Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Am J Physiol Endocrinol Metab ; 320(2): E379-E389, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33356995

RESUMO

Low-density lipoprotein receptor-related protein 1 (LRP1) is a member of LDL receptor family that plays a key role in systemic glucose and lipid homeostasis. LRP1 also regulates energy balance in the hypothalamus by mediating leptin's anorexigenic action, although the underlying neurocircuitry involved is still unclear. Because GABAergic neurons are a major mediator of hypothalamic leptin action, we studied the role of GABAergic LRP1 in energy balance and leptin action using mice lacking LRP1 in Vgat- or AgRP-expressing neurons (Vgat-Cre; LRP1loxP/loxP or AgRP-Cre; LRP1loxP/loxP). Here, we show that LRP1 deficiency in GABAergic neurons results in severe obesity in male and female mice fed a normal-chow diet. This effect is most likely due to increased food intake and decreased energy expenditure and locomotor activity. Increased adiposity in GABAergic neuron-specific LRP1-deficient mice is accompanied by hyperleptinemia and hyperinsulinemia. Insulin resistance and glucose intolerance in these mice are occurred without change in body weight. Importantly, LRP1 in GABAergic neurons is not required for leptin action, as evidenced by normal leptin's anorexigenic action and leptin-induced hypothalamic Stat3 phosphorylation. In contrast, LRP1 deficiency in AgRP neurons has no effect on adiposity and caloric intake. In conclusion, our data identify GABAergic neurons as a key neurocircuitry that underpins LRP1-dependent regulation of systemic energy balance and body-weight homeostasis. We further find that the GABAergic LRP1 signaling pathway modulates food intake and energy expenditure independently of leptin signaling and AgRP neurons.


Assuntos
Ingestão de Alimentos , Metabolismo Energético , Neurônios GABAérgicos/patologia , Leptina/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Obesidade/patologia , Receptores para Leptina/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Feminino , Neurônios GABAérgicos/metabolismo , Glucose/metabolismo , Homeostase , Resistência à Insulina , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Receptores para Leptina/genética
2.
Acta Neuropathol ; 139(2): 365-382, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31552482

RESUMO

Oligodendrocyte progenitor cells (OPCs) account for about 5% of total brain and spinal cord cells, giving rise to myelinating oligodendrocytes that provide electrical insulation to neurons of the CNS. OPCs have also recently been shown to regulate inflammatory responses and glial scar formation, suggesting functions that extend beyond myelination. Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifaceted phagocytic receptor that is highly expressed in several CNS cell types, including OPCs. Here, we have generated an oligodendroglia-specific knockout of LRP1, which presents with normal myelin development, but is associated with better outcomes in two animal models of demyelination (EAE and cuprizone). At a mechanistic level, LRP1 did not directly affect OPC differentiation into mature oligodendrocytes. Instead, animals lacking LRP1 in OPCs in the demyelinating CNS were characterized by a robust dampening of inflammation. In particular, LRP1-deficient OPCs presented with impaired antigen cross-presentation machinery, suggesting a failure to propagate the inflammatory response and thus promoting faster myelin repair and neuroprotection. Our study places OPCs as major regulators of neuroinflammation in an LRP1-dependent fashion.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Esclerose Múltipla/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/patologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Cuprizona , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Antígenos de Histocompatibilidade Classe I , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/etiologia , Esclerose Múltipla/patologia
3.
Brain Res ; 1723: 146379, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31415766

RESUMO

Abnormal accumulation of amyloid-ß (Aß) peptide defines progression of Alzheimer's disease (AD) pathology in brain. Here, we investigated expressive changes of two main Aß transport receptors low-density lipoprotein receptor related protein-1 (LRP1) and receptor for advanced glycation end products (RAGE) in a novel AD mice (APP23) with chronic cerebral hypoperfusion (CCH) model, moreover, examined a protective effect of a free radical scavenger edaravone (Eda). In contrast to wild type (WT) and APP23 mice, CCH strongly accelerated abnormal Aß40 depositions and cerebral amyloid angiopathy (CAA) pathology, increased both LRP1 and RAGE expressions in brain parenchyma, while a decrease of LRP1 and an increase of RAGE were observed in vascular endothelial cells at age 12 months (M) of AD mice. Furthermore, CCH strongly increased expressions of two hypoxia-related proteins hypoxia inducible factor-1α (HIF-1α) and heme oxygenase-1 (HO-1), two oxidative-related proteins 4-hydroxy-2-nonenal (4-HNE) and 8-hydroxy-2'-deoxyguanosine (8-OHdG), and decreased both two vital nutrient transporter proteins major facilitator super family domain containing 2a (Mfsd2a) and glucose transporter 1 (Glut1) expressions. Such the above abnormal pathological changes were significantly ameliorated by edaravone treatment. The present study demonstrated that CCH strongly enhanced primary AD pathology causing double imbalances of Aß efflux and influx transport related proteins in the cortical blood vessels in AD mice, and that such a neuropathologic abnormality was greatly ameliorated by Eda.


Assuntos
Doença de Alzheimer/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Isquemia Encefálica/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Edaravone/farmacologia , Células Endoteliais/metabolismo , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fármacos Neuroprotetores/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Receptores de LDL/metabolismo
4.
Pharmacol Ther ; 188: 12-25, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29444453

RESUMO

Atherosclerosis is a dynamic and progressive inflammatory process in the intimal layer of large and medium-sized arteries, and it is the major contributor to the atherosclerotic cardiovascular disease (ACVD), the leading cause of death worldwide. In an atherosclerotic plaque, phagocytosis of apoptotic cells occurs through an intricate process designated efferocytosis. Defective efferocytosis has emerged as a causal factor in the etiopathogenesis of atherosclerosis and its progression into overt ACVD. Both specialized phagocytes (macrophages and dendritic cells) and non-specialized cells with phagocytic capabilities (smooth muscle and endothelial cells) are involved in the efferocytotic process. Moreover, several signaling and regulatory molecules are involved in the different steps of efferocytosis, and they include "Find-Me" signals (lysophosphatidylcholine), "Eat-Me" signals [phosphatidylserine, Mer tyrosine kinase (MerTK), and milk fat globule-EGF factor 8], and "Don't Eat-Me" signals [cluster of differentiation 47 (CD47)]. Regulation of efferocytosis is in a close nexus with inflammation, the key component in atherosclerosis. The predominance of pro-inflammatory and anti-inflammatory molecules plays a crucial role in lesion progression and regression, respectively. Polarization of macrophages towards the M1 phenotype causes them to secrete proinflammatory cytokines, while polarization towards the M2 phenotype causes them to secrete of anti-inflammatory cytokines, including interleukin-10 and transforming growth factor ß, so tending to shift the balance towards resolution of the inflammation. Dysfunction of any regulatory signal may cause expansion of the necrotic core of an atherosclerotic plaque with ensuing conversion of the plaque into an unstable plaque with an increased susceptibility to rupture and to atherothrombotic complication. In this review we aim at elucidating the determinant factors and pathways of efferocytosis which can be considered as potential novel targets when striving to develop more personalized and efficient treatment regimens for patients with ACVD.


Assuntos
Aterosclerose/etiologia , Fagocitose/fisiologia , Antígenos de Superfície/fisiologia , Apoptose , Antígeno CD47/fisiologia , Calreticulina/fisiologia , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Macrófagos/fisiologia , Proteínas do Leite , c-Mer Tirosina Quinase/fisiologia
5.
Cell Mol Life Sci ; 75(9): 1671-1685, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29116364

RESUMO

Low density lipoprotein receptor-related protein (LRP) 1 modulates cell adhesion and motility under normal and pathological conditions. Previous studies documented that LRP1 binds several integrin receptors and mediates their trafficking to the cell surface and endocytosis. However, the mechanism by which LRP1 may regulate integrin activation remains unknown. Here we report that LRP1 promotes the activation and subsequent degradation of ß1 integrin and thus supports cell adhesion, spreading, migration and integrin signaling on fibronectin. LRP1 interacts with surface ß1 integrin, binds the integrin activator kindlin2 and stimulates ß1 integrin-kindlin2 complex formation. Specifically, serine 76 in the LRP1 cytoplasmic tail is crucial for the interaction with kindlin2, ß1 integrin activation and cell adhesion. Interestingly, a loss of LRP1 induces the accumulation of several integrin receptors on the cell surface. Following internalization, intracellular trafficking of integrins is driven by LRP1 in a protein kinase C- and class II myosin-dependent manner. Ultimately, LRP1 dictates the fate of endocytosed ß1 integrin by directing it down the pathway of lysosomal and proteasomal degradation. We propose that LRP1 mediates cell adhesion by orchestrating a multi-protein pathway to activate, traffic and degrade integrins. Thus, LRP1 may serve as a focal point in the integrin quality control system to ensure a firm connection to the extracellular matrix.


Assuntos
Integrina beta1/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Camundongos Knockout , Transporte Proteico/genética , Proteólise , Receptores de LDL/genética , Receptores de LDL/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
6.
Neuromolecular Med ; 19(2-3): 300-308, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28612181

RESUMO

Previous studies have demonstrated that the low-density lipoprotein receptor-related protein-1 (LRP1) plays conflicting roles in Alzheimer's disease (AD) pathogenesis, clearing ß-amyloid (Aß) from the brain while also enhancing APP endocytosis and resultant amyloidogenic processing. We have recently discovered that co-expression of mutant LRP1 C-terminal domain (LRP1-CT C4408R) with Swedish mutant amyloid precursor protein (APPswe) in Chinese hamster ovary (CHO) cells decreases Aß production, while also increasing sAPPα and APP α-C-terminal fragment (α-CTF), compared with CHO cells expressing APPswe alone. Surprisingly, the location of this mutation on LRP1 corresponded with the α-secretase cleavage site of APP. Further experimentation confirmed that in CHO cells expressing APPswe or wild-type APP (APPwt), co-expression of LRP1-CT C4408R decreases Aß and increases sAPPα and α-CTF compared with co-expression of wild-type LRP1-CT. In addition, LRP1-CT C4408R enhanced the unglycosylated form of LRP1-CT and reduced APP endocytosis as determined by flow cytometry. This finding identifies a point mutation in LRP1 which slows LRP1-CT-mediated APP endocytosis and amyloidogenic processing, while enhancing APP α-secretase cleavage, thus demonstrating a potential novel target for slowing AD pathogenesis.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Mutação de Sentido Incorreto , Mutação Puntual , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , Endocitose , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Domínios Proteicos , Proteínas Recombinantes/metabolismo
7.
Arthritis Rheumatol ; 69(6): 1246-1256, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28235248

RESUMO

OBJECTIVE: The aggrecanase ADAMTS-5 and the collagenase matrix metalloproteinase 13 (MMP-13) are constitutively secreted by chondrocytes in normal cartilage, but rapidly endocytosed via the cell surface endocytic receptor low-density lipoprotein receptor-related protein 1 (LRP-1) and subsequently degraded. This endocytic system is impaired in osteoarthritic (OA) cartilage due to increased ectodomain shedding of LRP-1. The aim of this study was to identify the LRP-1 sheddase(s) in human cartilage and to test whether inhibition of LRP-1 shedding prevents cartilage degradation in OA. METHODS: Cell-associated LRP-1 and soluble LRP-1 (sLRP-1) released from human cartilage explants and chondrocytes were measured by Western blot analysis. LRP-1 sheddases were identified by proteinase inhibitor profiling and gene silencing with small interfering RNAs. Specific monoclonal antibodies were used to selectively inhibit the sheddases. Degradation of aggrecan and collagen in human OA cartilage was measured by Western blot analysis using an antibody against an aggrecan neoepitope and a hydroxyproline assay, respectively. RESULTS: Shedding of LRP-1 was increased in OA cartilage compared with normal tissue. Shed sLRP-1 bound to ADAMTS-5 and MMP-13 and prevented their endocytosis without interfering with their proteolytic activities. Two membrane-bound metalloproteinases, ADAM-17 and MMP-14, were identified as the LRP-1 sheddases in cartilage. Inhibition of their activities restored the endocytic capacity of chondrocytes and reduced degradation of aggrecan and collagen in OA cartilage. CONCLUSION: Shedding of LRP-1 is a key link to OA progression. Local inhibition of LRP-1 sheddase activities of ADAM-17 and MMP-14 is a unique way to reverse matrix degradation in OA cartilage and could be effective as a therapeutic approach.


Assuntos
Anticorpos Monoclonais/farmacologia , Colagenases/efeitos dos fármacos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/efeitos dos fármacos , Osteoartrite/tratamento farmacológico , Proteólise/efeitos dos fármacos , Proteína ADAM17/análise , Proteína ADAM17/metabolismo , Proteína ADAMTS5/metabolismo , Adolescente , Adulto , Agrecanas/efeitos dos fármacos , Cartilagem Articular/metabolismo , Criança , Condrócitos/fisiologia , Colágeno/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Feminino , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 14 da Matriz/análise , Metaloproteinase 14 da Matriz/metabolismo , Pessoa de Meia-Idade , Osteoartrite/fisiopatologia , Adulto Jovem
8.
Proc Natl Acad Sci U S A ; 113(5): 1369-74, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26787872

RESUMO

LDL receptor-related protein-1 (LRP1) is an endocytic and cell-signaling receptor. In mice in which LRP1 is deleted in myeloid cells, the response to lipopolysaccharide (LPS) was greatly exacerbated. LRP1 deletion in macrophages in vitro, under the control of tamoxifen-activated Cre-ER(T) fusion protein, robustly increased expression of proinflammatory cytokines and chemokines. In LRP1-expressing macrophages, proinflammatory mediator expression was regulated by LRP1 ligands in a ligand-specific manner. The LRP1 agonists, α2-macroglobulin and tissue-type plasminogen activator, attenuated expression of inflammatory mediators, even in the presence of LPS. The antagonists, receptor-associated protein (RAP) and lactoferrin (LF), and LRP1-specific antibody had the entirely opposite effect, promoting inflammatory mediator expression and mimicking LRP1 deletion. NFκB was rapidly activated in response to RAP and LF and responsible for the initial increase in expression of proinflammatory mediators. RAP and LF also significantly increased expression of microRNA-155 (miR-155) after a lag phase of about 4 h. miR-155 expression reflected, at least in part, activation of secondary cell-signaling pathways downstream of TNFα. Although miR-155 was not involved in the initial induction of cytokine expression in response to LRP1 antagonists, miR-155 was essential for sustaining the proinflammatory response. We conclude that LRP1, NFκB, and miR-155 function as members of a previously unidentified system that has the potential to inhibit or sustain inflammation, depending on the continuum of LRP1 ligands present in the macrophage microenvironment.


Assuntos
Inflamação/prevenção & controle , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Macrófagos/metabolismo , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Animais , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos
9.
Neurobiol Dis ; 86: 170-6, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26639854

RESUMO

Correct positioning and differentiation of neurons during brain development is a key precondition for proper function. Focal cortical dysplasias (FCDs) are increasingly recognized as causes of therapy refractory epilepsies. Neuropathological analyses of respective surgical specimens from neurosurgery for seizure control often reveal aberrant cortical architecture and/or aberrantly shaped neurons in FCDs. However, the molecular pathogenesis particularly of FCDs with aberrant lamination (so-called FCD type I) is largely unresolved. Lipoproteins and particularly low-density lipoprotein receptor-related protein 12 (LRP12) are involved in brain development. Here, we have examined a potential role of LRP12 in the pathogenesis of FCDs. In vitro knockdown of LRP12 in primary neurons results in impaired neuronal arborization. In vivo ablation of LRP12 by intraventricularly in utero electroporated shRNAs elicits cortical maldevelopment, i.e. aberrant lamination by malpositioning of upper cortical layer neurons. Subsequent epilepsy phenotyping revealed pentylenetetrazol (PTZ)-induced seizures to be aggravated in cortical LRP12-silenced mice. Our data demonstrates IUE mediated cortical gene silencing as an excellent approach to study the role of distinct molecules for epilepsy associated focal brain lesions and suggests LRP12 and lipoprotein homeostasis as potential molecular target structures for the emergence of epilepsy-associated FCDs.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Neurônios/fisiologia , Convulsões/genética , Animais , Movimento Celular , Células Cultivadas , Eletroporação , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Pentilenotetrazol , RNA Interferente Pequeno/metabolismo , Convulsões/induzido quimicamente
10.
J Biol Chem ; 290(24): 14852-65, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-25918169

RESUMO

Low density lipoprotein receptor-related protein (LRP1) mediates the internalization of aggregated LDL (AgLDL), which in turn increases the expression of LRP1 in human vascular smooth muscle cells (hVSMCs). This positive feedback mechanism is thus highly efficient to promote the formation of hVSMC foam cells, a crucial vascular component determining the susceptibility of atherosclerotic plaque to rupture. Here we have determined the LRP1 domains involved in AgLDL recognition with the aim of specifically blocking AgLDL internalization in hVSMCs. The capacity of fluorescently labeled AgLDL to bind to functional LRP1 clusters was tested in a receptor-ligand fluorometric assay made by immobilizing soluble LRP1 "minireceptors" (sLRP1-II, sLRP1-III, and sLRP1-IV) recombinantly expressed in CHO cells. This assay showed that AgLDL binds to cluster II. We predicted three well exposed and potentially immunogenic peptides in the CR7-CR9 domains of this cluster (termed P1 (Cys(1051)-Glu(1066)), P2 (Asp(1090)-Cys(1104)), and P3 (Gly(1127)-Cys(1140))). AgLDL, but not native LDL, bound specifically and tightly to P3-coated wells. Rabbit polyclonal antibodies raised against P3 prevented AgLDL uptake by hVSMCs and were almost twice as effective as anti-P1 and anti-P2 Abs in reducing intracellular cholesteryl ester accumulation. Moreover, anti-P3 Abs efficiently prevented AgLDL-induced LRP1 up-regulation and counteracted the down-regulatory effect of AgLDL on hVSMC migration. In conclusion, domain CR9 appears to be critical for LRP1-mediated AgLDL binding and internalization in hVSMCs. Our results open new avenues for an innovative anti-VSMC foam cell-based strategy for the treatment of vascular lipid deposition in atherosclerosis.


Assuntos
Células Espumosas/citologia , Lipoproteínas LDL/fisiologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Músculo Liso Vascular/citologia , Sequência de Aminoácidos , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/química , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real , Homologia de Sequência de Aminoácidos
11.
J Cell Sci ; 128(8): 1475-80, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25736295

RESUMO

When tissues are injured and blood vessels clot, the local environment becomes ischemic, meaning that there is a lack of adequate supply of oxygen and glucose delivered to the surrounding cells. The heat shock protein-90 (Hsp90) family proteins protect tissues from various environmental insults and participate in the repair of damaged tissue. Here, we report discovery of a new ischemia-responsive mechanism in which the two Hsp90 isoforms Hsp90α and Hsp90ß (also known as HSP90AA1 and HSP90AB1, respectively) work together to promote cell motility in wounded skin and accelerate wound closure. We demonstrate that Hsp90α and Hsp90ß have distinct and non-exchangeable functions during wound healing. Under hypoxia and when there is a lack of serum factors, Hsp90ß binds to the cytoplasmic tail of the LDL receptor-related protein-1 (LRP-1) and stabilizes the receptor at the cell surface. Hsp90α, however, is secreted by the cell into extracellular space where it binds and signals through the LRP-1 receptor to promote cell motility, leading to wound closure. In addition to skin injury, we suggest that this repair mechanism applies broadly to other non-cutaneous injured tissues.


Assuntos
Movimento Celular , Fibroblastos/fisiologia , Proteínas de Choque Térmico HSP90/fisiologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Cicatrização/fisiologia , Animais , Hipóxia Celular , Células Cultivadas , Humanos , Isoformas de Proteínas/fisiologia , Pele/citologia , Suínos
12.
Clin Sci (Lond) ; 128(1): 17-28, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24865476

RESUMO

Low-density lipoprotein receptor-related protein 1 (LRP1) is an endocytic and multi-functional type I cell surface membrane protein, which is known to be phosphorylated by the activated platelet-derived growth factor receptor (PDGFR). The tyrosine kinase inhibitor imatinib, which inhibits PDGFR and c-Abl, and which has previously been reported to counteract ß-cell death and diabetes, has been suggested to reduce atherosclerosis by inhibiting PDGFR-induced LRP1 phosphorylation. The aim of the present study was to study LRP1 function in ß-cells and to what extent imatinib modulates LRP1 activity. LRP1 and c-Abl gene knockdown was performed by RNAi using rat INS-1 832/13 and human EndoC1-ßH1 cells. LRP1 was also antagonized by treatment with the antagonist low-density lipoprotein receptor-related protein associated protein 1 (LRPAP1). We have used PDGF-BB, a PDGFR agonist, and apolipoprotein E (ApoE), an LRP1 agonist, to stimulate the activities of PDGFR and LRP1 respectively. Knockdown or inhibition of LRP1 resulted in increased hydrogen peroxide (H2O2)- or cytokine-induced cell death, and glucose-induced insulin release was lowered in LRP1-silenced cells. These results indicate that LRP1 function is necessary for ß-cell function and that LRP1 is adversely affected by challenges to ß-cell health. PDGF-BB, or the combination of PDGF-BB+ApoE, induced phosphorylation of extracellular-signal-regulated kinase (ERK), Akt and LRP1. LRP1 silencing blocked this event. Imatinib blocked phosphorylation of LRP1 by PDGFR activation but induced phosphorylation of ERK. LRP1 silencing blocked imatinib-induced phosphorylation of ERK. Sunitinib also blocked LRP1 phosphorylation in response to PDGF-BB and induced phosphorylation of ERK, but this latter event was not affected by LRP1 knockdown. siRNA-mediated knockdown of the imatinib target c-Abl resulted in an increased ERK phosphorylation at basal conditions, with no further increase in response to imatinib. Imatinib-induced cell survival of tunicamycin-treated cells was partially mediated by ERK activation. We have concluded that imatinib promotes LRP1-dependent ERK activation, possibly via inhibition of c-Abl, and that this could contribute to the pro-survival effects of imatinib on ß-cells.


Assuntos
Benzamidas/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Piperazinas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Técnicas de Silenciamento de Genes , Mesilato de Imatinib , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/fisiologia , Ratos
13.
Pathol Biol (Paris) ; 62(2): 84-90, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24661974

RESUMO

The low-density lipoprotein receptor-related protein-1 (LRP-1) is a membrane receptor displaying both scavenging and signaling functions. The wide variety of extracellular ligands and of cytoplasmic scaffolding and signaling proteins interacting with LRP-1 gives it a major role not only in physiological processes, such as embryogenesis and development, but also in critical pathological situations, including cancer and neurological disorders. In this review, we describe the molecular mechanisms involved at distinct levels in the regulation of LRP-1, from its expression to the proper location and stability at the cell surface.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Animais , Membrana Celular/metabolismo , Microambiente Celular , Progressão da Doença , Endocitose/fisiologia , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/biossíntese , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/química , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Modelos Moleculares , Proteínas de Neoplasias/fisiologia , Neoplasias/patologia , Peptídeo Hidrolases/metabolismo , Fosforilação , Conformação Proteica , Dobramento de Proteína , Processamento de Proteína Pós-Traducional , Transporte Proteico , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade
14.
Arterioscler Thromb Vasc Biol ; 34(3): 487-98, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24504736

RESUMO

Low-density lipoprotein receptor-related protein-1 (LRP1) is a large endocytic and signaling receptor that is widely expressed. In the liver, LRP1 plays an important role in regulating the plasma levels of blood coagulation factor VIII (fVIII) by mediating its uptake and subsequent degradation. fVIII is a key plasma protein that is deficient in hemophilia A and circulates in complex with von Willebrand factor. Because von Willebrand factor blocks binding of fVIII to LRP1, questions remain on the molecular mechanisms by which LRP1 removes fVIII from the circulation. LRP1 also regulates cell surface levels of tissue factor, a component of the extrinsic blood coagulation pathway. This occurs when tissue factor pathway inhibitor bridges the fVII/tissue factor complex to LRP1, resulting in rapid LRP1-mediated internalization and downregulation of coagulant activity. In the vasculature LRP1 also plays protective role from the development of aneurysms. Mice in which the lrp1 gene is selectively deleted in vascular smooth muscle cells develop a phenotype similar to the progression of aneurysm formation in human patient, revealing that these mice are ideal for investigating molecular mechanisms associated with aneurysm formation. Studies suggest that LRP1 protects against elastin fiber fragmentation by reducing excess protease activity in the vessel wall. These proteases include high-temperature requirement factor A1, matrix metalloproteinase 2, matrix metalloproteinase-9, and membrane associated type 1-matrix metalloproteinase. In addition, LRP1 regulates matrix deposition, in part, by modulating levels of connective tissue growth factor. Defining pathways modulated by LRP1 that lead to aneurysm formation and defining its role in thrombosis may allow for more effective intervention in patients.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Aneurisma/prevenção & controle , Animais , Aterosclerose/metabolismo , Coagulação Sanguínea/fisiologia , Elastina/metabolismo , Endocitose/fisiologia , Matriz Extracelular/metabolismo , Fator VIII/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Fígado/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/química , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Modelos Animais , Modelos Moleculares , Músculo Liso Vascular/metabolismo , Especificidade de Órgãos , Peptídeo Hidrolases/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Conformação Proteica , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de LDL/fisiologia , Transdução de Sinais , Tromboplastina/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Fator de von Willebrand/metabolismo
15.
Blood ; 123(12): 1887-96, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24458438

RESUMO

Emerging evidence suggests a role of the cytokine midkine (MK) in inflammation. In this study, its functional relevance for recruitment of polymorphonuclear neutrophils (PMNs) during acute inflammation was investigated. Intravital microscopy and histologic analysis of tumor necrosis factor-α-stimulated cremaster muscle venules revealed severely compromised leukocyte adhesion and extravasation in MK(-/-) mice compared with MK(+/+) animals. Systemic administration of recombinant MK completely rescued the adhesion defect in MK(-/-) mice. In a hind limb ischemia model, leukocyte accumulation in MK(-/-) mice was significantly diminished compared with MK(+/+) animals. However, MK did not lead to an inflammatory activation of PMNs or endothelial cells suggesting that it does not serve as classical proinflammatory cytokine. Unexpectedly, immobilized MK mediated PMN adhesion under static and flow conditions, whereas PMN-derived MK was dispensable for the induction of adhesion. Furthermore, adhesion strengthening remained unaffected by MK. Flow cytometry revealed that immobilized, but not soluble MK, significantly promoted the high affinity conformation of ß2 integrins of PMNs. Blocking studies of low-density lipoprotein receptor-related protein 1 (LRP1) suggested that LRP1 may act as a receptor for MK on PMNs. Thus, MK seems to support PMN adhesion by promoting the high affinity conformation of ß2 integrins, thereby facilitating PMN trafficking during acute inflammation.


Assuntos
Antígenos CD18/fisiologia , Inflamação/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Neutrófilos/fisiologia , Animais , Antígenos CD11/fisiologia , Antígenos CD18/genética , Adesão Celular/imunologia , Adesão Celular/fisiologia , Citocinas/imunologia , Citocinas/fisiologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/imunologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Midkina , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/imunologia , Fatores de Crescimento Neural/fisiologia , Neutrófilos/imunologia , Neutrófilos/patologia , Receptores de LDL/imunologia , Receptores de LDL/fisiologia , Proteínas Supressoras de Tumor/imunologia , Proteínas Supressoras de Tumor/fisiologia
16.
PLoS One ; 8(11): e82026, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24312398

RESUMO

Valvular disease is common in patients with Marfan syndrome and can lead to cardiomyopathy. However, some patients develop cardiomyopathy in the absence of hemodynamically significant valve dysfunction, suggesting alternative mechanisms of disease progression. Disruption of LDL receptor-related protein-1 (Lrp1) in smooth muscle cells has been shown to cause vascular pathologies similar to Marfan syndrome, with activation of smooth muscle cells, vascular dysfunction and aortic aneurysms. This study used echocardiography and blood pressure monitoring in mouse models to determine whether inactivation of Lrp1 in vascular smooth muscle leads to cardiomyopathy, and if so, whether the mechanism is a consequence of valvular disease. Hemodynamic changes during treatment with captopril were also assessed. Dilation of aortic roots was observed in young Lrp1-knockout mice and progressed as they aged, whereas no significant aortic dilation was detected in wild type littermates. Diastolic blood pressure was lower and pulse pressure higher in Lrp1-knockout mice, which was normalized by treatment with captopril. Aortic dilation was followed by development of aortic insufficiency and subsequent dilated cardiomyopathy due to valvular disease. Thus, smooth muscle cell Lrp1 deficiency results in aortic dilation and insufficiency that causes secondary cardiomyopathy that can be improved by captopril. These findings provide novel insights into mechanisms of cardiomyopathy associated with vascular activation and offer a new model of valvular cardiomyopathy.


Assuntos
Insuficiência da Valva Aórtica/genética , Vasos Sanguíneos/fisiopatologia , Cardiomiopatias/fisiopatologia , Deleção de Genes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Músculo Liso/metabolismo , Envelhecimento/fisiologia , Animais , Insuficiência da Valva Aórtica/diagnóstico por imagem , Pressão Sanguínea , Cardiomiopatias/diagnóstico por imagem , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Proteína Smad2/metabolismo , Ultrassonografia
17.
Cardiovasc Res ; 100(2): 262-71, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23812296

RESUMO

AIM: Migration of vascular smooth muscle cells (VSMCs) is a crucial event in atherosclerosis and vascular repair. Low-density lipoprotein (LDL) infiltrated in the vessel wall become aggregated (agLDL) and internalized by VSMC through the LDL receptor-related protein LRP1, deriving in lipid-loaded cells with reduced motility capacity. The urokinase-plasminogen activator (UPA)/UPA receptor (UPAR) system plays a relevant role in vascular remodelling. Here, we investigated whether UPA-ligand binding is involved in the detrimental effects of lipid loading in VSMC migration. METHODS AND RESULTS: Animals fed a high-fat diet had 10-fold higher cholesterol-LDL plasma levels, >60% decrease in aortic UPA-protein expression, and VSMC showed impaired outgrowth from aortic explants. Angiotensin II infusion significantly increased aortic UPA expression and accelerated VSMC migration. Using an in vitro model of wound repair, we showed that agLDL inhibits UPA-mediated VSMC migration. UPA silencing reduced migration in control cells to levels observed in lipid-loaded VSMC. UPA silencing did not affect migration in lipid-loaded VSMC. UPA expression was significantly decreased in agLDL-exposed VSMC. agLDL also induced changes in the subcellular localization of UPA, with a reduction in colocalization with UPAR strongly evident at the front edge of agLDL-treated migrating cells. Rescue experiments showed that UPA acting as UPAR ligand restored migration capacity of agLDL-VSMC to control levels. The effects of UPA/UPAR on migration of lipid-loaded cells occurred through the binding to LRP-1. CONCLUSION: UPA-ligand binding regulates VSMC migration, a process that is interfered by LDL. Thus, tissue infiltrated LDL through the abrogation of UPA function reduces VSMC-regulated vascular repair.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Angiotensina II/farmacologia , Animais , Movimento Celular , Células Cultivadas , Humanos , Hipercolesterolemia/patologia , Lipoproteínas LDL/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Ativador de Plasminogênio Tipo Uroquinase/análise , Cicatrização
18.
Arterioscler Thromb Vasc Biol ; 33(5): 935-42, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23471233

RESUMO

OBJECTIVE: Macrophage (Mϕ) migration rests on the adhesion/detachment between Mϕ surface components and extracellular matrixes, and the contribution of numerous inflammatory disorders. Plasminogen activator inhibitor (PAI)-1, a serine protease inhibitor, influences Mϕ motility through an action distinct from its classical modulation of the plasmin-based fibrinolytic process. We rely here on a small molecule PAI-1 inhibitor (TM5275) to investigate the role of PAI-1 in Mϕ migration in the pathogenesis of renal injury. APPROACH AND RESULTS: Mϕ migration was inhibited both in vitro and in vivo by TM5275. It was also reduced in T-cell-deficient nude mice, but not in PAI-1-deficient mice. Mϕ migration hinged on the interaction of PAI-1 with low-density lipoprotein receptor-related protein, an interaction prevented by TM5275, but not with vitronectin, urokinase-type plasminogen activator, or tissue-type plasminogen activator. Fed to rats with anti-Thy-1-induced nephritis, TM5275 significantly decreased Mϕ accumulation and ameliorated the progression of renal injury. CONCLUSIONS: These findings suggest that a small molecule PAI-1 inhibitor represents a novel class of anti-inflammatory agents targeting Mϕ migration by the inhibition of the interaction of PAI-1 with low-density lipoprotein receptor-related protein.


Assuntos
Macrófagos/efeitos dos fármacos , Piperazinas/farmacologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , para-Aminobenzoatos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Movimento Celular/efeitos dos fármacos , Glomerulonefrite/patologia , Isoanticorpos/farmacologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Macrófagos/fisiologia , Camundongos , Ratos
19.
Yakugaku Zasshi ; 132(9): 1019-24, 2012.
Artigo em Japonês | MEDLINE | ID: mdl-23023418

RESUMO

To investigate cerebral infarction and intracranial hemorrhage (ICH) in relation to antithrombotic agents, we established an animal stroke model induced by using a combination of a photosensitive dye and local green photoillumination. Microplasmin (µPli), a derivative of plasmin lacking the five "kringle" domains, was administered in this model, and its effect was studied using magnetic resonance imaging. µPli treatment reduced cerebral damage 24 h after middle cerebral artery occlusion; it also reduced the expansion of the positive area on perfusion-weighted images between 1 and 24 h and the degree of neurological deficits. Tissue-type plasminogen activator (t-PA), a serine proteinase that converts plasminogen to plasmin, has been approved for treating acute ischemic stroke, but delayed treatment is associated with increased risk of ICH. Plasmin participates in the degradation of fibrin, causing clot lysis, and of various extracellular matrix proteins, either directly or via the activation of matrix metalloproteinases (MMPs). In this study, we observed that MMP-3 is relatively important in the enhanced risk of ICH induced by delayed t-PA treatment for ischemic stroke. In particular, the binding of t-PA with low-density lipoprotein receptor-related protein (LRP) results in the release of MMP-3 by endothelial cells. LRP production is upregulated in endothelial cells exposed to ischemia, and elevated LRP levels have been implicated in the increased ICH risk associated with delayed t-PA treatment. This implies that the t-PA/LRP/MMP-3 pathway may be a suitable target for developing strategies to improve the therapeutic efficacy of t-PA in acute ischemic stroke.


Assuntos
Hemorragia Cerebral/etiologia , Fibrinolíticos/uso terapêutico , Trombose Intracraniana/tratamento farmacológico , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Hemorragia Cerebral/prevenção & controle , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Avaliação de Medicamentos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Metaloproteinase 3 da Matriz/metabolismo , Camundongos
20.
Pharmacol Ther ; 136(1): 94-105, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22820095

RESUMO

Low-density lipoprotein receptor-related protein-1 (LRP1) is the main cell surface receptor involved in brain and systemic clearance of the Alzheimer's disease (AD) toxin amyloid-beta (Aß). In plasma, a soluble form of LRP1 (sLRP1) is the major transport protein for peripheral Aß. LRP1 in brain endothelium and mural cells mediates Aß efflux from brain by providing a transport mechanism for Aß across the blood-brain barrier (BBB). sLRP1 maintains a plasma 'sink' activity for Aß through binding of peripheral Aß which in turn inhibits re-entry of free plasma Aß into the brain. LRP1 in the liver mediates systemic clearance of Aß. In AD, LRP1 expression at the BBB is reduced and Aß binding to circulating sLRP1 is compromised by oxidation. Cell surface LRP1 and circulating sLRP1 represent druggable targets which can be therapeutically modified to restore the physiological mechanisms of brain Aß homeostasis. In this review, we discuss how increasing LRP1 expression at the BBB and liver with lifestyle changes, statins, plant-based active principles and/or gene therapy on one hand, and how replacing dysfunctional plasma sLRP1 on the other regulate Aß clearance from brain ultimately controlling the onset and/or progression of AD.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Homeostase , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Doença de Alzheimer/etiologia , Animais , Barreira Hematoencefálica , Exercício Físico , Terapia Genética , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...