Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Int Immunopharmacol ; 133: 112005, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38626543

RESUMO

BACKGROUND: Because the pathophysiology of osteoarthritis (OA) has not been fully elucidated, targeted treatments are lacking. In this study, we assessed the role and underlying mechanism apolipoprotein D (APOD) on the development of OA. METHODS: To establish an in vitro OA model, we extracted primary chondrocytes from the cartilage of C57BL/6 mice and stimulated the chondrocytes with IL-1ß. After APOD intervention or incubation with an overexpressing plasmid, we detected inflammatory-related markers using RT-qPCR, Western blotting, and ELISA. To detect apoptosis and autophagy-related markers, we used flow cytometry, immunofluorescence, and transmission electron microscopy (TEM). Finally, we measured the level of oxidative stress. We also used RNA-seq to identify the APOD-regulated downstream signaling pathways. We used an in vivo mice OA model of the anterior cruciate ligament transection (ACLT) and administered intra-articular adenovirus overexpressing APOD. To examine cartilage damage severity, we used immunohistochemical analysis (IHC), micro-CT, scanning electron microscopy (SEM), and Safranin O-fast green staining. RESULTS: Our results showed that APOD inhibited chondrocyte inflammation, degeneration, and apoptosis induced by IL-1ß. Additionally, APOD reversed autophagy inhibition and oxidative stress and also blocked activation of the PI3K/AKT/mTOR signaling pathway induced by IL-1ß. Finally, overexpression of the APOD gene through adenovirus was sufficient to mitigate OA progression. CONCLUSIONS: Our findings revealed that APOD had a chondroprotective role in OA progression by the PI3K/AKT/mTOR signaling pathway.


Assuntos
Apolipoproteínas D , Condrócitos , Osteoartrite do Joelho , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Serina-Treonina Quinases TOR , Animais , Masculino , Camundongos , Apolipoproteínas D/genética , Apolipoproteínas D/metabolismo , Apoptose , Autofagia , Cartilagem Articular/patologia , Cartilagem Articular/metabolismo , Células Cultivadas , Condrócitos/metabolismo , Modelos Animais de Doenças , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Osteoartrite do Joelho/patologia , Osteoartrite do Joelho/metabolismo , Estresse Oxidativo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
Rejuvenation Res ; 26(2): 42-50, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36571249

RESUMO

The current understanding of skin aging is that senescent fibroblasts accumulate within the dermis and subcutaneous fat to cause abnormal tissue remodeling and extracellular matrix dysfunction, triggering a senescence-associated secretory phenotype (SASP). A novel therapeutic approach to prevent skin aging is to specifically eliminate senescent dermal fibroblasts; this requires the identification of specific protein markers for senescent cells. Apolipoprotein D (ApoD) is involved in lipid metabolism and antioxidant responses and is abundantly expressed in tissues affected by age-related diseases such as Alzheimer's disease and atherosclerosis. However, its behavior and role in skin aging remain unclear. In this study, we examined whether ApoD functions as a marker of aging using human dermal fibroblast aging models. In cellular senescence models induced through replicative aging and ionizing radiation exposure, ApoD expression was upregulated at the gene and protein levels and correlated with senescence-associated ß-galactosidase activity and the decreased uptake of the proliferation marker bromodeoxyuridine, which was concomitant with the upregulation of SASP genes. Furthermore, ApoD-positive cells were found to be more abundant in the aging human dermis using fluorescence flow cytometry. These results suggest that ApoD is a potential clinical marker for identifying aging dermal fibroblasts.


Assuntos
Apolipoproteínas D , Envelhecimento da Pele , Humanos , Apolipoproteínas D/metabolismo , Células Cultivadas , Senescência Celular/genética , Fibroblastos/metabolismo , Rejuvenescimento
3.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638803

RESUMO

Adipose tissue releases a large range of bioactive factors called adipokines, many of which are involved in inflammation, glucose homeostasis and lipid metabolism. Under pathological conditions such as obesity, most of the adipokines are upregulated and considered as deleterious, due to their pro-inflammatory, pro-atherosclerotic or pro-diabetic properties, while only a few are downregulated and would be designated as beneficial adipokines, thanks to their counteracting properties against the onset of comorbidities. This review focuses on six adipose-derived lipid-binding proteins that have emerged as key factors in the development of obesity and diabetes: Retinol binding protein 4 (RBP4), Fatty acid binding protein 4 (FABP4), Apolipoprotein D (APOD), Lipocalin-2 (LCN2), Lipocalin-14 (LCN14) and Apolipoprotein M (APOM). These proteins share structural homology and capacity to bind small hydrophobic molecules but display opposite effects on glucose and lipid metabolism. RBP4 and FABP4 are positively associated with metabolic syndrome, while APOD and LCN2 are ubiquitously expressed proteins with deleterious or beneficial effects, depending on their anatomical site of expression. LCN14 and APOM have been recently identified as adipokines associated with healthy metabolism. Recent findings on these lipid-binding proteins exhibiting detrimental or protective roles in human and murine metabolism and their involvement in metabolic diseases are also discussed.


Assuntos
Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Síndrome Metabólica/metabolismo , Animais , Apolipoproteínas D/metabolismo , Apolipoproteínas M/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Humanos , Lipocalina-2/metabolismo , Síndrome Metabólica/etiologia , Obesidade/metabolismo , Proteínas Plasmáticas de Ligação ao Retinol/metabolismo
4.
Neuropharmacology ; 194: 108378, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-33160981

RESUMO

Parkinson's disease (PD) is the second most common age-related neurodegenerative disorder after Alzheimer's disease. Increasing evidence highlights the role of age-related chronic inflammation, oxidative stress and mitochondrial dysfunction in the pathogenesis of PD. A combination of these factors impairs the crosstalk between mitochondria and lysosomes, resulting in compromised cell homeostasis. Apolipoprotein D (APOD), an ancient and highly conserved anti-inflammatory and antioxidant lipocalin, and the transcription factor EB (TFEB), a master regulator of mitophagy, autophagy and lysosomal biogenesis, play key roles in these processes. Both APOD and TFEB have attracted attention as therapeutic targets for PD. The aim of this study was to investigate if the selective cyclooxygenase-2 inhibitor celecoxib (CXB) exerts a direct neuroprotective effect in 6-hydroxydopamine (6-OHDA) and paraquat (PQ) PD models. We found that CXB rescued SH-SY5Y cells challenged by 6-OHDA- and PQ-induced toxicity. Furthermore, treatment with CXB led to a marked and sustained upregulation of APOD and the two microphthalmia transcription factors TFEB and MITF. In sum, this study highlights the clinically approved drug CXB as a promising neuroprotective therapeutic tool in PD research that has the potential to increase the survival rate of dopaminergic neurons that are still alive at the time of diagnosis.


Assuntos
Apolipoproteínas D/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Celecoxib/farmacologia , Doença de Parkinson Secundária/tratamento farmacológico , Linhagem Celular Tumoral , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Neuroproteção/efeitos dos fármacos , Oxidopamina , Paraquat , Regulação para Cima
5.
Gene ; 756: 144874, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32554047

RESUMO

ApoD is a 25 to 30 kDa glycosylated protein, member of the lipocalin superfamily. As a transporter of several small hydrophobic molecules, its known biological functions are mostly associated to lipid metabolism and neuroprotection. ApoD is a multi-ligand, multi-function protein that is involved lipid trafficking, food intake, inflammation, antioxidative response and development and in different types of cancers. An important aspect of ApoD's role in lipid metabolism appears to involve the transport of arachidonic acid, and the modulation of eicosanoid production and delivery in metabolic tissues. ApoD expression in metabolic tissues has been associated positively and negatively with insulin sensitivity and glucose homeostasis in a tissue dependent manner. ApoD levels rise considerably in association with aging and neuropathologies such as Alzheimer's disease, stroke, meningoencephalitis, moto-neuron disease, multiple sclerosis, schizophrenia and Parkinson's disease. ApoD is also modulated in several animal models of nervous system injury/pathology.


Assuntos
Apolipoproteínas D/metabolismo , Animais , Apolipoproteínas D/química , Apolipoproteínas D/genética , Desenvolvimento Embrionário , Humanos , Neoplasias/metabolismo , Sistema Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Especificidade de Órgãos
6.
Histopathology ; 76(7): 976-987, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31994214

RESUMO

AIMS: Apolipoprotein D (ApoD) is a protein that is regulated by androgen and oestrogen, and is a major constituent of breast cysts. Although ApoD has been reported to be a marker of breast cancer, its prognostic importance in invasive breast cancer is unclear. The aim of this study was to investigate the relationship between ApoD protein expression, oestrogen receptor-α (ERα) expression and androgen receptor (AR) expression in predicting breast cancer outcome. METHODS AND RESULTS: ApoD levels were measured by the use of immunohistochemistry and video image analysis on tissue sections from a breast cancer cohort (n = 214). We assessed the associations of ApoD expression with disease-free survival (DFS), metastasis-free survival (MFS), and overall survival (OS). We also assessed the relationship between ApoD expression, AR expression and ERα expression in predicting OS. ApoD expression (>1% ApoD positivity) was found in 72% (154/214) of tissues. High ApoD positivity (≥20.7%, fourth quartile) was an independent predictor of MFS and OS, and conferred a 2.2-fold increased risk of developing metastatic disease and a 2.1-fold increased risk of breast cancer-related death. ApoD positivity was not associated with AR or ERα nuclear positivity. However, patients with (≥1%) ERα-positive cancers with low (<20.7%) ApoD positivity, or those showing high (≥78%) AR positivity and low (<20.7%) ApoD positivity had better OS than other patient groups. CONCLUSIONS: ApoD expression could be used to predict breast cancer prognosis independently of ERα and AR expression.


Assuntos
Apolipoproteínas D/metabolismo , Biomarcadores Tumorais/análise , Neoplasias da Mama/patologia , Adulto , Apolipoproteínas D/análise , Feminino , Humanos , Pessoa de Meia-Idade , Prognóstico , Resultado do Tratamento
7.
Biomolecules ; 9(12)2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31779197

RESUMO

Lecithin:cholesterol acyltransferase (LCAT) is an enzyme secreted by the liver and circulates with high-density lipoprotein (HDL) in the blood. The enzyme esterifies plasma cholesterol and increases the capacity of HDL to carry and potentially remove cholesterol from tissues. Cholesterol accumulates within the extracellular connective tissue matrix of the cornea stroma in individuals with genetic deficiency of LCAT. LCAT can be activated by apolipoproteins (Apo) including ApoD and ApoA1. ApoA1 also mediates cellular synthesis of HDL. This study examined the expression of LCAT by epithelial cells, keratocytes, and endothelial cells, the cell types that comprise from anterior to posterior the three layers of the cornea. LCAT and ApoD were immunolocalized to all three cell types within the cornea, while ApoA1 was immunolocalized to keratocytes and endothelium but not epithelium. In situ hybridization was used to detect LCAT, ApoD, and ApoA1 mRNA to learn what cell types within the cornea synthesize these proteins. No corneal cells showed mRNA for ApoA1. Keratocytes and endothelium both showed ApoD mRNA, but epithelium did not. Epithelium and endothelium both showed LCAT mRNA, but despite the presence of LCAT protein in keratocytes, keratocytes did not show LCAT mRNA. RNA sequencing analysis of serum-cultured dedifferentiated keratocytes (commonly referred to as corneal stromal fibroblasts) revealed the presence of both LCAT and ApoD (but not ApoA1) mRNA, which was accompanied by their respective proteins detected by immunolabeling of the cultured keratocytes and Western blot analysis of keratocyte lysates. The results indicate that keratocytes in vivo show both ApoA1 and LCAT proteins, but do not synthesize these proteins. Rather, keratocytes in vivo must take up ApoA1 and LCAT from the corneal interstitial tissue fluid.


Assuntos
Apolipoproteína A-I/metabolismo , Apolipoproteínas D/metabolismo , Colesterol/metabolismo , Córnea/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo , Idoso , Apolipoproteína A-I/sangue , Apolipoproteína A-I/genética , Apolipoproteínas D/sangue , Apolipoproteínas D/genética , Córnea/enzimologia , Córnea/patologia , Córnea/ultraestrutura , Distrofias Hereditárias da Córnea/genética , Distrofias Hereditárias da Córnea/metabolismo , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/metabolismo , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Queratinócitos/metabolismo , Deficiência da Lecitina Colesterol Aciltransferase/genética , Deficiência da Lecitina Colesterol Aciltransferase/metabolismo , Lipoproteínas HDL/sangue , Masculino , Microscopia Eletrônica , Pessoa de Meia-Idade , Fosfatidilcolina-Esterol O-Aciltransferase/sangue , Fosfatidilcolina-Esterol O-Aciltransferase/genética , Fosfolipídeos/metabolismo , RNA-Seq , Doença de Tangier/genética , Doença de Tangier/metabolismo
8.
J Mol Neurosci ; 67(1): 125-132, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30467822

RESUMO

Frontotemporal dementia (FTD) and Alzheimer's disease (AD) are the two common forms of dementia. FTD syndromes are characterized by lobar atrophy (frontotemporal lobar degeneration or FTLD) and the presence of either cellular TDP43 (FTLD-TDP), tau (FTLD-tau), or FUS aggregates, while extracellular ß-amyloid plaques and hyperphosphorylated tau tangles develop in AD. Oxidative stress can induce these pathological modifications in disease models, and is thought to play a role in these syndromes. Apolipoprotein D (apoD) is a glial-expressed lipocalin known to protect against oxidative stress, with increased levels in AD, supporting a protective role. The expression of apoD has not been studied in FTLD. This study assesses apoD expression in FTLD-TDP and FTLD-tau in comparison to AD and controls. It also analyzes the effect of apoD on TARDBP (TDP43 gene) and ß-amyloid precursor protein (APP). The expression of apoD was analyzed by Western blotting in FTLD-TDP, FTLD-tau, AD, and control post-mortem brain tissue. An apoD-overexpressing cell model was used to study the impact of increased apoD on APP and TARDBP expression. We confirm that apoD expression was increased in AD but surprisingly it was not affected in either of the two main pathological forms of FTLD. Under oxidative stress conditions, apoD had no effect on TDP43 expression but it did decrease APP expression. This suggests that apoD does not act as a neuroprotective factor in FTLD in the same way as in AD. This could contribute to the more rapid degeneration observed in FTLD.


Assuntos
Doença de Alzheimer/metabolismo , Apolipoproteínas D/genética , Demência Frontotemporal/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Apolipoproteínas D/metabolismo , Encéfalo/metabolismo , Linhagem Celular Tumoral , Feminino , Demência Frontotemporal/genética , Humanos , Masculino , Regulação para Cima
9.
Endocrine ; 61(2): 248-257, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29869155

RESUMO

PURPOSE: Apolipoprotein D (ApoD) is a lipocalin participating in lipid transport. It binds to a variety of ligands, with a higher affinity for arachidonic acid, and is thought to have a diverse array of functions. We investigated a potential role for ApoD in insulin sensitivity, inflammation, and thrombosis-processes related to lipid metabolism-in severely obese women. METHODS: We measured ApoD expression in a cohort of 44 severely obese women including dysmetabolic and non-dysmetabolic patients. Physical and metabolic characteristics of these women were determined from anthropometric measurements and blood samples. ApoD was quantified at the mRNA and protein levels in samples from three intra-abdominal adipose tissues (AT): omental, mesenteric and round ligament (RL). RESULTS: ApoD protein levels were highly variable between AT of the same individual. High ApoD protein levels, particularly in the RL depot, were linked to lower plasma insulin levels (-40%, p = 0.015) and insulin resistance (-47%, p = 0.022), and increased insulin sensitivity (+10%, p = 0.008). Lower circulating pro-inflammatory PAI-1 (-39%, p = 0.001), and TNF-α (-19%, p = 0.030) levels were also correlated to high ApoD protein in the RL AT. CONCLUSIONS: ApoD variability between AT was consistent with different accumulation efficiencies and/or metabolic functions according to the anatomic location of fat depots. Most statistically significant correlations implicated ApoD protein levels, in agreement with protein accumulation in target tissues. These correlations associated higher ApoD levels in fat depots with improved metabolic health in severely obese women.


Assuntos
Apolipoproteínas D/genética , Inflamação/sangue , Gordura Intra-Abdominal/metabolismo , Obesidade Mórbida/genética , Obesidade Mórbida/metabolismo , Ligamentos Redondos/metabolismo , Adulto , Apolipoproteínas D/metabolismo , Feminino , Humanos , Inflamação/complicações , Inflamação/metabolismo , Mediadores da Inflamação/sangue , Resistência à Insulina/genética , Interleucina-6/sangue , Metabolismo dos Lipídeos/fisiologia , Pessoa de Meia-Idade , Obesidade Mórbida/complicações , Obesidade Mórbida/patologia , Inibidor 1 de Ativador de Plasminogênio/sangue , Fator de Necrose Tumoral alfa/sangue , Adulto Jovem
10.
Glia ; 66(3): 670-687, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29222871

RESUMO

To compact the extracellular sides of myelin, an important transition must take place: from membrane sliding, while building the wraps, to membrane adhesion and water exclusion. Removal of the negatively charged glycocalyx becomes the limiting factor in such transition. What is required to initiate this membrane-zipping process? Knocking-out the Lipocalin Apolipoprotein D (ApoD), essential for lysosomal functional integrity in glial cells, results in a specific defect in myelin extracellular leaflet compaction in peripheral and central nervous system, which results in reduced conduction velocity and suboptimal behavioral outputs: motor learning is compromised. Myelination initiation, growth, intracellular leaflet compaction, myelin thickness or internodal length remain unaltered. Lack of ApoD specifically modifies Plp and P0 protein expression, but not Mbp or Mag. Late in myelin maturation period, ApoD affects lipogenic and growth-related, but not stress-responsive, signaling pathways. Without ApoD, the sialylated glycocalyx is maintained and ganglioside content remains high. In peripheral nervous system, Neu3 membrane sialidase and lysosomal Neu1 are coordinately expressed with ApoD in subsets of Schwann cells. ApoD-KO myelin becomes depleted of Neu3 and enriched in Fyn, a kinase with pivotal roles in transducing axon-derived signals into myelin properties. In the absence of ApoD, partial permeabilization of lysosomes alters Neu1 location as well. Exogenous ApoD rescues ApoD-KO hypersialylated glycocalyx in astrocytes, demonstrating that ApoD is necessary and sufficient to control glycocalyx composition in glial cells. By ensuring lysosomal functional integrity and adequate subcellular location of effector and regulatory proteins, ApoD guarantees the glycolipid recycling and glycocalyx removal required to complete myelin compaction.


Assuntos
Apolipoproteínas D/metabolismo , Glicocálix/metabolismo , Lisossomos/metabolismo , Bainha de Mielina/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Apolipoproteínas D/administração & dosagem , Apolipoproteínas D/genética , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Escherichia coli , Espaço Extracelular/metabolismo , Deficiências da Aprendizagem/metabolismo , Deficiências da Aprendizagem/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Mucolipidoses/metabolismo , Neuraminidase/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Nervo Isquiático/citologia , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/metabolismo
11.
FASEB J ; 31(12): 5568-5576, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28842423

RESUMO

Liver X receptors (LXRs) are important sensors and regulators for cholesterol, fatty acid, and glucose. LXRs play essential roles in the development and progression of cardiovascular diseases. We examined the effects of T0901317, a potent LXR agonist, on angiogenesis of human umbilical vein endothelial cells (HUVECs). Treatment with T0901317 inhibited the tube formation and migration of HUVECs and reduced the in vivo angiogenesis, as determined by chorioallantoic membrane assay. T0901317 stimulated gene and protein expression of LXR target gene apolipoprotein D (ApoD). Overexpression of ApoD suppressed the tube formation of HUVECs. ApoD interacted with scavenger receptor class B member 1 (SR-B1), while knockdown of SR-B1 blocked suppressive effects of T0901317 on HUVEC migration. T0901317 treatment or overexpression of ApoD lessened expression of proteins regulating angiogenesis, including phospho-eNOS S1177, phospho-Akt T308, phospho-Akt S473, eNOS, mammalian target of rapamycin, VEGF-A, VEGF-C, IL-8, RhoB, matrix metalloproteinase (MMP)-8, -9, and monocyte chemoattractant protein 1. Our study suggested that activation of LXR interferes with angiogenesis through induction of LXR target gene ApoD, which in turn suppresses PI3K-Akt-eNOS signaling, an essential pathway regulating angiogenesis. ApoD may be a potential therapeutic target for tumor angiogenesis.-Lai, C.-J., Cheng, H.-C., Lin, C.-Y., Huang, S.-H., Chen, T.-H., Chung, C.-J., Chang, C.-H., Wang, H.-D., Chuu, C.-P. Activation of liver X receptor suppresses angiogenesis via induction of ApoD.


Assuntos
Apolipoproteínas D/metabolismo , Receptores X do Fígado/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hidrocarbonetos Fluorados/farmacologia , Interleucina-8/metabolismo , Receptores X do Fígado/agonistas , Óxido Nítrico Sintase Tipo III/metabolismo , Receptores Depuradores Classe B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo
12.
PLoS One ; 12(3): e0171453, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28301514

RESUMO

BACKGROUND: Apolipoprotein D (ApoD) has been proposed as a predictor of breast cancer recurrence among estrogen receptor-positive (ER+), tamoxifen-treated patients. METHODS: We conducted a population-based case-control study nested in a population of 11,251 women aged 35-69 years at diagnosis with Stage I-III breast cancer between 1985 and 2001 on Denmark's Jutland Peninsula and registered with the Danish Breast Cancer Cooperative Group. We identified 541 recurrent or contralateral breast cancers cases among women with ER+ disease treated with tamoxifen for at least 1 year and 300 cases in women with ER- disease never treated with tamoxifen. We matched one control subject per case and assessed ApoD expression in the tumor cell nucleus and cytoplasm using tissue microarray immunohistochemistry. We computed the odds ratio (OR) associating ApoD expression with recurrence and adjusted for potential confounding using logistic regression. RESULTS: Cytoplasmic ApoD expression was seen in 68% of ER+ tumors, in 66% of ER- tumors, and in 66% of controls across both groups. In women with ER+ tumors, the associations of cytoplasmic ApoD expression with recurrence (OR = 1.0; 95% CI = 0.7 to 1.4) and increasing cytoplasmic expression with recurrence (OR = 1.0; 95% CI = 0.996 to 1.003) were null, as were those for women with ER- tumors. Associations for nuclear ApoD expression and combined nuclear and cytoplasmic expression were similarly near-null. CONCLUSION: ApoD expression is likely not a predictor of recurrence in tamoxifen-treated patients. IMPACT: This study eliminates the previously suggested marker ApoD as a predictor of recurrence among tamoxifen-treated women.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Apolipoproteínas D/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Recidiva Local de Neoplasia , Tamoxifeno/uso terapêutico , Adolescente , Idoso , Neoplasias da Mama/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Modelos Teóricos , Receptores de Estrogênio/metabolismo
13.
PLoS Genet ; 13(2): e1006603, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28182653

RESUMO

Environmental insults such as oxidative stress can damage cell membranes. Lysosomes are particularly sensitive to membrane permeabilization since their function depends on intraluminal acidic pH and requires stable membrane-dependent proton gradients. Among the catalog of oxidative stress-responsive genes is the Lipocalin Apolipoprotein D (ApoD), an extracellular lipid binding protein endowed with antioxidant capacity. Within the nervous system, cell types in the defense frontline, such as astrocytes, secrete ApoD to help neurons cope with the challenge. The protecting role of ApoD is known from cellular to organism level, and many of its downstream effects, including optimization of autophagy upon neurodegeneration, have been described. However, we still cannot assign a cellular mechanism to ApoD gene that explains how this protection is accomplished. Here we perform a comprehensive analysis of ApoD intracellular traffic and demonstrate its role in lysosomal pH homeostasis upon paraquat-induced oxidative stress. By combining single-lysosome in vivo pH measurements with immunodetection, we demonstrate that ApoD is endocytosed and targeted to a subset of vulnerable lysosomes in a stress-dependent manner. ApoD is functionally stable in this acidic environment, and its presence is sufficient and necessary for lysosomes to recover from oxidation-induced alkalinization, both in astrocytes and neurons. This function is accomplished by preventing lysosomal membrane permeabilization. Two lysosomal-dependent biological processes, myelin phagocytosis by astrocytes and optimization of neurodegeneration-triggered autophagy in a Drosophila in vivo model, require ApoD-related Lipocalins. Our results uncover a previously unknown biological function of ApoD, member of the finely regulated and evolutionary conserved gene family of extracellular Lipocalins. They set a lipoprotein-mediated regulation of lysosomal membrane integrity as a new mechanism at the hub of many cellular functions, critical for the outcome of a wide variety of neurodegenerative diseases. These results open therapeutic opportunities by providing a route of entry and a repair mechanism for lysosomes in pathological situations.


Assuntos
Astrócitos/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Apolipoproteínas D/genética , Apolipoproteínas D/metabolismo , Apolipoproteínas D/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/ultraestrutura , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral , Células Cultivadas , Drosophila , Células HEK293 , Herbicidas/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Immunoblotting , Lipocalinas/farmacologia , Lisossomos/química , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Modelos Biológicos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Neurônios/efeitos dos fármacos , Paraquat/farmacologia , Fagossomos/metabolismo
14.
Mol Neurobiol ; 54(6): 3948-3963, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27271124

RESUMO

Excitotoxicity due to the excessive activation of glutamatergic receptors leads to neuronal dysfunction and death. Excitotoxicity has been implicated in the pathogenesis of a myriad of neurodegenerative diseases with distinct etiologies such as Alzheimer's and Parkinson's. Numerous studies link apolipoprotein D (apoD), a secreted glycoprotein highly expressed in the central nervous system (CNS), to maintain and protect neurons in various mouse models of acute stress and neurodegeneration. Here, we used a mouse model overexpressing human apoD in neurons (H-apoD Tg) to test the neuroprotective effects of apoD in the kainic acid (KA)-lesioned hippocampus. Our results show that apoD overexpression in H-apoD Tg mice induces an increased resistance to KA-induced seizures, significantly attenuates inflammatory responses and confers protection against KA-induced cell apoptosis in the hippocampus. The apoD-mediated protection against KA-induced toxicity is imputable in part to increased plasma membrane Ca2+ ATPase type 2 expression (1.7-fold), decreased N-methyl-D-aspartate receptor (NMDAR) subunit NR2B levels (30 %) and lipid metabolism alterations. Indeed, we demonstrate that apoD can attenuate intracellular cholesterol content in primary hippocampal neurons and in brain of H-apoD Tg mice. In addition, apoD can be internalised by neurons and this internalisation is accentuated in ageing and injury conditions. Our results provide additional mechanistic information on the apoD-mediated neuroprotection in neurodegenerative conditions.


Assuntos
Apolipoproteínas D/metabolismo , Ácido Caínico/toxicidade , Neuroproteção , Neurotoxinas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Colesterol/metabolismo , Endocitose , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Inflamação/patologia , Camundongos Transgênicos , Modelos Biológicos , Neurônios/metabolismo , Neuroproteção/efeitos dos fármacos , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Subunidades Proteicas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/patologia , Regulação para Cima/efeitos dos fármacos
15.
Arkh Patol ; 78(5): 9-14, 2016.
Artigo em Russo | MEDLINE | ID: mdl-27804940

RESUMO

Prostate cancer is one of the most frequently detected malignancies in men. The gold standard for its diagnosis is morphological examination; at the same time the differential diagnosis of adenocarcinoma, high-grade prostatic intraepithelial neoplasia (HGPIN), and benign conditions that are able to mimic the malignancies is tremendously difficult in a number of cases, this being so, the hyperdiagnosis rate of HGPIN requiring mandatory repeat biopsy is as high as 24%. The currently available differential diagnostic panel of antibodies is imperfect, which necessitates a search for novel markers. AIM: to estimate the diagnostic and prognostic value of the expression of PTOV1, APOD, and EPHA4 in prostatic neoplasias. MATERIAL AND METHODS: A total of 90 samples from prostate cancer patients who had undergone radical prostatectomy were examined. The presence of adenocarcinoma and HGPIN was verified by immunohistochemical tests using antibodies to AMACR (P504S) and high molecular weight cytokeratin 34ßE12 in serial sections. The latter were also used to immunohistochemically analyze the expression of PTOV1, APOD, and EPHA4. RESULTS: APOD expression was noted in 76% of cases of both adenocarcinomas and HGPIN, in 4% in only cancer, and in 7% in only HGPIN. All the study samples showed a considerable decrease in PTOV1 expression in cancer and HGPIN compared to morphologically normal glands. Three samples also exhibited no PTOV1 expression in a number of morphologically normal glands. No difference was found in the expression of EPHA4 in morphologically normal glands, HGPIN, or cancer. CONCLUSION: The high rate of APOD expression in HGPIN and cancer, as well as the absence of its expression in the vast majority of morphologically normal glands allows the use of this protein as an additional marker in the differential diagnosis of prostatic neoplasms. The emerging trends in the difference of PTOV1 expression in morphologically normal prostate tissue, HGPIN, and cancer call for further investigations with a larger sample.


Assuntos
Adenocarcinoma/metabolismo , Apolipoproteínas D/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma in Situ/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/metabolismo , Receptor EphA4/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Apolipoproteínas D/genética , Biomarcadores Tumorais/genética , Carcinoma in Situ/patologia , Estudos de Casos e Controles , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Neoplasias da Próstata/patologia , Receptor EphA4/genética
16.
Metabolism ; 65(9): 1247-58, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27506732

RESUMO

BACKGROUND: Apolipoprotein D (ApoD) is a member of the lipocalin family known to transport small hydrophobic ligands. A major site of ApoD expression in mice is the central nervous system where evidence suggests that it plays a protective role. Gene expression of ApoD was reported in bone-forming osteoblasts but its impact on bone metabolism remains undocumented. METHODS: We compared basic bone parameters of ApoD(-/-) (null) and transgenic (tg) mice to wild-type (wt) littermates through microCT and histochemistry, as well as ApoD expression and secretion in osteoblasts under various culture conditions through real-time PCR and immunoblotting. RESULTS: ApoD-null females displayed progressive bone loss with aging, resulting in a 50% reduction in trabecular bone volume and a 23% reduction in cortical bone volume by 9months of age. Only cortical bone volume was significantly reduced in ApoD-null males by an average of 24%. Histochemistry indicated significantly higher osteoblast surface and number of osteoclasts in femora from ApoD-null females. ApoD gene expression was confirmed in primary cultures of bone marrow mesenchymal cells (MSC), with higher expression levels in MSC from females compared to males. ApoD-null MSC exhibited impaired proliferation and differentiation potentials. Moreover, exogenous ApoD partially rescued the osteogenic potential of null MSC, which were shown to readily uptake the protein from media. ApoD expression was upregulated under low proliferation conditions, by contact inhibition and osteoblastic differentiation in MC3T3-E1 osteoblast-like cells. CONCLUSION: Our results indicate that ApoD influences bone metabolism in mice in a gender-specific manner, potentially through an auto-/paracrine pathway.


Assuntos
Envelhecimento/genética , Apolipoproteínas D/deficiência , Desenvolvimento Ósseo/genética , Remodelação Óssea/genética , Osteoblastos , Células 3T3 , Animais , Apolipoproteínas D/genética , Apolipoproteínas D/metabolismo , Células da Medula Óssea/metabolismo , Ciclo Celular/genética , Diferenciação Celular , Proliferação de Células , Feminino , Fêmur/citologia , Fêmur/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Osteoblastos/metabolismo , Cultura Primária de Células
17.
Menopause ; 23(7): 719-30, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27163517

RESUMO

OBJECTIVE: Ospemifene (Osp) is a novel selective estrogen-receptor modulator (SERM) accepted for the treatment of dyspareunia, a symptom of postmenopausal vulvovaginal atrophy. We aimed to analyze the effects of Osp on human breast tissue (HBT), in comparison with the clinically established SERMs raloxifene (Ral) and tamoxifen (Tam), using ex vivo explant cultures. METHODS: HBT samples were obtained from postmenopausal women undergoing mammoplasty and cultured with or without Osp, Ral, Tam, or 17ß-estradiol (E2) for 7 and 14 days, and studied for morphology, proliferation, and apoptosis. The expression of epithelial markers, the estrogen-receptor alpha (ERα), the androgen receptor (AR), TFF1, and apolipoprotein D was evaluated using immunohistochemistry and quantitative reverse transcription-polymerase chain reaction. The PvuII polymorphism of ERS1 was determined. RESULTS: Osp, similar to Ral and Tam, decreased the number of proliferating cells in a concentration-dependent manner (at 100 nM, P < 0.01) and strongly opposed 10 nM E2-stimulated proliferation (P < 0.001). Corresponding effects were observed in the proportions of cells expressing ERα and TFF1 (P < 0.001). At 14 days apoptosis was increased by 100 nM SERMs (P < 0.01), but, notably, decreased by 1 nM Osp and Ral at day 7 (P < 0.05). The SERMs exerted ER-agonist effects on AR-positive cell populations at 1 nM (P < 0.05), but not at 100 nM concentrations. The effects on proliferation and ERα expressing cell numbers were associated with the ERS1 PvuII genotype. CONCLUSIONS: In summary, Osp inhibited proliferation and opposed E2 stimulation in normal HBT in an efficacious, but less potent way than Ral and Tam. The ESR1 PvuII polymorphisms may influence the responsiveness of HBT to E2 and SERMs.


Assuntos
Mama/efeitos dos fármacos , Pós-Menopausa/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/análogos & derivados , Idoso , Apolipoproteínas D/metabolismo , Apoptose/efeitos dos fármacos , Mama/metabolismo , Mama/patologia , Proliferação de Células/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Polimorfismo Genético , Cloridrato de Raloxifeno/farmacologia , Receptores Androgênicos/metabolismo , Tamoxifeno/farmacologia , Fator Trefoil-1/metabolismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-26365933

RESUMO

AIMS: We sought to identify biochemical predictors that indicate susceptibility to in-stent restenosis (ISR) after coronary artery bare-metal stenting. METHODS: A total of 111 consecutive patients with post-percutaneous coronary intervention (PCI) in-stent restenosis of a target lesion within 12 months were matched for age, sex, vessel diameter, and diabetes with 111 controls without post-PCI ISR. Plasma or serum levels of biochemical markers were measured: matrix metalloproteinases (MMP) 2, 3, 9; myeloperoxidase (MPO); asymmetric dimethylarginine (ADMA); lipoprotein (a) (Lp[a]); apolipoproteins E and D (ApoE and D); and lecitin-cholesterol acyltransferase (LCAT). Multivariable logistic regression association tests were performed. RESULTS: Increased plasma MMP-3 (OR: 1.013; 95% CI: 1.004-1.023; P = 0.005), MMP-9 (OR: 1.014; 95% CI: 1.008-1.020; P < 0.0001) or MPO (OR: 1,003; 95% CI: 1.001-1.005; P = 0.002) was significantly associated with increased risk of ISR. Increased levels of ADMA (OR: 0.212; 95% CI: 0.054-0.827; P = 0.026), ApoE (OR: 0.924; 95% CI: 0.899-0.951; P < 0.0001), ApoD (OR: 0.919; 95% CI: 0.880-0.959; P = 0.0001), or LCAT (OR: 0.927; 95% CI: 0.902-0.952; P < 0.0001) was associated with risk reduction. No correlation was found between plasma MMP-2 or Lp (a) and ISR risk. CONCLUSIONS: Increased levels of MMP-3, MMP-9, and MPO represent predictors of ISR after bare-metal stent implantation. In contrast, increased ADMA, LCAT, and Apo E and D indicate a decreased in-stent restenosis occurrence.


Assuntos
Biomarcadores/metabolismo , Reestenose Coronária/diagnóstico , Oclusão de Enxerto Vascular/diagnóstico , Stents , Idoso , Apolipoproteínas D/metabolismo , Apolipoproteínas E/metabolismo , Arginina/análogos & derivados , Arginina/metabolismo , Estudos de Casos e Controles , Reestenose Coronária/fisiopatologia , Feminino , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Lipoproteína(a)/metabolismo , Masculino , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Intervenção Coronária Percutânea , Peroxidase/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo , Grau de Desobstrução Vascular/fisiologia
19.
J Biol Chem ; 290(26): 16077-87, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25918162

RESUMO

Apolipoprotein D (apoD), a member of the lipocalin family, is a 29-kDa secreted glycoprotein that binds and transports small lipophilic molecules. Expressed in several tissues, apoD is up-regulated under different stress stimuli and in a variety of pathologies. Numerous studies have revealed that overexpression of apoD led to neuroprotection in various mouse models of acute stress and neurodegeneration. This multifunctional protein is internalized in several cells types, but the specific internalization mechanism remains unknown. In this study, we demonstrate that the internalization of apoD involves a specific cell surface receptor in 293T cells, identified as the transmembrane glycoprotein basigin (BSG, CD147); more particularly, its low glycosylated form. Our results show that internalized apoD colocalizes with BSG into vesicular compartments. Down-regulation of BSG disrupted the internalization of apoD in cells. In contrast, overexpression of basigin in SH-5YSY cells, which poorly express BSG, restored the uptake of apoD. Cyclophilin A, a known ligand of BSG, competitively reduced apoD internalization, confirming that BSG is a key player in the apoD internalization process. In summary, our results demonstrate that basigin is very likely the apoD receptor and provide additional clues on the mechanisms involved in apoD-mediated functions, including neuroprotection.


Assuntos
Apolipoproteínas D/metabolismo , Basigina/metabolismo , Apolipoproteínas D/genética , Basigina/genética , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Humanos , Transporte Proteico
20.
Mol Med Rep ; 11(6): 4463-7, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25633187

RESUMO

It has been suggested that the adenosine triphosphate­binding cassette sub­family C member 11 (ABCC11) gene polymorphism and apolipoprotein D (ApoD), an odor precursor carrier, may be important in the formation of axillary odor. To date, few studies have examined the potential correlation between these two factors. The present study aimed to investigate the association between a 538 G>A single­nucleotide polymorphism (SNP) of the ABCC11 gene and the mRNA expression levels of ApoD in the apocrine gland of patients with osmidrosis. The 538 G>A polymorphism genotypes of 33 patients with a clinical diagnosis of osmidrosis were analyzed by polymerase chain reaction (PCR) and a base­quenched probe method, and they were divided into two groups according to the results. The G allele functions as a dominant gene; therefore, patients with the GG or GA genotype were allocated to Group I (n=28) and patients with the AA genotype to Group II (n=5). The mRNA expression levels of ApoD in the apocrine glands were determined by reverse transcription­PCR. The results indicated that the mRNA expression levels of ApoD were significantly higher in the apocrine glands of patients in Group I compared with those in Group II (P<0.01). In conclusion, the results indicated that the ABCC11 gene SNP of the 538 G>A allele was associated with a downregulation of the mRNA expression of ApoD in the apocrine glands, which may indicate a role for the ABCC11 gene in the mediation of osmidrosis by enhancing the transition of odor precursors via the ApoD pathway.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Glândulas Apócrinas/metabolismo , Apolipoproteínas D/metabolismo , Polimorfismo de Nucleotídeo Único , Doenças das Glândulas Sudoríparas/genética , Adolescente , Adulto , Alelos , Apolipoproteínas D/genética , Feminino , Genótipo , Humanos , Masculino , RNA Mensageiro/metabolismo , Índice de Gravidade de Doença , Doenças das Glândulas Sudoríparas/patologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA