivery of IDUAe fusion protein into CNS. In this study, we evaluated the long-term CNS biodistribution, dose-correlation, and therapeutic benefits of IDUAe after systemic, sustained delivery via hematopoietic stem cell (HSC)-mediated gene therapy with expression restricted to erythroid/megakaryocyte lineages. Compared to the highest dosage group treated by nontargeted control IDUAc (165 U/ml), physiological levels of IDUAe in the circulation (12 U/ml) led to better CNS benefits in MPS I mice as demonstrated in glycosaminoglycan accumulation, histopathology analysis, and neurological behavior. Long-term brain metabolic correction and normalization of exploratory behavior deficits in MPS I mice were observed by peripheral enzyme therapy with physiological levels of IDUAe derived from clinically attainable levels of HSC transduction efficiency (0.1). Importantly, these levels of IDUAe proved to be more beneficial on correction of cerebrum pathology and behavioral deficits in MPS I mice than wild-type HSCs fully engrafted in MPS I chimeras. These results provide compelling evidence for CNS efficacy of IDUAe and its prospective translation to clinical application.
Collapse
Affiliation(s)
- Salim S El-Amouri
- Molecular and Cell Therapy Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mei Dai
- Molecular and Cell Therapy Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jing-Fen Han
- Molecular and Cell Therapy Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Roscoe O Brady
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Dao Pan
- Molecular and Cell Therapy Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
110
|
Mechanisms of U87 astrocytoma cell uptake and trafficking of monomeric versus protofibril Alzheimer's disease amyloid-β proteins. PLoS One 2014; 9:e99939. [PMID: 24941200 PMCID: PMC4062444 DOI: 10.1371/journal.pone.0099939] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 05/20/2014] [Indexed: 01/02/2023] Open
Abstract
A significant hallmark of Alzheimer’s disease is the formation of senile plaques in the brain due to the unbalanced levels of amyloid-beta (Aβ). However, although how Aβ is produced from amyloid precursor proteins is well understood, little is known regarding the clearance and metabolism of various Aβ aggregates from the brain. Similarly, little is known regarding how astrocytes internalize and degrade Aβ, although astrocytes are known to play an important role in plaque maintenance and Aβ clearance. The objective of this study is to investigate the cellular mechanisms that mediate the internalization of soluble monomeric versus oligomeric Aβ by astrocytes. We used a combination of laser confocal microscopy and genetic and pharmacological experiments to dissect the internalization of sAβ42 and oAβ42 and their postendocytic transport by U87 human brain astrocytoma cell line. Both Aβ42 species were internalized by U87 cells through fluid phase macropinocytosis, which required dynamin 2. Depleting LDL receptor-related protein 1 (LRP1) decreased sAβ42 uptake more significantly than that of oAβ42. We finally show that both Aβ42 species were rapidly transported to lysosomes through an endolytic pathway and subjected to proteolysis after internalization, which had no significant toxic effects to the U87 cells under relatively low concentrations. We propose that macropinocytic sAβ42 and oAβ42 uptake and their subsequent proteolytic degradation in astroglial cells is a significant mechanism underlying Aβ clearance from the extracellular milieu. Understanding the molecular events involved in astrocytic Aβ internalization may identify potential therapeutic targets for Alzheimer’s disease.
Collapse
|
111
|
Fang Y, Feng M, Han B, Lu X, Ramadan H, Li J. In-depth proteomics characterization of embryogenesis of the honey bee worker (Apis mellifera ligustica). Mol Cell Proteomics 2014; 13:2306-20. [PMID: 24895377 DOI: 10.1074/mcp.m114.037846] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Identifying proteome changes of honey bee embryogenesis is of prime importance for unraveling the molecular mechanisms that they underlie. However, many proteomic changes during the embryonic period are not well characterized. We analyzed the proteomic alterations over the complete time course of honey bee worker embryogenesis at 24, 48, and 72 h of age, using mass spectrometry-based proteomics, label-free quantitation, and bioinformatics. Of the 1460 proteins identified the embryo of all three ages, the core proteome (proteins shared by the embryos of all three ages, accounting for 40%) was mainly involved in protein synthesis, metabolic energy, development, and molecular transporter, which indicates their centrality in driving embryogenesis. However, embryos at different developmental stages have their own specific proteome and pathway signatures to coordinate and modulate developmental events. The young embryos (<24 h) stronger expression of proteins related to nutrition storage and nucleic acid metabolism may correlate with the cell proliferation occurring at this stage. The middle aged embryos (24-48 h) enhanced expression of proteins associated with cell cycle control, transporters, antioxidant activity, and the cytoskeleton suggest their roles to support rudimentary organogenesis. Among these proteins, the biological pathways of aminoacyl-tRNA biosynthesis, β-alanine metabolism, and protein export are intensively activated in the embryos of middle age. The old embryos (48-72 h) elevated expression of proteins implicated in fatty acid metabolism and morphogenesis indicate their functionality for the formation and development of organs and dorsal closure, in which the biological pathways of fatty acid metabolism and RNA transport are highly activated. These findings add novel understanding to the molecular details of honey bee embryogenesis, in which the programmed activation of the proteome matches with the physiological transition observed during embryogenesis. The identified biological pathways and key node proteins allow for further functional analysis and genetic manipulation for both the honey bee embryos and other eusocial insects.
Collapse
Affiliation(s)
- Yu Fang
- From the ‡Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mao Feng
- From the ‡Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Bin Han
- From the ‡Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoshan Lu
- From the ‡Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Haitham Ramadan
- From the ‡Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianke Li
- From the ‡Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
112
|
Kanekiyo T, Bu G. The low-density lipoprotein receptor-related protein 1 and amyloid-β clearance in Alzheimer's disease. Front Aging Neurosci 2014; 6:93. [PMID: 24904407 PMCID: PMC4033011 DOI: 10.3389/fnagi.2014.00093] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/28/2014] [Indexed: 11/24/2022] Open
Abstract
Accumulation and aggregation of amyloid-β (Aβ) peptides in the brain trigger the development of progressive neurodegeneration and dementia associated with Alzheimer’s disease (AD). Perturbation in Aβ clearance, rather than Aβ production, is likely the cause of sporadic, late-onset AD, which accounts for the majority of AD cases. Since cellular uptake and subsequent degradation constitute a major Aβ clearance pathway, the receptor-mediated endocytosis of Aβ has been intensely investigated. Among Aβ receptors, the low-density lipoprotein receptor-related protein 1 (LRP1) is one of the most studied receptors. LRP1 is a large endocytic receptor for more than 40 ligands, including apolipoprotein E, α2-macroglobulin and Aβ. Emerging in vitro and in vivo evidence demonstrates that LRP1 is critically involved in brain Aβ clearance. LRP1 is highly expressed in a variety of cell types in the brain including neurons, vascular cells and glial cells, where LRP1 functions to maintain brain homeostasis and control Aβ metabolism. LRP1-mediated endocytosis regulates cellular Aβ uptake by binding to Aβ either directly or indirectly through its co-receptors or ligands. Furthermore, LRP1 regulates several signaling pathways, which also likely influences Aβ endocytic pathways. In this review, we discuss how LRP1 regulates the brain Aβ clearance and how this unique endocytic receptor participates in AD pathogenesis. Understanding of the mechanisms underlying LRP1-mediated Aβ clearance should enable the rational design of novel diagnostic and therapeutic strategies for AD.
Collapse
Affiliation(s)
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville FL, USA
| |
Collapse
|
113
|
Protection of SK-N-MC cells against β-amyloid peptide-induced degeneration using neuron growth factor-loaded liposomes with surface lactoferrin. Biomaterials 2014; 35:5954-64. [PMID: 24746790 DOI: 10.1016/j.biomaterials.2014.03.082] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/28/2014] [Indexed: 01/26/2023]
Abstract
A liposomal system with surface lactoferrin (Lf) was developed for delivering neuron growth factor (NGF) across the blood-brain barrier (BBB) and improving the viability of neuron-like SK-N-MC cells with deposited β-amyloid peptide (Aβ). The Lf-grafted liposomes carrying NGF (Lf/NGF-liposomes) were applied to a monolayer of human brain-microvascular endothelial cells (HBMECs) regulated by human astrocytes (HAs) and to fibrillar Aβ1-42-insulted SK-N-MC cells. An increase in cholesterol mole percentage enhanced the particle size, absolute value of zeta potential, and physical stability, however, reduced the entrapment efficiency and release rate of NGF. In addition, an increase in Lf concentration increased the particle size, surface nitrogen percentage, NGF permeability across the BBB, and viability of HBMECs, HAs, and SK-N-MC cells, however, decreased the absolute value of zeta potential, surface phosphorus percentage, and loading efficiency of Lf. After treating with Lf/NGF-liposomes, a higher Aβ concentration yielded a lower survival of SK-N-MC cells. The current Lf/NGF-liposomes are efficacious drug carriers to target the BBB and inhibit the Aβ-induced neurotoxicity as potential pharmacotherapy for Alzheimer's disease.
Collapse
|
114
|
Liu Q, Zhang J. Lipid metabolism in Alzheimer's disease. Neurosci Bull 2014; 30:331-45. [PMID: 24733655 DOI: 10.1007/s12264-013-1410-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 02/25/2014] [Indexed: 12/14/2022] Open
Abstract
Lipids play crucial roles in cell signaling and various physiological processes, especially in the brain. Impaired lipid metabolism in the brain has been implicated in neurodegenerative diseases, such as Alzheimer's disease (AD), and other central nervous system insults. The brain contains thousands of lipid species, but the complex lipid compositional diversity and the function of each of lipid species are currently poorly understood. This review integrates current knowledge about major lipid changes with the molecular mechanisms that underlie AD pathogenesis.
Collapse
Affiliation(s)
- Qiang Liu
- CAS Key Laboratory of Brain Function and Disease and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China,
| | | |
Collapse
|
115
|
Förster E. Reelin, neuronal polarity and process orientation of cortical neurons. Neuroscience 2014; 269:102-11. [PMID: 24657457 DOI: 10.1016/j.neuroscience.2014.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/01/2014] [Accepted: 03/04/2014] [Indexed: 12/22/2022]
Abstract
Deficient reelin signaling leads to characteristic layering malformations in the cerebral cortex and causes polarity defects of cortical neurons. Since the discovery of reelin much has been learned about the molecular mechanisms that underlie the characteristic defects of layering defects in the reeler mutant. More recent studies provided insights in the crosstalk between reelin signaling and molecular pathways that control polarity development of radially migrating neurons. The present review summarizes and discusses recent findings on the role of reelin in modulating polarization and process orientation of neurons in the neocortex and hippocampus.
Collapse
Affiliation(s)
- E Förster
- Institute of Neuroanatomy, University Medical Center Hamburg Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
116
|
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
|
117
|
Cheng Y, Morshed RA, Auffinger B, Tobias AL, Lesniak MS. Multifunctional nanoparticles for brain tumor imaging and therapy. Adv Drug Deliv Rev 2014; 66:42-57. [PMID: 24060923 PMCID: PMC3948347 DOI: 10.1016/j.addr.2013.09.006] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/28/2013] [Accepted: 09/13/2013] [Indexed: 12/16/2022]
Abstract
Brain tumors are a diverse group of neoplasms that often carry a poor prognosis for patients. Despite tremendous efforts to develop diagnostic tools and therapeutic avenues, the treatment of brain tumors remains a formidable challenge in the field of neuro-oncology. Physiological barriers including the blood-brain barrier result in insufficient accumulation of therapeutic agents at the site of a tumor, preventing adequate destruction of malignant cells. Furthermore, there is a need for improvements in brain tumor imaging to allow for better characterization and delineation of tumors, visualization of malignant tissue during surgery, and tracking of response to chemotherapy and radiotherapy. Multifunctional nanoparticles offer the potential to improve upon many of these issues and may lead to breakthroughs in brain tumor management. In this review, we discuss the diagnostic and therapeutic applications of nanoparticles for brain tumors with an emphasis on innovative approaches in tumor targeting, tumor imaging, and therapeutic agent delivery. Clinically feasible nanoparticle administration strategies for brain tumor patients are also examined. Furthermore, we address the barriers towards clinical implementation of multifunctional nanoparticles in the context of brain tumor management.
Collapse
Affiliation(s)
- Yu Cheng
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Ramin A Morshed
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Brenda Auffinger
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Alex L Tobias
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Maciej S Lesniak
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
118
|
Jiang S, Li Y, Zhang X, Bu G, Xu H, Zhang YW. Trafficking regulation of proteins in Alzheimer's disease. Mol Neurodegener 2014; 9:6. [PMID: 24410826 PMCID: PMC3891995 DOI: 10.1186/1750-1326-9-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 12/15/2013] [Indexed: 12/12/2022] Open
Abstract
The β-amyloid (Aβ) peptide has been postulated to be a key determinant in the pathogenesis of Alzheimer’s disease (AD). Aβ is produced through sequential cleavage of the β-amyloid precursor protein (APP) by β- and γ-secretases. APP and relevant secretases are transmembrane proteins and traffic through the secretory pathway in a highly regulated fashion. Perturbation of their intracellular trafficking may affect dynamic interactions among these proteins, thus altering Aβ generation and accelerating disease pathogenesis. Herein, we review recent progress elucidating the regulation of intracellular trafficking of these essential protein components in AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Yun-wu Zhang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
119
|
|
120
|
Lee JH, Kim BK, Seo YI, Choi JH, Kang SW, Kang CK, Park WG, Kim HW. Four cDNAs encoding lipoprotein receptors from shrimp (Pandalopsis japonica): structural characterization and expression analysis during maturation. Comp Biochem Physiol B Biochem Mol Biol 2013; 169:51-62. [PMID: 24389120 DOI: 10.1016/j.cbpb.2013.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
Abstract
As in all other oviparous animals, lipoprotein receptors play a critical role in lipid metabolism and reproduction in decapod crustaceans. Four full-length cDNAs encoding lipoprotein receptors (Paj-VgR, Paj-LpR1, Paj-LpR2A, and Paj-LpR2B) were identified from Pandalopsis japonica through a combination of EST screening and PCR-based cloning. Paj-LpR1 appears to be the first crustacean ortholog of insect lipophorin receptors, and its two paralogs, Paj-LpR2A and Paj-LpR2B, exhibited similar structural characteristics. Several transcriptional isoforms were also identified for all three Paj-LpRs. Each expression pattern was unique, suggesting different physiological roles for these proteins. Paj-VgR is an ortholog of vitellogenin (Vg) receptors from other decapod crustaceans. A phylogenetic analysis of lipoproteins and their receptors suggested that the nomenclature of Vgs from decapod crustaceans may need to be changed. A PCR-based transcriptional analysis showed that Paj-VgR and Paj-LpR2B are expressed almost exclusively in the ovary, whereas Paj-LpR1 and Paj-LpR2A are expressed in multiple tissues. The various transcriptional isoforms of the three Paj-LpRs exhibited unique tissue distribution profiles. A transcriptional analysis of each receptor using tissues with different GSI values showed that the change in transcription of Paj-VgRs, Paj-LpR2A and Paj-LpR1 was not as significant as that of Vgs during maturation. However, the transcriptional levels of Paj-LpR2B decreased in ovary at maturation, suggesting that their transcriptional regulation is involved in reproduction.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Interdisciplinary program of Biomedical Engineering, Pukyong National University, Busan, 608-737, South Korea
| | - Bo Kwang Kim
- Interdisciplinary program of Biomedical Engineering, Pukyong National University, Busan, 608-737, South Korea
| | - Young-Il Seo
- Fisheries Resources Research Division, National Fisheries Research and Development Institute, Busan, 619-902, South Korea
| | - Jung Hwa Choi
- Fisheries Resources Research Division, National Fisheries Research and Development Institute, Busan, 619-902, South Korea
| | - Seung-Wan Kang
- Gyeongsangnam-do Fisheries Resources Research Institute, South Korea
| | - Chang-Keun Kang
- POSTECH Ocean Science and Technology Institute, Pohang University of Science and Technology (POSTECH), Pohang 790-784, South Korea
| | - Won-gyu Park
- Department of Marine Biology, Pukyong National University, Busan 608-737, South Korea
| | - Hyun-Woo Kim
- Department of Marine Biology, Pukyong National University, Busan 608-737, South Korea; Interdisciplinary program of Biomedical Engineering, Pukyong National University, Busan, 608-737, South Korea.
| |
Collapse
|
121
|
Morshed RA, Cheng Y, Auffinger B, Wegscheid ML, Lesniak MS. The potential of polymeric micelles in the context of glioblastoma therapy. Front Pharmacol 2013; 4:157. [PMID: 24416018 PMCID: PMC3874582 DOI: 10.3389/fphar.2013.00157] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/29/2013] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM), a type of malignant glioma, is the most common form of brain cancer found in adults. The current standard of care for GBM involves adjuvant temozolomide-based chemotherapy in conjunction with radiotherapy, yet patients still suffer from poor outcomes with a median survival of 14.6 months. Many novel therapeutic agents that are toxic to GBM cells in vitro cannot sufficiently accumulate at the site of an intracranial tumor after systemic administration. Thus, new delivery strategies must be developed to allow for adequate intratumoral accumulation of such therapeutic agents. Polymeric micelles offer the potential to improve delivery to brain tumors as they have demonstrated the capacity to be effective carriers of chemotherapy drugs, genes, and proteins in various preclinical GBM studies. In addition to this, targeting moieties and trigger-dependent release mechanisms incorporated into the design of these particles can promote more specific delivery of a therapeutic agent to a tumor site. However, despite these advantages, there are currently no micelle formulations targeting brain cancer in clinical trials. Here, we highlight key aspects of the design of polymeric micelles as therapeutic delivery systems with a review of their clinical applications in several non-brain tumor cancer types. We also discuss their potential to serve as nanocarriers targeting GBM, the major barriers preventing their clinical implementation in this disease context, as well as current approaches to overcome these limitations.
Collapse
Affiliation(s)
- Ramin A Morshed
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine Chicago, IL, USA
| | - Yu Cheng
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine Chicago, IL, USA
| | - Brenda Auffinger
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine Chicago, IL, USA
| | - Michelle L Wegscheid
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine Chicago, IL, USA
| | - Maciej S Lesniak
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine Chicago, IL, USA
| |
Collapse
|
122
|
Molecular patterning of the mammalian dentition. Semin Cell Dev Biol 2013; 25-26:61-70. [PMID: 24355560 DOI: 10.1016/j.semcdb.2013.12.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 11/20/2013] [Accepted: 12/09/2013] [Indexed: 01/15/2023]
Abstract
Four conserved signaling pathways, including the bone morphogenetic proteins (Bmp), fibroblast growth factors (Fgf), sonic hedgehog (Shh), and wingless-related (Wnt) pathways, are each repeatedly used throughout tooth development. Inactivation of any of these resulted in early tooth developmental arrest in mice. The mutations identified thus far in human patients with tooth agenesis also affect these pathways. Recent studies show that these signaling pathways interact through positive and negative feedback loops to regulate not only morphogenesis of individual teeth but also tooth number, shape, and spatial pattern. Increased activity of each of the Fgf, Shh, and canonical Wnt signaling pathways revitalizes development of the physiologically arrested mouse diastemal tooth germs whereas constitutive activation of canonical Wnt signaling in the dental epithelium is able to induce supernumerary tooth formation even in the absence of Msx1 and Pax9, two transcription factors required for normal tooth development beyond the early bud stage. Bmp4 and Msx1 act in a positive feedback loop to drive sequential tooth formation whereas the Osr2 transcription factor restricts Msx1-mediated expansion of the mesenchymal odontogenic field along both the buccolingual and anteroposterior axes to pattern mouse molar teeth in a single row. Moreover, the ectodermal-specific ectodysplasin (EDA) signaling pathway controls tooth number and tooth shape through regulation of Fgf20 expression in the dental epithelium, whereas Shh suppresses Wnt signaling through a negative feedback loop to regulate spatial patterning of teeth. In this article, we attempt to integrate these exciting findings in the understanding of the molecular networks regulating tooth development and patterning.
Collapse
|
123
|
Alvarez JI, Katayama T, Prat A. Glial influence on the blood brain barrier. Glia 2013; 61:1939-58. [PMID: 24123158 PMCID: PMC4068281 DOI: 10.1002/glia.22575] [Citation(s) in RCA: 407] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/13/2013] [Accepted: 08/19/2013] [Indexed: 12/14/2022]
Abstract
The Blood Brain Barrier (BBB) is a specialized vascular structure tightly regulating central nervous system (CNS) homeostasis. Endothelial cells are the central component of the BBB and control of their barrier phenotype resides on astrocytes and pericytes. Interactions between these cells and the endothelium promote and maintain many of the physiological and metabolic characteristics that are unique to the BBB. In this review we describe recent findings related to the involvement of astroglial cells, including radial glial cells, in the induction of barrier properties during embryogenesis and adulthood. In addition, we describe changes that occur in astrocytes and endothelial cells during injury and inflammation with a particular emphasis on alterations of the BBB phenotype. GLIA 2013;61:1939–1958
Collapse
Affiliation(s)
- Jorge Ivan Alvarez
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | | | | |
Collapse
|
124
|
Taibi AV, Lighthouse JK, Grady RC, Shroyer KR, Holdener BC. Development of a conditional Mesd (mesoderm development) allele for functional analysis of the low-density lipoprotein receptor-related family in defined tissues. PLoS One 2013; 8:e75782. [PMID: 24124512 PMCID: PMC3790828 DOI: 10.1371/journal.pone.0075782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/21/2013] [Indexed: 11/19/2022] Open
Abstract
The Low-density lipoprotein receptor-Related Protein (LRP) family members are essential for diverse processes ranging from the regulation of gastrulation to the modulation of lipid homeostasis. Receptors in this family bind and internalize a diverse array of ligands in the extracellular matrix (ECM). As a consequence, LRPs regulate a wide variety of cellular functions including, but not limited to lipid metabolism, membrane composition, cell motility, and cell signaling. Not surprisingly, mutations in single human LRPs are associated with defects in cholesterol metabolism and development of atherosclerosis, abnormalities in bone density, or aberrant eye vasculature, and may be a contributing factor in development of Alzheimer’s disease. Often, members of this diverse family of receptors perform overlapping roles in the same tissues, complicating the analysis of their function through conventional targeted mutagenesis. Here, we describe development of a mouse Mesd (Mesoderm Development) conditional knockout allele, and demonstrate that ubiquitous deletion of Mesd using Cre-recombinase blocks gastrulation, as observed in the traditional knockout and albino-deletion phenotypes. This conditional allele will serve as an excellent tool for future characterization of the cumulative contribution of LRP members in defined tissues.
Collapse
Affiliation(s)
- Andrew V. Taibi
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Janet K. Lighthouse
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Richard C. Grady
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Kenneth R. Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, New York, United States of America
| | - Bernadette C. Holdener
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
125
|
Luo W, Ito Y, Mizuta H, Massaki K, Hiramatsu N, Todo T, Reading BJ, Sullivan CV, Hara A. Molecular cloning and partial characterization of an ovarian receptor with seven ligand binding repeats, an orthologue of low-density lipoprotein receptor, in the cutthroat trout (Oncorhynchus clarki). Comp Biochem Physiol A Mol Integr Physiol 2013; 166:263-71. [DOI: 10.1016/j.cbpa.2013.06.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/23/2013] [Accepted: 06/26/2013] [Indexed: 11/25/2022]
|
126
|
Sharaf A, Bock HH, Spittau B, Bouché E, Krieglstein K. ApoER2 and VLDLr are required for mediating reelin signalling pathway for normal migration and positioning of mesencephalic dopaminergic neurons. PLoS One 2013; 8:e71091. [PMID: 23976984 PMCID: PMC3745466 DOI: 10.1371/journal.pone.0071091] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 06/24/2013] [Indexed: 12/19/2022] Open
Abstract
The migration of mesencephalic dopaminergic (mDA) neurons from the subventricular zone to their final positions in the substantia nigra compacta (SNc), ventral tegmental area (VTA), and retrorubral field (RRF) is controlled by signalling from neurotrophic factors, cell adhesion molecules (CAMs) and extracellular matrix molecules (ECM). Reelin and the cytoplasmic adaptor protein Disabled-1 (Dab1) have been shown to play important roles in the migration and positioning of mDA neurons. Mice lacking Reelin and Dab1 both display phenotypes characterised by the failure of nigral mDA neurons to migrate properly. ApoER2 and VLDLr are receptors for Reelin signalling and are therefore part of the same signal transduction pathway as Dab1. Here we describe the roles of ApoER2 and VLDLr in the proper migration and positioning of mDA neurons in mice. Our results demonstrate that VLDLr- and ApoER2-mutant mice have both a reduction in and abnormal positioning of mDA neurons. This phenotype was more pronounced in VLDLr-mutant mice. Moreover, we provide evidence that ApoER2/VLDLr double-knockout mice show a phenotype comparable with the phenotypes observed for Reelin- and Dab1- mutant mice. Taken together, our results demonstrate that the Reelin receptors ApoER2 and VLDLr play essential roles in Reelin-mediated migration and positioning of mDA neurons.
Collapse
Affiliation(s)
- Ahmed Sharaf
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs-Universität, Freiburg, Germany
| | - Hans H. Bock
- Center for Neuroscience, Albert-Ludwigs-Universität, Freiburg, Germany
- Department of Medicine II, Albert-Ludwigs-Universität, Freiburg, Germany
| | - Björn Spittau
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs-Universität, Freiburg, Germany
| | - Elisabeth Bouché
- Center for Neuroscience, Albert-Ludwigs-Universität, Freiburg, Germany
| | - Kerstin Krieglstein
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs-Universität, Freiburg, Germany
- * E-mail:
| |
Collapse
|
127
|
Granic A, Potter H. Mitotic spindle defects and chromosome mis-segregation induced by LDL/cholesterol-implications for Niemann-Pick C1, Alzheimer's disease, and atherosclerosis. PLoS One 2013; 8:e60718. [PMID: 23593294 PMCID: PMC3625184 DOI: 10.1371/journal.pone.0060718] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 03/01/2013] [Indexed: 12/17/2022] Open
Abstract
Elevated low-density lipoprotein (LDL)-cholesterol is a risk factor for both Alzheimer's disease (AD) and Atherosclerosis (CVD), suggesting a common lipid-sensitive step in their pathogenesis. Previous results show that AD and CVD also share a cell cycle defect: chromosome instability and up to 30% aneuploidy-in neurons and other cells in AD and in smooth muscle cells in atherosclerotic plaques in CVD. Indeed, specific degeneration of aneuploid neurons accounts for 90% of neuronal loss in AD brain, indicating that aneuploidy underlies AD neurodegeneration. Cell/mouse models of AD develop similar aneuploidy through amyloid-beta (Aß) inhibition of specific microtubule motors and consequent disruption of mitotic spindles. Here we tested the hypothesis that, like upregulated Aß, elevated LDL/cholesterol and altered intracellular cholesterol homeostasis also causes chromosomal instability. Specifically we found that: 1) high dietary cholesterol induces aneuploidy in mice, satisfying the hypothesis' first prediction, 2) Niemann-Pick C1 patients accumulate aneuploid fibroblasts, neurons, and glia, demonstrating a similar aneugenic effect of intracellular cholesterol accumulation in humans 3) oxidized LDL, LDL, and cholesterol, but not high-density lipoprotein (HDL), induce chromosome mis-segregation and aneuploidy in cultured cells, including neuronal precursors, indicating that LDL/cholesterol directly affects the cell cycle, 4) LDL-induced aneuploidy requires the LDL receptor, but not Aß, showing that LDL works differently than Aß, with the same end result, 5) cholesterol treatment disrupts the structure of the mitotic spindle, providing a cell biological mechanism for its aneugenic activity, and 6) ethanol or calcium chelation attenuates lipoprotein-induced chromosome mis-segregation, providing molecular insights into cholesterol's aneugenic mechanism, specifically through its rigidifying effect on the cell membrane, and potentially explaining why ethanol consumption reduces the risk of developing atherosclerosis or AD. These results suggest a novel, cell cycle mechanism by which aberrant cholesterol homeostasis promotes neurodegeneration and atherosclerosis by disrupting chromosome segregation and potentially other aspects of microtubule physiology.
Collapse
Affiliation(s)
- Antoneta Granic
- Department of Neurology and Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Institute for Ageing and Health, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Huntington Potter
- Department of Neurology and Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
128
|
Ahn Y, Sims C, Logue JM, Weatherbee SD, Krumlauf R. Lrp4 and Wise interplay controls the formation and patterning of mammary and other skin appendage placodes by modulating Wnt signaling. Development 2013; 140:583-93. [PMID: 23293290 DOI: 10.1242/dev.085118] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The future site of skin appendage development is marked by a placode during embryogenesis. Although Wnt/β-catenin signaling is known to be essential for skin appendage development, it is unclear which cellular processes are controlled by the signaling and how the precise level of the signaling activity is achieved during placode formation. We have investigated roles for Lrp4 and its potential ligand Wise (Sostdc1) in mammary and other skin appendage placodes. Lrp4 mutant mice displayed a delay in placode initiation and changes in distribution and number of mammary precursor cells leading to abnormal morphology, number and position of mammary placodes. These Lrp4 mammary defects, as well as limb defects, were associated with elevated Wnt/β-catenin signaling and were rescued by reducing the dose of the Wnt co-receptor genes Lrp5 and Lrp6, or by inactivating the gene encoding β-catenin. Wise-null mice phenocopied a subset of the Lrp4 mammary defects and Wise overexpression reduced the number of mammary precursor cells. Genetic epistasis analyses suggest that Wise requires Lrp4 to exert its function and that, together, they have a role in limiting mammary fate, but Lrp4 has an early Wise-independent role in facilitating placode formation. Lrp4 and Wise mutants also share defects in vibrissa and hair follicle development, suggesting that the roles played by Lrp4 and Wise are common to skin appendages. Our study presents genetic evidence for interplay between Lrp4 and Wise in inhibiting Wnt/β-catenin signaling and provides an insight into how modulation of Wnt/β-catenin signaling controls cellular processes important for skin placode formation.
Collapse
Affiliation(s)
- Youngwook Ahn
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | | | | | | |
Collapse
|
129
|
Cortes VA, Busso D, Mardones P, Maiz A, Arteaga A, Nervi F, Rigotti A. Retracted: Advances in the physiological and pathological implications of cholesterol. Biol Rev Camb Philos Soc 2013; 88:825-43. [DOI: 10.1111/brv.12025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 01/22/2013] [Accepted: 01/25/2013] [Indexed: 01/07/2023]
Affiliation(s)
- Victor A. Cortes
- Department of Nutrition Diabetes and Metabolism; School of Medicine; Faculty of Medicine; Pontificia Universidad Catolica de Chile; Marcoleta 367 Edifico de Gastroenterologia 4 piso Santiago Chile
| | - Dolores Busso
- Department of Nutrition Diabetes and Metabolism; School of Medicine; Faculty of Medicine; Pontificia Universidad Catolica de Chile; Marcoleta 367 Edifico de Gastroenterologia 4 piso Santiago Chile
| | - Pablo Mardones
- Department of Nutrition Diabetes and Metabolism; School of Medicine; Faculty of Medicine; Pontificia Universidad Catolica de Chile; Marcoleta 367 Edifico de Gastroenterologia 4 piso Santiago Chile
| | - Alberto Maiz
- Department of Nutrition Diabetes and Metabolism; School of Medicine; Faculty of Medicine; Pontificia Universidad Catolica de Chile; Marcoleta 367 Edifico de Gastroenterologia 4 piso Santiago Chile
| | - Antonio Arteaga
- Department of Nutrition Diabetes and Metabolism; School of Medicine; Faculty of Medicine; Pontificia Universidad Catolica de Chile; Marcoleta 367 Edifico de Gastroenterologia 4 piso Santiago Chile
| | - Flavio Nervi
- Department of Gastroenterology; School of Medicine; Faculty of Medicine; Pontificia Universidad Catolica de Chile; Santiago Chile
| | - Attilio Rigotti
- Department of Nutrition Diabetes and Metabolism; School of Medicine; Faculty of Medicine; Pontificia Universidad Catolica de Chile; Marcoleta 367 Edifico de Gastroenterologia 4 piso Santiago Chile
| |
Collapse
|
130
|
Apolipoprotein e sets the stage: response to injury triggers neuropathology. Neuron 2013; 76:871-85. [PMID: 23217737 DOI: 10.1016/j.neuron.2012.11.020] [Citation(s) in RCA: 271] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2012] [Indexed: 01/04/2023]
Abstract
Apolipoprotein (apo) E4 is the major genetic risk factor for Alzheimer's disease and is associated with poor clinical outcome following traumatic brain injury and other neuropathological disorders. Protein instability and an isoform-specific apoE property called domain interaction are responsible for these neuropathological effects. ApoE4 is the most neurotoxic isoform and can induce neuropathology through various cellular pathways. Neuronal damage or stress induces apoE synthesis as part of the repair response; however, when apoE4 is expressed in neurons, its unique conformation makes it susceptible to proteolysis, resulting in the generation of neurotoxic fragments. These fragments cause pathological mitochondrial dysfunction and cytoskeletal alterations. Here, we review data supporting the hypothesis that apoE4 (> apoE3 > apoE2) has direct neurotoxic effects and highlight studies showing that blocking domain interaction reverses these detrimental effects.
Collapse
|
131
|
Sagare AP, Bell RD, Zlokovic BV. Neurovascular defects and faulty amyloid-β vascular clearance in Alzheimer's disease. J Alzheimers Dis 2013; 33 Suppl 1:S87-100. [PMID: 22751174 PMCID: PMC4416477 DOI: 10.3233/jad-2012-129037] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The evidence that neurovascular dysfunction is an integral part of Alzheimer's disease (AD) pathogenesis has continued to emerge in the last decade. Changes in the brain vasculature have been shown to contribute to the onset and progression of the pathological processes associated with AD, such as microvascular reductions, blood brain barrier (BBB) breakdown, and faulty clearance of amyloid β-peptide (Aβ) from the brain. Herein, we review the role of the neurovascular unit and molecular mechanisms in cerebral vascular cells behind the pathogenesis of AD. In particular, we focus on molecular pathways within cerebral vascular cells and the systemic circulation that contribute to BBB dysfunction, brain hypoperfusion, and impaired clearance of Aβ from the brain. We aim to provide a summary of recent research findings implicated in neurovascular defects and faulty Aβ vascular clearance contributing to AD pathogenesis.
Collapse
Affiliation(s)
- Abhay P. Sagare
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert D. Bell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
132
|
Ruzali WAW, Kehoe PG, Love S. LRP1 expression in cerebral cortex, choroid plexus and meningeal blood vessels: relationship to cerebral amyloid angiopathy and APOE status. Neurosci Lett 2012; 525:123-8. [PMID: 22967844 DOI: 10.1016/j.neulet.2012.07.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/28/2012] [Accepted: 07/30/2012] [Indexed: 01/20/2023]
Abstract
APOE genotype is a risk factor for Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). The risk and severity of CAA increase with possession of APOE ε4, whereas APOE ε2 increases the risk of vessel rupture. Uptake of Aβ by cerebrovascular smooth muscle cells (CVSMCs) is mediated by low-density lipoprotein receptor-related protein-1 (LRP1). To determine whether APOE influences CAA by altering LRP1 expression, particularly by CVSMCs, we analysed APOE genotype, CAA severity, and LRP1 levels in post-mortem cerebral cortex, choroid plexus and meningeal vessels. LRP1 mRNA and protein were not related to CAA severity and presence. LRP1 mRNA was increased in meningeal vessels, but not cortex or choroid plexus, in AD and in association with APOE ε4, and was decreased in association with APOE ε3. In brains with CAA, APOE ε2 was associated with decreased LRP1 protein in meningeal vessels, and ε3 with increased LRP1 in choroid plexus. These findings suggest that APOE may influence the severity of CAA through altered expression of LRP1.
Collapse
Affiliation(s)
- Wan Adriyani W Ruzali
- Dementia Research Group, Institute of Clinical Neurosciences, Frenchay Hospital, Bristol, BS16 1LE, UK
| | | | | |
Collapse
|
133
|
Krol S, Macrez R, Docagne F, Defer G, Laurent S, Rahman M, Hajipour MJ, Kehoe PG, Mahmoudi M. Therapeutic Benefits from Nanoparticles: The Potential Significance of Nanoscience in Diseases with Compromise to the Blood Brain Barrier. Chem Rev 2012; 113:1877-903. [DOI: 10.1021/cr200472g] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Silke Krol
- Fondazione IRCCS Institute of Neurology “Carlo Besta”, Milan, Italy
| | - Richard Macrez
- Inserm U919, University Caen Basse Normandie, Serine Proteases and Pathophysiology of the Neurovascular Unit, GIP CYCERON, F-14074 Caen, France
- Department of Neurology, University Hospital of Caen, Caen, France
| | - Fabian Docagne
- Inserm U919, University Caen Basse Normandie, Serine Proteases and Pathophysiology of the Neurovascular Unit, GIP CYCERON, F-14074 Caen, France
| | - Gilles Defer
- Inserm U919, University Caen Basse Normandie, Serine Proteases and Pathophysiology of the Neurovascular Unit, GIP CYCERON, F-14074 Caen, France
- Department of Neurology, University Hospital of Caen, Caen, France
| | - Sophie Laurent
- Department of General, Organic, and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau, 19, B-7000 Mons, Belgium
| | - Masoud Rahman
- Laboratory of NanoBio Interactions , Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad J. Hajipour
- Laboratory of NanoBio Interactions , Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Patrick G. Kehoe
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, John James Laboratories, Frenchay Hospital, Bristol, U.K
| | - Morteza Mahmoudi
- Laboratory of NanoBio Interactions , Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Current address: School of Chemical Sciences, University of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
| |
Collapse
|
134
|
Cholesterol: its regulation and role in central nervous system disorders. CHOLESTEROL 2012; 2012:292598. [PMID: 23119149 PMCID: PMC3483652 DOI: 10.1155/2012/292598] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 09/03/2012] [Accepted: 09/10/2012] [Indexed: 02/08/2023]
Abstract
Cholesterol is a major constituent of the human brain, and the brain is the most cholesterol-rich organ. Numerous lipoprotein receptors and apolipoproteins are expressed in the brain. Cholesterol is tightly regulated between the major brain cells and is essential for normal brain development. The metabolism of brain cholesterol differs markedly from that of other tissues. Brain cholesterol is primarily derived by de novo synthesis and the blood brain barrier prevents the uptake of lipoprotein cholesterol from the circulation. Defects in cholesterol metabolism lead to structural and functional central nervous system diseases such as Smith-Lemli-Opitz syndrome, Niemann-Pick type C disease, and Alzheimer's disease. These diseases affect different metabolic pathways (cholesterol biosynthesis, lipid transport and lipoprotein assembly, apolipoproteins, lipoprotein receptors, and signaling molecules). We review the metabolic pathways of cholesterol in the CNS and its cell-specific and microdomain-specific interaction with other pathways such as the amyloid precursor protein and discuss potential treatment strategies as well as the effects of the widespread use of LDL cholesterol-lowering drugs on brain functions.
Collapse
|
135
|
Krol MB, Gromadzinska J, Wasowicz W. SeP, ApoER2 and megalin as necessary factors to maintain Se homeostasis in mammals. J Trace Elem Med Biol 2012; 26:262-6. [PMID: 22683052 DOI: 10.1016/j.jtemb.2012.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 03/19/2012] [Indexed: 11/23/2022]
Abstract
Selenoprotein P (SeP) is an extracellular protein containing ten selenium atoms in the form of selenocysteine, secreted mainly from the liver. About 60% of the whole plasma selenium level is present in SeP, which makes it a useful biomarker of selenium nutritional status. The main functions of SeP are transport and storage of selenium in plasma. It is especially an important protein for the brain, testes and kidneys where the supplementation of the proper amount of Se ensures the synthesis of selenoenzymes with antioxidant properties.Recently, it has been found that SeP uptake in kidneys, testes and brain depends on the apolipoprotein receptor 2 (ApoER2) and lipoprotein megalin receptor (Lrp2). Megalin receptor represents a physiological SeP receptor in kidneys, mediating the re-uptake of secreted SeP from the primary urine. The absence of a functional megalin receptor causes a significant reduction of plasma selenium and the SeP levels as a result of Se excretion. ApoER2 is a SeP receptor in the brain and testes which uptakes Se from the extracellular fluid. Deletion of ApoER2 in mice leads to a lowered selenium level in the brain and testes, neurological dysfunction, production of abnormal spermatozoa, infertility and even death when the subjects are fed a low-selenium diet.
Collapse
Affiliation(s)
- Magdalena Beata Krol
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | | | | |
Collapse
|
136
|
Sagare AP, Bell RD, Zlokovic BV. Neurovascular dysfunction and faulty amyloid β-peptide clearance in Alzheimer disease. Cold Spring Harb Perspect Med 2012; 2:cshperspect.a011452. [PMID: 23028132 DOI: 10.1101/cshperspect.a011452] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurovascular dysfunction is an integral part of Alzheimer disease (AD). Changes in the brain vascular system may contribute in a significant way to the onset and progression of cognitive decline and the development of a chronic neurodegenerative process associated with accumulation of amyloid β-peptide (Aβ) in brain and cerebral vessels in AD individuals and AD animal models. Here, we review the role of the neurovascular unit and molecular mechanisms in cerebral vascular cells behind the pathogenesis of AD. In particular, we focus on blood-brain barrier (BBB) dysfunction, decreased cerebral blood flow, and impaired vascular clearance of Aβ from brain. The data reviewed here support an essential role of the neurovascular and BBB mechanisms in AD pathogenesis.
Collapse
Affiliation(s)
- Abhay P Sagare
- Center for Neurodegenerative and Vascular Brain Disorders and Interdisciplinary Program in Dementia Research, Arthur Kornberg Medical Research Building, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | |
Collapse
|
137
|
Lipoproteins in Drosophila melanogaster--assembly, function, and influence on tissue lipid composition. PLoS Genet 2012; 8:e1002828. [PMID: 22844248 PMCID: PMC3406001 DOI: 10.1371/journal.pgen.1002828] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/25/2012] [Indexed: 11/19/2022] Open
Abstract
Interorgan lipid transport occurs via lipoproteins, and altered lipoprotein levels correlate with metabolic disease. However, precisely how lipoproteins affect tissue lipid composition has not been comprehensively analyzed. Here, we identify the major lipoproteins of Drosophila melanogaster and use genetics and mass spectrometry to study their assembly, interorgan trafficking, and influence on tissue lipids. The apoB-family lipoprotein Lipophorin (Lpp) is the major hemolymph lipid carrier. It is produced as a phospholipid-rich particle by the fat body, and its secretion requires Microsomal Triglyceride Transfer Protein (MTP). Lpp acquires sterols and most diacylglycerol (DAG) at the gut via Lipid Transfer Particle (LTP), another fat body-derived apoB-family lipoprotein. The gut, like the fat body, is a lipogenic organ, incorporating both de novo-synthesized and dietary fatty acids into DAG for export. We identify distinct requirements for LTP and Lpp-dependent lipid mobilization in contributing to the neutral and polar lipid composition of the brain and wing imaginal disc. These studies define major routes of interorgan lipid transport in Drosophila and uncover surprising tissue-specific differences in lipoprotein lipid utilization.
Collapse
|
138
|
Spuch C, Ortolano S, Navarro C. LRP-1 and LRP-2 receptors function in the membrane neuron. Trafficking mechanisms and proteolytic processing in Alzheimer's disease. Front Physiol 2012; 3:269. [PMID: 22934024 PMCID: PMC3429044 DOI: 10.3389/fphys.2012.00269] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/26/2012] [Indexed: 11/13/2022] Open
Abstract
Low density lipoprotein receptor-related protein (LRP) belongs to the low-density lipoprotein receptor family, generally recognized as cell surface endocytic receptors, which bind and internalize extracellular ligands for degradation in lysosomes. Neurons require cholesterol to function and keep the membrane rafts stable. Cholesterol uptake into the neuron is carried out by ApoE via LRPs receptors on the cell surface. In neurons the most important are LRP-1 and LRP-2, even it is thought that a causal factor in Alzheimer's disease (AD) is the malfunction of this process which cause impairment intracellular signaling as well as storage and/or release of nutrients and toxic compounds. Both receptors are multifunctional cell surface receptors that are widely expressed in several tissues including neurons and astrocytes. LRPs are constituted by an intracellular (ICD) and extracellular domain (ECD). Through its ECD, LRPs bind at least 40 different ligands ranging from lipoprotein and protease inhibitor complex to growth factors and extracellular matrix proteins. These receptors has also been shown to interact with scaffolding and signaling proteins via its ICD in a phosphorylation-dependent manner and to function as a co-receptor partnering with other cell surface or integral membrane proteins. Thus, LRPs are implicated in two major physiological processes: endocytosis and regulation of signaling pathways, which are both involved in diverse biological roles including lipid metabolism, cell growth processes, degradation of proteases, and tissue invasion. Interestingly, LRPs were also localized in neurons in different stages, suggesting that both receptors could be implicated in signal transduction during embryonic development, neuronal outgrowth or in the pathogenesis of AD.
Collapse
Affiliation(s)
- Carlos Spuch
- Department of Pathology and Neuropathology, University Hospital of VigoVigo, Spain
| | | | | |
Collapse
|
139
|
Lee TW, Montgomery JM, Birch NP. The serine protease inhibitor neuroserpin regulates the growth and maturation of hippocampal neurons through a non-inhibitory mechanism. J Neurochem 2012; 121:561-74. [PMID: 22191421 DOI: 10.1111/j.1471-4159.2011.07639.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuroserpin is a brain-specific serine protease inhibitor that is expressed in the developing and adult nervous system. Its expression profile led to suggestions that it played roles in neuronal growth and connectivity. In this study, we provide direct evidence to support a role for neuroserpin in axon and dendritic growth. We report that axon growth is enhanced while axon and dendrite diameter are reduced following neuroserpin treatment of hippocampal neurons. More complex effects are seen on dendritic growth and branching with neuroserpin-stimulating dendritic growth and branching in young neurons but switching to an inhibitory response in older neurons. The protease inhibitory activity of neuroserpin is not required to activate changes in neuronal morphology and a proportion of responses are modulated by an antagonist to the LRP1 receptor. Collectively, these findings support a key role for neuroserpin as a regulator of neuronal development through a non-inhibitory mechanism and suggest a basis for neuroserpin's effects on complex emotional behaviours and recent link to schizophrenia.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
140
|
Huang XY, Shi GM, Devbhandari RP, Ke AW, Wang Y, Wang XY, Wang Z, Shi YH, Xiao YS, Ding ZB, Dai Z, Xu Y, Jia WP, Tang ZY, Fan J, Zhou J. Low level of low-density lipoprotein receptor-related protein 1 predicts an unfavorable prognosis of hepatocellular carcinoma after curative resection. PLoS One 2012; 7:e32775. [PMID: 22427881 PMCID: PMC3299691 DOI: 10.1371/journal.pone.0032775] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 01/30/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor involved in receptor-mediated endocytosis and cell signaling. The aim of this study was to elucidate the expression and mechanism of LRP1 in hepatocellular carcinoma (HCC). METHODS LRP1 expression in 4 HCC cell lines and 40 HCC samples was detected. After interruption of LRP1 expression in a HCC cell line either with specific lentiviral-mediated shRNA LRP1 or in the presence of the LRP1-specific chaperone, receptor-associated protein (RAP), the role of LRP1 in the migration and invasion of HCC cells was assessed in vivo and in vitro, and the expression of matrix metalloproteinase (MMP) 9 in cells and the bioactivity of MMP9 in the supernatant were assayed. The expression and prognostic value of LRP1 were investigated in 327 HCC specimens. RESULTS Low LRP1 expression was associated with poor HCC prognosis, with low expression independently related to shortened overall survival and increased tumor recurrence rate. Expression of LRP1 in non-recurrent HCC samples was significantly higher than that in early recurrent samples. LRP1 expression in HCC cell lines was inversely correlated with their metastatic potential. After inhibition of LRP1, low-metastatic SMCC-7721 cells showed enhanced migration and invasion and increased expression and bioactivity of MMP9. Correlation analysis showed a negative correlation between LRP1 and MMP9 expression in HCC patients. The prognostic value of LRP1 expression was validated in the independent data set. CONCLUSIONS LRP1 modulated the level of MMP9 and low level of LRP1 expression was associated with aggressiveness and invasiveness in HCCs. LRP1 offered a possible strategy for tumor molecular therapy.
Collapse
Affiliation(s)
- Xiao-Yong Huang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, People's Republic of China,
| | - Guo-Ming Shi
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | | | - Ai-Wu Ke
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Yuwei Wang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Xiao-Ying Wang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Zheng Wang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Ying-Hong Shi
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Yong-Sheng Xiao
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Zhen-Bin Ding
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Zhi Dai
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Yang Xu
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Wei-Ping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Zhao-You Tang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Jia Fan
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Cancer Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, People's Republic of China,
| | - Jian Zhou
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Cancer Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, People's Republic of China,
| |
Collapse
|
141
|
Basak JM, Verghese PB, Yoon H, Kim J, Holtzman DM. Low-density lipoprotein receptor represents an apolipoprotein E-independent pathway of Aβ uptake and degradation by astrocytes. J Biol Chem 2012; 287:13959-71. [PMID: 22383525 DOI: 10.1074/jbc.m111.288746] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Accumulation of the amyloid β (Aβ) peptide within the brain is hypothesized to be one of the main causes underlying the pathogenic events that occur in Alzheimer disease (AD). Consequently, identifying pathways by which Aβ is cleared from the brain is crucial for better understanding of the disease pathogenesis and developing novel therapeutics. Cellular uptake and degradation by glial cells is one means by which Aβ may be cleared from the brain. In the current study, we demonstrate that modulating levels of the low-density lipoprotein receptor (LDLR), a cell surface receptor that regulates the amount of apolipoprotein E (apoE) in the brain, altered both the uptake and degradation of Aβ by astrocytes. Deletion of LDLR caused a decrease in Aβ uptake, whereas increasing LDLR levels significantly enhanced both the uptake and clearance of Aβ. Increasing LDLR levels also enhanced the cellular degradation of Aβ and facilitated the vesicular transport of Aβ to lysosomes. Despite the fact that LDLR regulated the uptake of apoE by astrocytes, we found that the effect of LDLR on Aβ uptake and clearance occurred in the absence of apoE. Finally, we provide evidence that Aβ can directly bind to LDLR, suggesting that an interaction between LDLR and Aβ could be responsible for LDLR-mediated Aβ uptake. Therefore, these results identify LDLR as a receptor that mediates Aβ uptake and clearance by astrocytes, and provide evidence that increasing glial LDLR levels may promote Aβ degradation within the brain.
Collapse
Affiliation(s)
- Jacob M Basak
- Department of Neurology, Hope Center for Neurological Disorders, Charles F and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
142
|
Kikuchi A, Yamamoto H, Sato A, Matsumoto S. New insights into the mechanism of Wnt signaling pathway activation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 291:21-71. [PMID: 22017973 DOI: 10.1016/b978-0-12-386035-4.00002-1] [Citation(s) in RCA: 208] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Wnts compromise a large family of secreted, hydrophobic glycoproteins that control a variety of developmental and adult processes in all metazoan organisms. Recent advances in the Wnt-signal studies have revealed that distinct Wnts activate multiple intracellular cascades that regulate cellular proliferation, differentiation, migration, and polarity. Although the mechanism by which Wnts regulate different pathways selectively remains to be clarified, evidence has accumulated that in addition to the formation of ligand-receptor pairs, phosphorylation of receptors, receptor-mediated endocytosis, acidification, and the presence of cofactors, such as heparan sulfate proteoglycans, are also involved in the activation of specific Wnt pathways. Here, we review the mechanism of activation in Wnt signaling initiated on the cell-surface membrane. In addition, the mechanisms for fine-tuning by cross talk between Wnt and other signaling are also discussed.
Collapse
Affiliation(s)
- Akira Kikuchi
- Department of Biochemistry, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | |
Collapse
|
143
|
Zhang B, Liang C, Bates R, Yin Y, Xiong WC, Mei L. Wnt proteins regulate acetylcholine receptor clustering in muscle cells. Mol Brain 2012; 5:7. [PMID: 22309736 PMCID: PMC3296622 DOI: 10.1186/1756-6606-5-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 02/06/2012] [Indexed: 01/08/2023] Open
Abstract
Background The neuromuscular junction (NMJ) is a cholinergic synapse that rapidly conveys signals from motoneurons to muscle cells and exhibits a high degree of subcellular specialization characteristic of chemical synapses. NMJ formation requires agrin and its coreceptors LRP4 and MuSK. Increasing evidence indicates that Wnt signaling regulates NMJ formation in Drosophila, C. elegans and zebrafish. Results In the study we systematically studied the effect of all 19 different Wnts in mammals on acetylcholine receptor (AChR) cluster formation. We identified five Wnts (Wnt9a, Wnt9b, Wnt10b, Wnt11, and Wnt16) that are able to stimulate AChR clustering, of which Wnt9a and Wnt11 are expressed abundantly in developing muscles. Using Wnt9a and Wnt11 as example, we demonstrated that Wnt induction of AChR clusters was dose-dependent and non-additive to that of agrin, suggesting that Wnts may act via similar pathways to induce AChR clusters. We provide evidence that Wnt9a and Wnt11 bind directly to the extracellular domain of MuSK, to induce MuSK dimerization and subsequent tyrosine phosphorylation of the kinase. In addition, Wnt-induced AChR clustering requires LRP4. Conclusions These results identify Wnts as new players in AChR cluster formation, which act in a manner that requires both MuSK and LRP4, revealing a novel function of LRP4.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurology and Institute of Molecular Medicine and Genetics, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | | | | | | | | | |
Collapse
|
144
|
Lindhorst T, Kurz H, Sibbe M, Meseke M, Förster E. Congruence of vascular network remodeling and neuronal dispersion in the hippocampus of reelin-deficient mice. Histochem Cell Biol 2012; 137:629-39. [PMID: 22261923 DOI: 10.1007/s00418-012-0912-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2012] [Indexed: 12/18/2022]
Abstract
In the hippocampus, neurons and fiber projections are strictly organized in layers and supplied with oxygen via a vascular network that also develops layer-specific characteristics in wild-type mice, as shown in the present study for the first time in a quantitative manner. By contrast, in the reeler mutant, well known for its neuronal migration defects due to the lack of the extracellular matrix protein reelin, emerging layer-specific characteristics of the vascular pattern were found to be remodeled during development of the dentate gyrus. Remarkably, in the first postnatal week, when a granule cell layer was still discernable in the reeler dentate gyrus, also the reeler vascular pattern resembled wild type. Thus, at postnatal day 6, unbranched microvessels traversed the granule cell layer and bifurcated when reaching the subgranular zone. Only after the first postnatal week vascular network remodeling in the reeler dentate gyrus became apparent, when the proportion of dispersed granule cells increased. Hence, vessel bifurcation frequency decreased in the maturing reeler dentate gyrus, but increased in wild type, resulting in significant differences (approx. 100%; p < 0.01) between adult wild type and reeler. Moreover, layer-specific vessel bifurcation frequencies disappeared in the maturing reeler dentate gyrus. Finally, a wild type-like vascular pattern was also found in the dentate gyrus of mice deficient for the reelin receptor very low density lipoprotein receptor (VLDLR), precluding a requirement of VLDLR for normal vascular pattern formation in the dentate gyrus. In sum, our findings show that vascular network remodeling in the reeler dentate gyrus is closely linked to the progression of granule cell dispersion.
Collapse
Affiliation(s)
- Tina Lindhorst
- Department of Hematology and Oncology, University Medical Center Freiburg, Freiburg, Germany
| | | | | | | | | |
Collapse
|
145
|
The role of APP proteolytic processing in lipid metabolism. Exp Brain Res 2011; 217:365-75. [DOI: 10.1007/s00221-011-2975-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 12/01/2011] [Indexed: 12/14/2022]
|
146
|
Kievit FM, Zhang M. Cancer nanotheranostics: improving imaging and therapy by targeted delivery across biological barriers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H217-47. [PMID: 21842473 PMCID: PMC3397249 DOI: 10.1002/adma.201102313] [Citation(s) in RCA: 350] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/12/2011] [Indexed: 05/03/2023]
Abstract
Cancer nanotheranostics aims to combine imaging and therapy of cancer through use of nanotechnology. The ability to engineer nanomaterials to interact with cancer cells at the molecular level can significantly improve the effectiveness and specificity of therapy to cancers that are currently difficult to treat. In particular, metastatic cancers, drug-resistant cancers, and cancer stem cells impose the greatest therapeutic challenge for targeted therapy. Targeted therapy can be achieved with appropriately designed drug delivery vehicles such as nanoparticles, adult stem cells, or T cells in immunotherapy. In this article, we first review the different types of nanotheranostic particles and their use in imaging, followed by the biological barriers they must bypass to reach the target cancer cells, including the blood, liver, kidneys, spleen, and particularly the blood-brain barrier. We then review how nanotheranostics can be used to improve targeted delivery and treatment of cancer cells. Finally, we discuss development of nanoparticles to overcome current limitations in cancer therapy.
Collapse
Affiliation(s)
- Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
147
|
Bourhis E, Wang W, Tam C, Hwang J, Zhang Y, Spittler D, Huang OW, Gong Y, Estevez A, Zilberleyb I, Rouge L, Chiu C, Wu Y, Costa M, Hannoush RN, Franke Y, Cochran AG. Wnt antagonists bind through a short peptide to the first β-propeller domain of LRP5/6. Structure 2011; 19:1433-42. [PMID: 21944579 DOI: 10.1016/j.str.2011.07.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/30/2011] [Accepted: 07/02/2011] [Indexed: 01/27/2023]
Abstract
The Wnt pathway inhibitors DKK1 and sclerostin (SOST) are important therapeutic targets in diseases involving bone loss or damage. It has been appreciated that Wnt coreceptors LRP5/6 are also important, as human missense mutations that result in bone overgrowth (bone mineral density, or BMD, mutations) cluster to the E1 propeller domain of LRP5. Here, we report a crystal structure of LRP6 E1 bound to an antibody, revealing that the E1 domain is a peptide recognition module. Remarkably, the consensus E1 binding sequence is a close match to a conserved tripeptide motif present in all Wnt inhibitors that bind LRP5/6. We show that this motif is important for DKK1 and SOST binding to LRP6 and for inhibitory function, providing a detailed structural explanation for the effect of the BMD mutations.
Collapse
Affiliation(s)
- Eric Bourhis
- Department of Early Discovery Biochemistry, Genentech Research and Early Development, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Low-density lipoprotein receptor-related protein is decreased in optic neuropathy of Alzheimer disease. J Neuroophthalmol 2011; 31:139-46. [PMID: 21593627 DOI: 10.1097/wno.0b013e31821b602c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Alzheimer disease (AD) is associated with optic nerve degeneration, yet the underlying pathophysiology of this disease and the optic nerve disorder remain poorly understood. Low-density lipoprotein receptor-related protein (LRP) is implicated in the pathogenesis of AD by mediating the transport of amyloid-β (Aβ) out of the brain into the systemic circulation. As a key player in the reaction to central nervous system injury, astrocytes associate with LRP in AD. This study investigates the role of LRP and astrocytes in the pathogenesis of AD optic neuropathy. METHODS To investigate the role of LRP and astrocytes in the pathogenesis of AD optic neuropathy, we conducted immunohistochemical studies on postmortem optic nerves in AD patients (n = 11) and age-matched controls (n = 10) to examine the presence of LRP. Quantitative analyses using imaging software were used to document the extent of LRP in neural tissues. Axonal integrity was assessed by performing immunohistochemistry on the subjects' optic nerves with an antibody to neurofilament (NF) protein. Double-immunofluorescence labeling was performed to investigate whether LRP colocalized with astrocytes, expressing glial fibrillary acidic protein. RESULTS LRP expression was decreased in AD optic nerves compared to that in controls (P < 0.001). LRP immunoreactivity was observed in the microvasculature and perivascularly in close proximity to the astrocytic processes. Colocalization of LRP in the astrocytes of optic nerves was also demonstrated. The presence of optic neuropathy was confirmed in the AD optic nerves by demonstrating greatly reduced immunostaining for NF protein as compared to controls. CONCLUSIONS The reduction of LRP in the AD degenerative optic nerves supports the hypothesis that LRP may play a role in the pathophysiology of AD optic neuropathy.
Collapse
|
149
|
Yip YP, Zhou G, Kubo KI, Nakajima K, Yip JW. Reelin inhibits migration of sympathetic preganglionic neurons in the spinal cord of the chick. J Comp Neurol 2011; 519:1970-8. [PMID: 21452229 DOI: 10.1002/cne.22616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The present study examined the effects of Reelin in the migration of sympathetic preganglionic neurons (SPN) in the spinal cord of the chick. SPN in the chick first migrate from the neuroepithelium to the ventrolateral spinal cord. They then undergo a secondary migration to cluster adjacent to the central canal, forming the column of Terni (CT). During secondary migration, abundant Reelin is found in large areas of the ventral spinal cord; the only areas devoid of Reelin are areas occupied by SPN or somatic motor neurons and the pathway along which SPN migrate. Ectopic expression of Reelin in the pathway of SPN through electroporation of full-length Reelin DNA stopped SPN migration toward their destination. The spatiotemporal pattern of Reelin expression, along with the inhibition of SPN migration by exogenous Reelin, suggests that Reelin functions as a barrier to SPN migration during normal development of the spinal cord.
Collapse
Affiliation(s)
- Yee Ping Yip
- Department of Neurobiology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | |
Collapse
|
150
|
Xia CH, Lu E, Liu H, Du X, Beutler B, Gong X. The role of Vldlr in intraretinal angiogenesis in mice. Invest Ophthalmol Vis Sci 2011; 52:6572-9. [PMID: 21757581 DOI: 10.1167/iovs.10-7082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE. To identify and characterize the r26 mouse line, which displays depigmented patches in the retina, and to determine the causative gene mutation and study the underlying mechanism. METHODS. Fundus examination, fluorescein angiography, histology, and immunostaining were used to determine the retinal phenotypes. Genome-wide linkage analysis, DNA sequencing, and an allelic test were used to identify the causative gene mutation. Wild-type and mutant gene products were examined by Western blot and transient transfection. RESULTS. Homozygous r26/r26 mice displayed depigmented patches in the fundus that overlapped the hyperfluorescent spots in the angiogram. Histology showed overgrown retinal vessels in the subretinal space. Immunostaining verified the presence of endothelial cells in the photoreceptor layer. Chromosome mapping and DNA sequencing revealed a point mutation, c.2239C>T, in the very-low-density lipoprotein receptor (Vldlr) gene. An allelic test in Vldlr knockout (-/-) mice confirmed that r26/(-) mice display a phenotype similar to that of r26/r26 mice. The Vldlr protein was predominantly localized at the plasma membrane of transfected cells, whereas the truncated Vldlr was diffusely expressed in the cell cytosol. The r26 truncated Vldlr was undetectable in mutant retinas by Western blot. CONCLUSIONS. The r26 is a recessive mutant caused by a missense mutation in the Vldlr gene. This results in a truncated Vldlr protein that lacks the C-terminal 127 amino acid residues including the single transmembrane domain and fails to localize at the plasma membrane. Thus, the r26 is a loss-of-function Vldlr mutation. Vldlr on the cell surface probably mediates an antiangiogenic signal to prevent retinal endothelial cells from migrating into the photoreceptor cell layer.
Collapse
Affiliation(s)
- Chun-Hong Xia
- School of Optometry and Vision Science Program, University of California, Berkeley, USA.
| | | | | | | | | | | |
Collapse
|