Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 261
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Obes (Lond) ; 48(7): 973-980, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38491190

RESUMO

BACKGROUND: The adiponectin is one of the rare adipokines down-regulated with obesity and protects against obesity-related disorders. Similarly, the apolipoprotein M (apoM) is expressed in adipocytes and its expression in adipose tissue is associated with metabolic health. We compared circulating apoM with adiponectin regarding their relationship with metabolic parameters and insulin sensitivity and examined their gene expression patterns in adipocytes and in the adipose tissue. METHODS: Circulating apoM and adiponectin were examined in 169 men with overweight in a cross-sectional study, and 13 patients with obesity during a surgery-induced slimming program. Correlations with clinical parameters including the insulin resistance index (HOMA-IR) were analyzed. Multiple regression analyses were performed on HOMA-IR. The APOM and ADIPOQ gene expression were measured in the adipose tissue from 267 individuals with obesity and a human adipocyte cell line. RESULTS: Participants with type 2 diabetes had lower circulating adiponectin and apoM, while apoM was higher in individuals with dyslipidemia. Similar to adiponectin, apoM showed negative associations with HOMA-IR and hs-CRP (r < -0.2), and positive correlations with HDL markers (HDL-C and apoA-I, r > 0.3). Unlike adiponectin, apoM was positively associated with LDL markers (LDL-C and apoB100, r < 0.20) and negatively correlated with insulin and age (r < -0.2). The apoM was the sole negative determinant of HOMA-IR in multiple regression models, while adiponectin not contributing significantly. After surgery, the change in HOMA-IR was negatively associated with the change in circulating apoM (r = -0.71), but not with the change in adiponectin. The APOM and ADIPOQ gene expression positively correlated in adipose tissue (r > 0.44) as well as in adipocytes (r > 0.81). In adipocytes, APOM was downregulated by inflammatory factors and upregulated by adiponectin. CONCLUSIONS: The apoM rises as a new partner of adiponectin regarding insulin sensitivity. At the adipose tissue level, the adiponectin may be supported by apoM to promote a healthy adipose tissue. TRIAL REGISTRATION: NCT01277068, registered 13 January 2011; NCT02332434, registered 5 January 2015; and NCT00390637, registered 20 October 2006.


Assuntos
Adiponectina , Apolipoproteínas M , Resistência à Insulina , Humanos , Masculino , Apolipoproteínas M/sangue , Resistência à Insulina/fisiologia , Adiponectina/sangue , Estudos Transversais , Pessoa de Meia-Idade , Adulto , Obesidade/sangue , Obesidade/metabolismo , Feminino , Adipócitos/metabolismo , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Biomarcadores/sangue , Tecido Adiposo/metabolismo , Apolipoproteínas/sangue
2.
Int J Neuropsychopharmacol ; 27(7)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38970624

RESUMO

BACKGROUND: Alcohol use disorder (AUD) courses with inflammation and cognitive decline. Apolipoproteins have emerged as novel target compounds related to inflammatory processes and cognition. METHODS: A cross-sectional study was performed on abstinent AUD patients with at least 1 month of abstinence (n = 33; 72.7% men) and healthy controls (n = 34; 47.1% men). A battery of plasma apolipoproteins (APOAI, APOAII, APOB, APOCII, APOE, APOJ, and APOM), plasma inflammatory markers (LPS, LBP), and their influence on cognition and presence of the disorder were investigated. RESULTS: Higher levels of plasma APOAI, APOB, APOE, and APOJ, as well as the proinflammatory LPS, were observed in the AUD group, irrespective of sex, whereas APOM levels were lower vs controls. Hierarchical logistic regression analyses, adjusting for covariates (age, sex, education), associated APOM with the absence of cognitive impairment in AUD and identified APOAI and APOM as strong predictors of the presence or absence of the disorder, respectively. APOAI and APOM did not correlate with alcohol abuse variables or liver status markers, but they showed an opposite profile in their associations with LPS (positive for APOAI; negative for APOM) and cognition (negative for APOAI; positive for APOM) in the entire sample. CONCLUSIONS: The HDL constituents APOAI and APOM were differentially regulated in the plasma of AUD patients compared with controls, playing divergent roles in the disorder identification and associations with inflammation and cognitive decline.


Assuntos
Alcoolismo , Apolipoproteína A-I , Apolipoproteínas M , Disfunção Cognitiva , Inflamação , Humanos , Masculino , Feminino , Apolipoproteínas M/sangue , Projetos Piloto , Disfunção Cognitiva/sangue , Disfunção Cognitiva/etiologia , Pessoa de Meia-Idade , Estudos Transversais , Inflamação/sangue , Alcoolismo/sangue , Adulto , Apolipoproteína A-I/sangue , Biomarcadores/sangue , Apolipoproteínas/sangue
3.
J Am Soc Nephrol ; 34(10): 1629-1646, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37545036

RESUMO

SIGNIFICANCE STATEMENT: Activation of the type 1 IL-1 receptor (IL-1R1) triggers a critical innate immune signaling cascade that contributes to the pathogenesis of AKI. However, blockade of IL-1 signaling in AKI has not consistently demonstrated kidney protection. The current murine experiments show that IL-1R1 activation in the proximal tubule exacerbates toxin-induced AKI and cell death through local suppression of apolipoprotein M. By contrast, IL-1R1 activation in endothelial cells ameliorates AKI by restoring VEGFA-dependent endothelial cell viability. Using this information, future delivery strategies can maximize the protective effects of blocking IL-1R1 while mitigating unwanted actions of IL-1R1 manipulation. BACKGROUND: Activation of the type 1 IL-1 receptor (IL-1R1) triggers a critical innate immune signaling cascade that contributes to the pathogenesis of AKI. IL-1R1 is expressed on some myeloid cell populations and on multiple kidney cell lineages, including tubular and endothelial cells. Pharmacological inhibition of the IL-1R1 does not consistently protect the kidney from injury, suggesting there may be complex, cell-specific effects of IL-1R1 stimulation in AKI. METHODS: To examine expression of IL-1 and IL-1R1 in intrinsic renal versus infiltrating immune cell populations during AKI, we analyzed single-cell RNA sequencing (scRNA-seq) data from kidney tissues of humans with AKI and mice with acute aristolochic acid exposure. We then investigated cell-specific contributions of renal IL-1R1 signaling to AKI using scRNA-seq, RNA microarray, and pharmacological interventions in mice with IL-1R1 deletion restricted to the proximal tubule or endothelium. RESULTS: scRNA-seq analyses demonstrated robust IL-1 expression in myeloid cell populations and low-level IL-1R1 expression in kidney parenchymal cells during toxin-induced AKI. Our genetic studies showed that IL-1R1 activation in the proximal tubule exacerbated toxin-induced AKI and cell death through local suppression of apolipoprotein M. By contrast, IL-1R1 activation in endothelial cells ameliorated aristolochic acid-induced AKI by restoring VEGFA-dependent endothelial cell viability and density. CONCLUSIONS: These data highlight opposing cell-specific effects of IL-1 receptor signaling on AKI after toxin exposure. Disrupting pathways activated by IL-1R1 in the tubule, while preserving those triggered by IL-1R1 activation on endothelial cells, may afford renoprotection exceeding that of global IL-1R1 inhibition while mitigating unwanted actions of IL-1R1 blockade.


Assuntos
Injúria Renal Aguda , Receptores de Interleucina-1 , Humanos , Camundongos , Animais , Receptores de Interleucina-1/genética , Apolipoproteínas M , Células Endoteliais/metabolismo , Injúria Renal Aguda/patologia , Camundongos Knockout , Interleucina-1 , Endotélio/metabolismo , Camundongos Endogâmicos C57BL
4.
Breast Cancer Res Treat ; 201(3): 571-576, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37490173

RESUMO

PURPOSE: Apolipoprotein M (APOM) is a plasma apolipoprotein closely involved with lipid metabolism and inflammation. In vitro studies suggest that APOM may also have a tumor-suppressive role in breast cancer. In the present study, we aimed to evaluate the impact of plasma APOM levels on the prognosis of breast cancer patients. METHODS: We measured APOM levels using an enzyme-linked immunosorbent assay in 75 patients with ER-positive/HER2-negative metastatic breast cancer. The endpoint was overall survival (OS) at 24 months. RESULTS: During the 24-month follow-up period, 34.7% of the patients died. Baseline APOM levels were significantly reduced in patients who deceased during follow-up compared to survivors (42.7 ± 14.5 µg/mL versus 52.2 ± 13.8 µg/mL; P = 0.003). Cox regression analysis showed a hazard ratio of 0.30 [95% confidence interval 0.15-0.61]; P < 0.001 per doubling of APOM levels. Correction for age, C-reactive protein, menopausal state, histology of the primary tumor, metastatic site, number of metastases, endocrine resistance, scheduled therapy line, and kind of scheduled therapy indicated that circulating APOM predicted OS independently of these parameters (HRper doubling = 0.23 [0.09-0.56; P = 0.001). CONCLUSIONS: Our study suggests that circulating APOM is significantly linked with reduced mortality in metastatic breast cancer patients.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Apolipoproteínas , Apolipoproteínas M , Ensaio de Imunoadsorção Enzimática , Menopausa
5.
Arch Biochem Biophys ; 743: 109642, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37211224

RESUMO

Renal cell carcinoma is one of the most common malignancies worldwide, and kidney renal clear cell carcinoma (KIRC) is the most common histopathological type of renal cell carcinoma. However, the mechanism of KIRC progression remains poorly understood. Apolipoprotein M (ApoM) is a plasma apolipoprotein and a member of the lipid transport protein superfamily. Lipid metabolism is essential for tumor progression, and its related proteins can be used as therapeutic targets for tumors. ApoM influences the development of several cancers, but its relationship with KIRC remains unclear. In this study, we aimed to investigate the biological function of ApoM in KIRC and to reveal its potential molecular mechanisms. We found that ApoM expression was significantly reduced in KIRC and was strongly correlated with patient prognosis. ApoM overexpression significantly inhibited KIRC cell proliferation in vitro, suppressed the epithelial mesenchymal transition (EMT) of KIRC cells, and decreased their metastatic capacity. Additionally, the growth of KIRC cells was inhibited by ApoM overexpression in vivo. In addition, we found that overexpression of ApoM in KIRC attenuated Hippo-YAP protein expression and YAP stability and thus inhibited KIRC growth and progression. Therefore, ApoM may be a potential target for the treatment of KIRC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Apolipoproteínas M/metabolismo , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Rim/patologia , Neoplasias Renais/metabolismo , Transdução de Sinais , Proteínas de Sinalização YAP
6.
Curr Opin Lipidol ; 33(3): 199-207, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35695616

RESUMO

PURPOSE OF REVIEW: To better define the metabolism of sphingosine-1-phosphate (S1P), its transport in plasma and its interactions with S1P receptors on vascular cells, and to evaluate the effect of statin treatment on the subnormal plasma levels of high-density lipoprotein (HDL)-bound S1P characteristic of the atherogenic dyslipidemia of metabolic syndrome (MetS). RECENT FINDINGS: Neither clinical intervention trials targeted to raising high-density lipoprotein-cholesterol (HDL-C) levels nor human genome-wide association studies (GWAS) studies have provided evidence to support an atheroprotective role of HDL. Recently however a large monogenic univariable Mendelian randomization on the N396S mutation in the gene encoding endothelial lipase revealed a causal protective effect of elevated HDL-C on coronary artery disease conferred by reduced enzyme activity. Given the complexity of the HDL lipidome and proteome, components of HDL other than cholesterol may in all likelihood contribute to such a protective effect. Among HDL lipids, S1P is a bioactive sphingolipid present in a small proportion of HDL particles (about 5%); indeed, S1P is preferentially enriched in small dense HDL3. As S1P is bound to apolipoprotein (apo) M in HDL, such enrichment is consistent with the elevated apoM concentration in HDL3. When HDL/apoM-bound S1P acts on S1P1 or S1P3 receptors in endothelial cells, potent antiatherogenic and vasculoprotective effects are exerted; those exerted by albumin-bound S1P at these receptors are typically weaker. When HDL/apoM-bound S1P binds to S1P2 receptors, proatherogenic effects may potentially be induced. Subnormal plasma levels of HDL-associated S1P are typical of dyslipidemic individuals at high cardiovascular risk and in patients with coronary heart disease. International Guidelines recommend statin treatment as first-line lipid lowering therapy in these groups. The cardiovascular benefits of statin therapy are derived primarily from reduction in low-density lipoprotein (LDL)-cholesterol, although minor contributions from pleiotropic actions cannot be excluded. Might statin treatment therefore normalize, directly or indirectly, the subnormal levels of S1P in dyslipidemic subjects at high cardiovascular risk? Our unpublished findings in the CAPITAIN study (ClinicalTrials.gov: NCT01595828), involving a cohort of obese, hypertriglyceridemic subjects (n = 12) exhibiting the MetS, showed that pitavastatin calcium (4 mg/day) treatment for 180days was without effect on either total plasma or HDL-associated S1P levels, suggesting that statin-mediated improvement of endothelial function is not due to normalization of HDL-bound S1P. Statins may however induce the expression of S1P1 receptors in endothelial cells, thereby potentiating increase in endothelial nitric oxide synthase response to HDL-bound S1P, with beneficial downstream vasculoprotective effects. SUMMARY: Current evidence indicates that S1P in small dense HDL3 containing apoM exerts antiatherogenic effects and that statins exert vasculoprotective effects through activation of endothelial cell S1P1 receptors in response to HDL/apoM-bound S1P.


Assuntos
Dislipidemias , Inibidores de Hidroximetilglutaril-CoA Redutases , Apolipoproteínas M/genética , Colesterol , Células Endoteliais/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Lipoproteínas HDL/metabolismo , Lisofosfolipídeos/genética , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/genética , Esfingosina/metabolismo
7.
J Recept Signal Transduct Res ; 42(3): 285-292, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34006168

RESUMO

Apolipoprotein M (apoM), an apolipoprotein predominantly associated with high-density lipoprotein (HDL), is considered a mediator of the numerous roles of HDL, including reverse cholesterol transport, anti-atherosclerotic, anti-inflammatory and anti-oxidant, and mediates pre-ß-HDL formation. ApoM expression is known to be regulated by a variety of in vivo and in vitro factors. The transcription factors farnesoid X receptor, small heterodimer partner, liver receptor homolog-1, and liver X receptor comprise the signaling cascade network that regulates the expression and secretion of apoM. Moreover, hepatocyte nuclear factor-1α and c-Jun/JunB have been demonstrated to exert opposing regulatory effects on apoM through competitive binding to the same sites in the proximal region of the apoM gene. Furthermore, as a carrier and modulator of sphingosine 1-phosphate (S1P), apoM binds to S1P within its hydrophobic-binding pocket. The apoM/S1P axis has been discovered to play a crucial role in the apoM signaling pathway through its ability to regulate glucose and lipid metabolism, vascular barrier homeostasis, inflammatory response and other pathological and physiological processes. Using the findings of previous studies, the present review aimed to summarize the regulation of apoM expression by various factors and its role in different physiological and pathological conditions, and provide a new perspective for the further treatment of these diseases.


Assuntos
Aterosclerose , Lisofosfolipídeos , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas M/genética , Apolipoproteínas M/metabolismo , Humanos , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Lisofosfolipídeos/metabolismo , Transdução de Sinais , Esfingosina/genética , Esfingosina/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 41(10): e468-e479, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34407633

RESUMO

Objective: ApoM enriches S1P (sphingosine-1-phosphate) within HDL (high-density lipoproteins) and facilitates the activation of the S1P1 (S1P receptor type 1) by S1P, thereby preserving endothelial barrier function. Many protective functions exerted by HDL in extravascular tissues raise the question of how S1P regulates transendothelial HDL transport. Approach and Results: HDL were isolated from plasma of wild-type mice, Apom knockout mice, human apoM transgenic mice or humans and radioiodinated to trace its binding, association, and transport by bovine or human aortic endothelial cells. We also compared the transport of fluorescently-labeled HDL or Evans Blue, which labels albumin, from the tail vein into the peritoneal cavity of apoE-haploinsufficient mice with (apoE-haploinsufficient mice with endothelium-specific knockin of S1P1) or without (control mice, ie, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1) endothelium-specific knockin of S1P1. The binding, association, and transport of HDL from Apom knockout mice and human apoM-depleted HDL by bovine aortic endothelial cells was significantly lower than that of HDL from wild-type mice and human apoM-containing HDL, respectively. The binding, uptake, and transport of 125I-HDL by human aortic endothelial cells was increased by an S1P1 agonist but decreased by an S1P1 inhibitor. Silencing of SR-BI (scavenger receptor BI) abrogated the stimulation of 125I-HDL transport by the S1P1 agonist. Compared with control mice, that is, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1, apoE-haploinsufficient mice with endothelium-specific knockin of S1P1 showed decreased transport of Evans Blue but increased transport of HDL from blood into the peritoneal cavity and SR-BI expression in the aortal endothelium. Conclusions: ApoM and S1P1 promote transendothelial HDL transport. Their opposite effect on transendothelial transport of albumin and HDL indicates that HDL passes endothelial barriers by specific mechanisms rather than passive filtration.


Assuntos
Apolipoproteínas M/metabolismo , Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Lipoproteínas HDL/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Transporte Biológico , Bovinos , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Permeabilidade , Placa Aterosclerótica , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Receptores de Esfingosina-1-Fosfato/genética
9.
Mol Biol Rep ; 49(2): 1171-1179, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34775573

RESUMO

BACKGROUND: To investigate the effects and mechanism of action of apolipoprotein M (ApoM) on the growth of breast cancer (BC) cells. METHODS AND RESULTS: Bioinformatics, cell experiments and animal experiments were used to verify the effect of ApoM on breast cancer cell lines and breast tumor growth in vivo. ApoM expression was significantly reduced in BC tissues, and patients with lower ApoM mRNA expression had a poorer prognosis (P < 0.0001). Besides, ApoM can partially inhibit the proliferative, migratory and invasive processes of BC cells. In vivo, the difference between ApoM-OE and NC groups was no significant. The level of vitamin D receptor (VDR) protein in MDA-MB-231 cells was increased by overexpression of ApoM (P < 0.05), while in MCF-7 cells, VDR levels decreased (P < 0.05). CONCLUSIONS: ApoM can partially inhibit the growth of BC cells. VDR may play a role, but is not the main pathway.


Assuntos
Apolipoproteínas M/metabolismo , Neoplasias da Mama/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas M/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Biologia Computacional/métodos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , RNA Mensageiro/genética , Receptores de Calcitriol/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Drug Dev Res ; 83(2): 397-406, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34418885

RESUMO

Propofol (PRO), a clinical potent intravenous anesthetic, plays a significant role in relieving inflammatory diseases by repressing the release of inflammatory cytokines. The present study was aimed to reveal a novel mechanism by which PRO alleviates acute lung injury (ALI). Lipopolysaccharide (LPS) was utilized to induce human pulmonary microvascular endothelial cells (HPMECs) so as to simulate the microenvironment of ALI, and the expression of apolipoprotein M (APOM) was examined with western blotting. Then, APOM was silenced and profopol was used to treat the LPS-injured HPMECs. The cell viability, migration, and apoptosis were respectively observed after the processes of cell counting kit-8, wound healing, transwell, and TUNEL assay. Meanwhile, the inflammatory response was detected by determining the contents of inflammatory cytokines. Subsequently, the relationship between phosphoinositide-3 kinase (PI3K)/protein kinase B (AKT) signaling pathway and PRO was analyzed by western blotting. PI3K/AKT inhibitor LY294002 was employed to evaluate whether the effects of PRO on LPS-challenged HPMECs injury were mediated by this pathway. Results revealed that APOM was notably downregulated in HPMECs after LPS exposure. PRO treatment promoted cell proliferation and migration while alleviated inflammation and apoptosis of LPS-treated HPMECs, which was reversed by APOM-downregulation. PRO brought about the upregulation of proteins in PI3K/AKT signaling pathway, and LY294002 intervention further accentuated the impacts of APOM-knockdown on LPS-challenged HPMECs injury. To conclude, PRO promotes migration and alleviates inflammation and apoptosis of LPS-treated HPMECs by PI3K/AKT signaling pathway via upregulating APOM, which laid an experimental foundation for the future study and clinical application of PRO.


Assuntos
Lesão Pulmonar Aguda , Propofol , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Apolipoproteínas M/metabolismo , Apolipoproteínas M/farmacologia , Apoptose , Citocinas/metabolismo , Células Endoteliais/metabolismo , Humanos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Lipopolissacarídeos/efeitos adversos , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Propofol/efeitos adversos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
11.
Int J Mol Sci ; 23(22)2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36430543

RESUMO

High-density lipoprotein (HDL)-bound apolipoprotein M/sphingosine 1-phosphate (ApoM/S1P) complex in cardiovascular diseases serves as a bridge between HDL and endothelial cells, maintaining a healthy endothelial barrier. To date, S1P and ApoM in patients with untreated heterozygous familial hypercholesterolemia (HeFH) have not been extensively studied. Eighty-one untreated patients with HeFH and 32 healthy control subjects were included in this study. Serum S1P, ApoM, sCD40L, sICAM-1, sVCAM-1, oxLDL, and TNFα concentrations were determined by ELISA. PON1 activities were measured spectrophotometrically. Lipoprotein subfractions were detected by Lipoprint. We diagnosed FH using the Dutch Lipid Clinic Network criteria. Significantly higher serum S1P and ApoM levels were found in HeFH patients compared to controls. S1P negatively correlated with large HDL and positively with small HDL subfractions in HeFH patients and the whole study population. S1P showed significant positive correlations with sCD40L and MMP-9 levels and PON1 arylesterase activity, while we found significant negative correlation between sVCAM-1 and S1P in HeFH patients. A backward stepwise multiple regression analysis showed that the best predictors of serum S1P were large HDL subfraction and arylesterase activity. Higher S1P and ApoM levels and their correlations with HDL subfractions and inflammatory markers in HeFH patients implied their possible role in endothelial protection.


Assuntos
Células Endoteliais , Hiperlipoproteinemia Tipo II , Humanos , Apolipoproteínas M , Células Endoteliais/metabolismo , Apolipoproteínas/metabolismo , Biomarcadores , Arildialquilfosfatase
12.
Int J Mol Sci ; 23(15)2022 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-35955730

RESUMO

High-density lipoproteins (HDL) undergo adverse remodeling and loss of function in the presence of comorbidities. We assessed the potential of lipid-lowering approaches (diet and rosuvastatin) to rescue hypercholesterolemia-induced HDL dysfunction. Hypercholesterolemia was induced in 32 pigs for 10 days. Then, they randomly received one of the 30-day interventions: (I) hypercholesterolemic (HC) diet; (II) HC diet + rosuvastatin; (III) normocholesterolemic (NC) diet; (IV) NC diet + rosuvastatin. We determined cholesterol efflux capacity (CEC), antioxidant potential, HDL particle number, HDL apolipoprotein content, LDL oxidation, and lipid levels. Hypercholesterolemia time-dependently impaired HDL function (−62% CEC, −11% antioxidant index (AOI); p < 0.01), increased HDL particles numbers 2.8-fold (p < 0.0001), reduced HDL-bound APOM (−23%; p < 0.0001), and increased LDL oxidation 1.7-fold (p < 0.0001). These parameters remained unchanged in animals on HC diet alone up to day 40, while AOI deteriorated up to day 25 (−30%). The switch to NC diet reversed HDL dysfunction, restored apolipoprotein M content and particle numbers, and normalized cholesterol levels at day 40. Rosuvastatin improved HDL, AOI, and apolipoprotein M content. Apolipoprotein A-I and apolipoprotein C-III remained unchanged. Lowering LDL-C levels with a low-fat diet rescues HDL CEC and antioxidant potential, while the addition of rosuvastatin enhances HDL antioxidant capacity in a pig model of hypercholesterolemia. Both strategies restore HDL-bound apolipoprotein M content.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Hipercolesterolemia , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Apolipoproteínas M , Colesterol/uso terapêutico , HDL-Colesterol , Dieta , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/etiologia , Lipoproteínas HDL/uso terapêutico , Lipoproteínas LDL , Modelos Animais , Rosuvastatina Cálcica/farmacologia , Rosuvastatina Cálcica/uso terapêutico , Suínos
13.
Circulation ; 141(18): 1463-1476, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32237898

RESUMO

BACKGROUND: Apo (apolipoprotein) M mediates the physical interaction between high-density lipoprotein (HDL) particles and sphingosine-1-phosphate (S1P). Apo M exerts anti-inflammatory and cardioprotective effects in animal models. METHODS: In a subset of PHFS (Penn Heart Failure Study) participants (n=297), we measured apo M by Enzyme-Linked ImmunoSorbent Assay (ELISA). We also measured total S1P by liquid chromatography-mass spectrometry and isolated HDL particles to test the association between apo M and HDL-associated S1P. We confirmed the relationship between apo M and outcomes using modified aptamer-based apo M measurements among 2170 adults in the PHFS and 2 independent cohorts: the Washington University Heart Failure Registry (n=173) and a subset of TOPCAT (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist Trial; n=218). Last, we examined the relationship between apo M and ≈5000 other proteins (SomaScan assay) to identify biological pathways associated with apo M in heart failure. RESULTS: In the PHFS, apo M was inversely associated with the risk of death (standardized hazard ratio, 0.56 [95% CI, 0.51-0.61]; P<0.0001) and the composite of death/ventricular assist device implantation/heart transplantation (standardized hazard ratio, 0.62 [95% CI, 0.58-0.67]; P<0.0001). This relationship was independent of HDL cholesterol or apo AI levels. Apo M remained associated with death (hazard ratio, 0.78 [95% CI, 0.69-0.88]; P<0.0001) and the composite of death/ventricular assist device/heart transplantation (hazard ratio, 0.85 [95% CI, 0.76-0.94]; P=0.001) in models that adjusted for multiple confounders. This association was present in both heart failure with reduced and preserved ejection fraction and was replicated in the Washington University cohort and a cohort with heart failure with preserved ejection fraction only (TOPCAT). The S1P and apo M content of isolated HDL particles strongly correlated (R=0.81, P<0.0001). The top canonical pathways associated with apo M were inflammation (negative association), the coagulation system (negative association), and liver X receptor/retinoid X receptor activation (positive association). The relationship with inflammation was validated with multiple inflammatory markers measured with independent assays. CONCLUSIONS: Reduced circulating apo M is independently associated with adverse outcomes across the spectrum of human heart failure. Further research is needed to assess whether the apo M/S1P axis is a suitable therapeutic target in heart failure.


Assuntos
Apolipoproteínas M/sangue , Insuficiência Cardíaca/sangue , Proteoma , Idoso , Biomarcadores/sangue , Regulação para Baixo , Feminino , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/mortalidade , Insuficiência Cardíaca/terapia , Humanos , Lipoproteínas HDL/sangue , Lisofosfolipídeos/sangue , Masculino , Pessoa de Meia-Idade , Prognóstico , Proteômica , Ensaios Clínicos Controlados Aleatórios como Assunto , Sistema de Registros , Medição de Risco , Fatores de Risco , Esfingosina/análogos & derivados , Esfingosina/sangue , Fatores de Tempo , Estados Unidos
14.
FASEB J ; 34(11): 14655-14670, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32918529

RESUMO

Hepatobiliary cholesterol handling, mediated by Niemann-Pick C1-like 1 protein (NPC1L1) and ABCG5/8, is well-known to contribute to the homeostasis of cholesterol. We attempted to elucidate the impact of hepatobiliary cholesterol handling on the homeostasis of sphingolipids and lysophospholipids, especially sphingosine 1-phosphate (S1P). We induced the overexpression of NPC1L1 or ABCG5/8 in the mouse liver. Hepatic NPC1L1 overexpression increased the plasma and hepatic S1P levels, while it decreased the biliary S1P levels, and all of these changes were inhibited by ezetimibe. The ability of HDL to activate Akt in the endothelial cells was augmented by hepatic NPC1L1 overexpression. NPC1L1-mediated S1P transport was confirmed by both in vitro and in vivo studies conducted using C17 S1P, an exogenous S1P analog. Upregulation of apolipoprotein M (apoM) was involved in these modulations, although apoM was not necessary for these modulations. Moreover, the increase in the plasma S1P levels also observed in ABCG5/8-overexpressing mice was dependent on the elevation of the plasma apoM levels. In regard to other sphingolipids and lysophospholipids, ceramides were similarly modulated by NPC1L1 to S1P, while other lipids were differently influenced by NPC1L1 or ABCG5/8 from S1P. Hepatobiliary cholesterol handling might also regulate the functional lipids, such as S1P.


Assuntos
Colesterol/metabolismo , Fígado/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Apolipoproteínas M/metabolismo , Ezetimiba/farmacologia , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fígado/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Esfingosina/metabolismo
15.
Nature ; 523(7560): 342-6, 2015 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-26053123

RESUMO

Lipid mediators influence immunity in myriad ways. For example, circulating sphingosine-1-phosphate (S1P) is a key regulator of lymphocyte egress. Although the majority of plasma S1P is bound to apolipoprotein M (ApoM) in the high-density lipoprotein (HDL) particle, the immunological functions of the ApoM-S1P complex are unknown. Here we show that ApoM-S1P is dispensable for lymphocyte trafficking yet restrains lymphopoiesis by activating the S1P1 receptor on bone marrow lymphocyte progenitors. Mice that lacked ApoM (Apom(-/-)) had increased proliferation of Lin(-) Sca-1(+) cKit(+) haematopoietic progenitor cells (LSKs) and common lymphoid progenitors (CLPs) in bone marrow. Pharmacological activation or genetic overexpression of S1P1 suppressed LSK and CLP cell proliferation in vivo. ApoM was stably associated with bone marrow CLPs, which showed active S1P1 signalling in vivo. Moreover, ApoM-bound S1P, but not albumin-bound S1P, inhibited lymphopoiesis in vitro. Upon immune stimulation, Apom(-/-) mice developed more severe experimental autoimmune encephalomyelitis, characterized by increased lymphocytes in the central nervous system and breakdown of the blood-brain barrier. Thus, the ApoM-S1P-S1P1 signalling axis restrains the lymphocyte compartment and, subsequently, adaptive immune responses. Unique biological functions imparted by specific S1P chaperones could be exploited for novel therapeutic opportunities.


Assuntos
Apolipoproteínas/metabolismo , Sistema Nervoso Central/patologia , Lipoproteínas HDL/metabolismo , Linfócitos/citologia , Linfócitos/metabolismo , Linfopoese , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Apolipoproteínas/deficiência , Apolipoproteínas/genética , Apolipoproteínas M , Barreira Hematoencefálica/patologia , Movimento Celular , Proliferação de Células/genética , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Cloridrato de Fingolimode/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Linfócitos/imunologia , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/metabolismo , Lisofosfolipídeos/agonistas , Lisofosfolipídeos/sangue , Lisofosfolipídeos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Esfingosina/agonistas , Esfingosina/sangue , Esfingosina/genética , Esfingosina/metabolismo
16.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33805921

RESUMO

Colorectal cancer (CRC) is a highly prevalent malignancy with multifactorial etiology, which includes metabolic alterations as contributors to disease development. Studies have shown that lipid status disorders are involved in colorectal carcinogenesis. In line with this, previous studies have also suggested that the serum high-density lipoprotein cholesterol (HDL-C) level decreases in patients with CRC, but more recently, the focus of investigations has shifted toward the exploration of qualitative properties of HDL in this malignancy. Herein, a comprehensive overview of available evidences regarding the putative role of HDL in CRC will be presented. We will analyze existing findings regarding alterations of HDL-C levels but also HDL particle structure and distribution in CRC. In addition, changes in HDL functionality in this malignancy will be discussed. Moreover, we will focus on the genetic regulation of HDL metabolism, as well as the involvement of HDL in disturbances of cholesterol trafficking in CRC. Finally, possible therapeutic implications related to HDL will be presented. Given the available evidence, future studies are needed to resolve all raised issues concerning the suggested protective role of HDL in CRC, its presumed function as a biomarker, and eventual therapeutic approaches based on HDL.


Assuntos
Neoplasias Colorretais/metabolismo , Lipoproteínas HDL/metabolismo , Animais , Apolipoproteína A-I/metabolismo , Apolipoproteínas M/metabolismo , Arildialquilfosfatase/metabolismo , Biomarcadores/metabolismo , Carcinogênese , Colesterol/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , HDL-Colesterol/metabolismo , Homeostase , Humanos , Camundongos , Polimorfismo de Nucleotídeo Único , Medicina de Precisão , Receptores Depuradores Classe B/metabolismo
17.
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
18.
J Cell Mol Med ; 24(15): 8836-8848, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32597038

RESUMO

This study aims to explore the possible mechanism of TUG1 regulating ApoM in AS. To this end, expression levels of TUG1 and ApoM were measured in high fat dieted C57BL/6J mice, normal dieted C57BL/6J mice, ob/ob mice and db/db mice. LV-TUG1 or sh-TUG1 was injected into C57BL/6J mice before isolating peritoneal macrophages to measure cholesterol efflux (CE) and expression levels of ABCA1, ABCG1 and SR-BI. Meanwhile, CE in RAW264.7 cells was also measured after cell transfection. Dual luciferase reporter assay and anti-AGO2 RIP were applied to verify the relationship among TUG1, FXR1 and miR-92a. Total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterin (LDL-C), high-density lipoprotein cholesterol (HDL-C) as well as expressions of inflammatory cytokines (TNF-α, IL-1ß and IL-6) in plasma were measured. Knock-down or expressed TUG1, FXR1 or miR-92a in NCTC 1469 cells or in ApoE-/- AS mice to determine the alteration on ApoM and plaque size. TUG1 was highly expressed while ApoM was down-regulated in high fat dieted C57BL/6J mice, b/ob and db/db mice. Overexpression of TUG1 could reduce the expression of ApoM, ABCA1 and ABCG1 in addition to slowing down CE rate. Reversed expression pattern was found in cells with knock-down of TUG1. TUG1 can compete with FXR1 to bind miR-92a. FXR1 negatively target ApoM. Overexpression of TUG1 in ApoE-/- mice can increase plaque size and enhance macrophage contents accordingly. TUG1 can inhibit ApoM in both liver tissues and plasma to inhibit CE through regulating miR-92a/ FXR1 axis. TUG1 is a promising target for AS treatment.


Assuntos
Apolipoproteínas M/genética , Aterosclerose/genética , Aterosclerose/patologia , Regulação da Expressão Gênica , MicroRNAs/genética , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas M/metabolismo , Aterosclerose/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Genes Reporter , Mediadores da Inflamação/metabolismo , Fígado/metabolismo , Fígado/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Células RAW 264.7 , Interferência de RNA , Proteínas de Ligação a RNA/metabolismo
19.
FASEB J ; 33(4): 5181-5195, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30629456

RESUMO

Because the association between sphingosine 1-phosphate (S1P)/apolipoprotein M (ApoM) and chronic kidney diseases has not been established, we investigated the involvement of S1P/ApoM in the phenotypes of IgA nephropathy in hyper-IgA (HIGA) mice. The overexpression of ApoM in adenoviral gene transfer ameliorated the phenotypes of IgA nephropathy in HIGA mice, whereas the knockdown of ApoM with siRNA caused deterioration. When ApoM-overexpressing HIGA mice were treated with VPC23019, an antagonist against S1P receptor 1 (S1P1) and 3 (S1P3), we observed that the protective effects of ApoM were reversed, whereas JTE013, an antagonist against S1P2, did not inhibit the effects. We also found that S1P bound to albumin accelerated the proliferation of MES13 cells and the fibrotic changes of HK2 cells, which were inhibited by JTE013, whereas S1P bound to ApoM suppressed these changes, which were inhibited by VPC23019. These results suggest that S1P bound to ApoM possesses properties protective against the phenotypes of IgA nephropathy through S1P1 and S1P3, whereas S1P bound to albumin exerts deteriorating effects through S1P2. ApoM may be useful as a therapeutic target to treat or retard the progression of IgA nephropathy.-Kurano, M., Tsuneyama, K., Morimoto, Y., Nishikawa, M., Yatomi, Y. Apolipoprotein M suppresses the phenotypes of IgA nephropathy in hyper-IgA mice.


Assuntos
Apolipoproteínas M/uso terapêutico , Glomerulonefrite por IGA/sangue , Glomerulonefrite por IGA/tratamento farmacológico , Imunoglobulina A/sangue , Lisofosfolipídeos/uso terapêutico , Esfingosina/análogos & derivados , Animais , Western Blotting , Linhagem Celular , Creatinina/sangue , Creatinina/urina , Feminino , Glomerulonefrite por IGA/metabolismo , Glomerulonefrite por IGA/urina , Imunoglobulina A/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingosina/uso terapêutico , Receptores de Esfingosina-1-Fosfato/antagonistas & inibidores , Receptores de Esfingosina-1-Fosfato/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 39(11): 2411-2430, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31554421

RESUMO

OBJECTIVE: Clinical evidence has linked low HDL (high-density lipoprotein) cholesterol levels with high cardiovascular disease risk; however, its significance as a therapeutic target remains unestablished. We hypothesize that HDLs functional heterogeneity is comprised of metabolically distinct proteins, each on distinct HDL sizes and that are affected by diet. Approach and Results: Twelve participants were placed on 2 healthful diets high in monounsaturated fat or carbohydrate. After 4 weeks on each diet, participants completed a metabolic tracer study. HDL was isolated by Apo (apolipoprotein) A1 immunopurification and separated into 5 sizes. Tracer enrichment and metabolic rates for 8 HDL proteins-ApoA1, ApoA2, ApoC3, ApoE, ApoJ, ApoL1, ApoM, and LCAT (lecithin-cholesterol acyltransferase)-were determined by parallel reaction monitoring and compartmental modeling, respectively. Each protein had a unique, size-specific distribution that was not altered by diet. However, carbohydrate, when replacing fat, increased the fractional catabolic rate of ApoA1 and ApoA2 on alpha3 HDL; ApoE on alpha3 and alpha1 HDL; and ApoM on alpha2 HDL. Additionally, carbohydrate increased the production of ApoC3 on alpha3 HDL and ApoJ and ApoL1 on the largest alpha0 HDL. LCAT was the only protein studied that diet did not affect. Finally, global proteomics showed that diet did not alter the distribution of the HDL proteome across HDL sizes. CONCLUSIONS: This study demonstrates that HDL in humans is composed of a complex system of proteins, each with its own unique size distribution, metabolism, and diet regulation. The carbohydrate-induced hypercatabolic state of HDL proteins may represent mechanisms by which carbohydrate alters the cardioprotective properties of HDL.


Assuntos
Dieta Hiperlipídica , Carboidratos da Dieta/administração & dosagem , Gorduras Insaturadas na Dieta/administração & dosagem , Lipoproteínas HDL/sangue , Proteoma , Apolipoproteína A-I/sangue , Apolipoproteína A-II/sangue , Apolipoproteína C-III/sangue , Apolipoproteína L1/sangue , Apolipoproteínas E/sangue , Apolipoproteínas M/sangue , Clusterina/sangue , Feminino , Humanos , Lipoproteínas HDL/química , Masculino , Fosfatidilcolina-Esterol O-Aciltransferase/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA