Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Clin Transl Oncol ; 22(11): 1952-1962, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32306242

RESUMEN

Apolipoproteins, the key components of lipoproteins, play vital roles in the combination and transportation of lipids. Numerous research articles have accumulated solid evidence that lipoproteins are closely related to various types of tumorigenesis. In this review, we focused on the associations between several apolipoproteins and breast carcinoma and distinguished the effects and significance of apolipoproteins in different locations to validate their roles in breast carcinoma development. For example, apoD and apoE in serum are viewed as risk factors for breast carcinoma. ApoD, apoE and apoA-I in mammary tissues inhibit tumor growth. Moreover, apoB, apoJ and apoA-I have the potential to function as diagnostic or prognostic markers in the clinic. ApoEdp and apoJ treatment on breast carcinoma could significantly restrict tumor growth. In general, the aim of this review was to further analyze the associations between some members of the apolipoprotein family and breast cancer.


Asunto(s)
Apolipoproteínas/fisiología , Neoplasias de la Mama/etiología , Proteínas Portadoras/fisiología , Neoplasias de la Mama/patología , Femenino , Humanos , Metabolismo de los Lípidos
2.
Microbiol Immunol ; 63(10): 401-406, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31342548

RESUMEN

The family Flaviviridae comprises four genera, namely, Flavivirus, Pestivirus, Pegivirus, and Hepacivirus. These viruses have similar genome structures, but the genomes of Pestivirus and Flavivirus encode the secretory glycoproteins Erns and NS1, respectively. Erns plays an important role in virus particle formation and cell entry, whereas NS1 participates in the formation of replication complexes and virus particles. Conversely, apolipoproteins are known to participate in the formation of infectious particles of hepatitis C virus (HCV) and various secretory glycoproteins play a similar role in HCV particles formation, suggesting that there is no strong specificity for the function of secretory glycoproteins in infectious-particle formation. In addition, recent studies have shown that host-derived apolipoproteins and virus-derived Erns and NS1 play comparable roles in infectious-particle formation of both HCV and pestiviruses. In this review, we summarize the roles of secretory glycoproteins in the formation of Flaviviridae virus particles.


Asunto(s)
Apolipoproteínas/fisiología , Infecciones por Flaviviridae/virología , Flaviviridae , Glicoproteínas/fisiología , Virión/fisiología , Flaviviridae/patogenicidad , Flaviviridae/fisiología , Interacciones Microbiota-Huesped , Humanos , Ensamble de Virus
3.
Uirusu ; 68(1): 63-70, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-31105136

RESUMEN

Hepatitis C virus (HCV) infects over 170 million people worldwide and is a major cause of life-threatening liver diseases such as liver cirrhosis and hepatocellular carcinoma. In current research, we aimed to clarify the mechanism of hepatic tropism of HCV infection. Although non-hepatic cells could not permit replication of HCV RNA, exogenous expression of liver specific miRNA, miR-122 facilitated efficient replication of viral RNA through direct interaction with 5'UTR of viral genome, indicating that miR-122 is one of the key determinants for hepatic tropism of HCV infection. In spite of efficient replication of viral RNA, formation of infectious particles was not observed in non-hepatic cells exogenously expressing miR-122. We found that expression of apolipoprotein E (ApoE) facilitated the formation of infectious HCV particles in non-hepatic cells, indicating that not only miR-122 but also ApoE participate in tissue tropism of HCV infection. To understand the exact roles of miR-122 and apolipoproteins in hepatic tropism of HCV, we established miR-122 and ApoB/ApoE knockout (KO) Huh7 cells, respectively. Although slight increase of intracellular HCV RNA and infectious titers in the culture supernatants was observed, propagation of HCV was impaired in miR-122 KO Huh7 cells. After serial passages of HCV in miR-122 KO cells, we obtained an adaptive mutant that possessed G28A substitutions in the 5'UTR of the HCV genome and exhibited efficient translation and replication in both miR-122 KO Huh7 and non-hepatic cells without exogenous expression of miR-122. These results suggest that HCV mutants replicating in non-hepatic cells in an miR-122-independent manner participate in the induction of extrahepatic manifestations in chronic hepatitis C patients. Deficiency of both ApoB and ApoE strongly inhibited the formation of infectious HCV particles. Interestingly, expression not only of ApoE but also of ApoA or ApoC could rescue the production of infectious HCV particles in ApoB/ApoE KO cells, suggesting that exchangeable apolipoproteins redundantly participate in the formation of infectious HCV particles.


Asunto(s)
Hepacivirus/genética , Hepacivirus/fisiología , Hígado/virología , Tropismo Viral , Apolipoproteínas/metabolismo , Apolipoproteínas/fisiología , Genoma Viral/genética , Humanos , Hígado/metabolismo , MicroARNs , Tropismo Viral/genética , Virión/genética , Replicación Viral/genética
4.
World J Microbiol Biotechnol ; 34(1): 11, 2017 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-29255943

RESUMEN

Pullulan produced by Aureobasidium pullulans presents various applications in food manufacturing and pharmaceutical industry. However, the pullulan biosynthesis mechanism remains unclear. This work proposed a pathway suggesting that heavy oil and melanin may correlate with pullulan production. The effects of overexpression or deletion of genes encoding apolipoprotein, UDPG-pyrophosphorylase, glucosyltransferase, and α-phosphoglucose mutase on the production of pullulan, heavy oil, and melanin were examined. Pullulan production increased by 16.93 and 8.52% with the overexpression of UDPG-pyrophosphorylase and apolipoprotein genes, respectively. Nevertheless, the overexpression or deletion of other genes exerted little effect on pullulan biosynthesis. Heavy oil production increased by 146.30, 64.81, and 33.33% with the overexpression of UDPG-pyrophosphorylase, α-phosphoglucose mutase, and apolipoprotein genes, respectively. Furthermore, the syntheses of pullulan, heavy oil, and melanin can compete with one another. This work may provide new guidance to improve the production of pullulan, heavy oil, and melanin through genetic approach.


Asunto(s)
Apolipoproteínas/genética , Apolipoproteínas/fisiología , Ascomicetos/genética , Ascomicetos/metabolismo , Glucanos/biosíntesis , Melaninas/biosíntesis , Aceites/metabolismo , Ascomicetos/enzimología , Metabolismo de los Hidratos de Carbono , Activación Enzimática , Fermentación , Proteínas Fúngicas/genética , Proteínas Fúngicas/fisiología , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación Fúngica de la Expresión Génica , Genes Fúngicos/genética , Genes Fúngicos/fisiología , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Redes y Vías Metabólicas/genética , Redes y Vías Metabólicas/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transformación Genética , UTP-Glucosa-1-Fosfato Uridililtransferasa/genética , UTP-Glucosa-1-Fosfato Uridililtransferasa/metabolismo
5.
J Am Soc Nephrol ; 28(11): 3227-3238, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28696248

RESUMEN

Population genetic approaches have uncovered a strong association between kidney diseases and two sequence variants of the APOL1 gene, called APOL1 risk variant G1 and variant G2, compared with the nonrisk G0 allele. However, the mechanism whereby these variants lead to disease manifestation and, in particular, whether this involves an intracellular or extracellular pool of APOL1 remains unclear. Herein, we show a predominantly intracellular localization of APOL1 G0 and the renal risk variants, which localized to membranes of the endoplasmic reticulum in podocyte cell lines. This localization did not depend on the N-terminal signal peptide that mediates APOL1 secretion into the circulation. Additionally, a fraction of these proteins localized to structures surrounding mitochondria. In vitro overexpression of G1 or G2 lacking the signal peptide inhibited cell viability, triggered phosphorylation of stress-induced kinases, increased the phosphorylation of AMP-activated protein kinase, reduced intracellular potassium levels, and reduced mitochondrial respiration rates. These findings indicate that functions at intracellular membranes, specifically those of the endoplasmic reticulum and mitochondria, are crucial factors in APOL1 renal risk variant-mediated cell injury.


Asunto(s)
Apolipoproteínas , Metabolismo Energético , Lipoproteínas HDL , Apolipoproteína L1 , Apolipoproteínas/análisis , Apolipoproteínas/genética , Apolipoproteínas/fisiología , Células Cultivadas , Retículo Endoplásmico/química , Variación Genética , Humanos , Lipoproteínas HDL/análisis , Lipoproteínas HDL/genética , Lipoproteínas HDL/fisiología , Membranas Mitocondriales/química , Factores de Riesgo
6.
J Endocrinol ; 233(2): R95-R107, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28314771

RESUMEN

It is well appreciated that high-density lipoprotein (HDL) and bone physiology and pathology are tightly linked. Studies, primarily in mouse models, have shown that dysfunctional and/or disturbed HDL can affect bone mass through many different ways. Specifically, reduced HDL levels have been associated with the development of an inflammatory microenvironment that affects the differentiation and function of osteoblasts. In addition, perturbation in metabolic pathways of HDL favors adipoblastic differentiation and restrains osteoblastic differentiation through, among others, the modification of specific bone-related chemokines and signaling cascades. Increased bone marrow adiposity also deteriorates bone osteoblastic function and thus bone synthesis, leading to reduced bone mass. In this review, we present the current knowledge and the future directions with regard to the HDL-bone mass connection. Unraveling the molecular phenomena that underline this connection will promote the deeper understanding of the pathophysiology of bone-related pathologies, such as osteoporosis or bone metastasis, and pave the way toward the development of novel and more effective therapies against these conditions.


Asunto(s)
Huesos/fisiología , Lipoproteínas HDL/fisiología , Adiposidad , Animales , Apolipoproteína A-I/fisiología , Apolipoproteínas/fisiología , Apolipoproteínas E/fisiología , Densidad Ósea , Médula Ósea/fisiología , Huesos/metabolismo , Cartílago/fisiología , Diferenciación Celular , Modelos Animales de Enfermedad , Homeostasis , Humanos , Lipoproteínas HDL/metabolismo , Ratones , Obesidad , Osteoblastos/fisiología , Osteoclastos/fisiología , Osteogénesis
7.
Curr Rheumatol Rep ; 19(3): 13, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28265848

RESUMEN

PURPOSE OF REVIEW: Systemic lupus erythematosus (SLE) confers up to a 50-fold increased risk of cardiovascular disease (CVD), and African Americans with SLE experience accelerated damage accrual and doubled cardiovascular risk when compared to their European American counterparts. RECENT FINDINGS: Genome-wide association studies have identified a substantial signal at 22q13, now assigned to variation at apolipoprotein L1 (APOL1), which has associated with progressive nondiabetic nephropathy, cardiovascular disease, and many immune-associated renal diseases, including lupus nephritis. We contend that alterations in crucial APOL1 intracellular pathways may underpin associated disease states based on structure-functional differences between variant and ancestral forms. While ancestral APOL1 may be a key driver of autophagy, nonconserved primary structure changes result in a toxic gain of function with attenuation of autophagy and an unsupervised pore-forming feature. Thus, the divergent intracellular biological pathways of ancestral and variant APOL1 may explain a worsened prognosis as demonstrated in SLE.


Asunto(s)
Apolipoproteínas/genética , Lipoproteínas HDL/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Apolipoproteína L1 , Apolipoproteínas/fisiología , Autofagia/genética , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/inmunología , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Lipoproteínas HDL/fisiología , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología
8.
J Am Soc Nephrol ; 28(4): 1117-1130, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27864431

RESUMEN

APOL1 harbors C-terminal sequence variants (G1 and G2), which account for much of the increased risk for kidney disease in sub-Saharan African ancestry populations. Expression of the risk variants has also been shown to cause injury to podocytes and other cell types, but the underlying mechanisms are not understood. We used Drosophila melanogaster and Saccharomyces cerevisiae to help clarify these mechanisms. Ubiquitous expression of the human APOL1 G1 and G2 disease risk alleles caused near-complete lethality in D. melanogaster, with no effect of the G0 nonrisk APOL1 allele, corresponding to the pattern of human disease risk. We also observed a congruent pattern of cellular damage with tissue-specific expression of APOL1. In particular, expression of APOL1 risk variants in D. melanogaster nephrocytes caused cell-autonomous accumulation of the endocytic tracer atrial natriuretic factor-red fluorescent protein at early stages and nephrocyte loss at later stages. We also observed differential toxicity of the APOL1 risk variants compared with the APOL1 nonrisk variants in S. cerevisiae, including impairment of vacuole acidification. Yeast strains defective in endosomal trafficking or organelle acidification but not those defective in autophagy displayed augmented APOL1 toxicity with all isoforms. This pattern of differential injury by the APOL1 risk alleles compared with the nonrisk alleles across evolutionarily divergent species is consistent with an impairment of conserved core intracellular endosomal trafficking processes. This finding should facilitate the identification of cell injury pathways and corresponding therapeutic targets of interest in these amenable experimental platforms.


Asunto(s)
Apolipoproteínas/metabolismo , Apolipoproteínas/fisiología , Muerte Celular/fisiología , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/fisiología , Alelos , Animales , Apolipoproteína L1 , Apolipoproteínas/genética , Drosophila melanogaster/citología , Humanos , Concentración de Iones de Hidrógeno , Lipoproteínas HDL/genética , Transporte de Proteínas , Saccharomyces cerevisiae/citología
10.
Rinsho Byori ; 64(2): 186-92, 2016 Feb.
Artículo en Japonés | MEDLINE | ID: mdl-27311283

RESUMEN

We have studied the physiological function of four apolipoproteins. First, apo A-I is a major component of HDL and plays a crucial role in reverse cholesterol transport. The lipid-poor apo A-I concentration in plasma was significantly increased in patients with coronary artery disease compared with healthy controls, which may be caused by the impairment of the reverse cholesterol transport pathway. Second, the plasma A-IV concentration was significantly elevated in uremic patients, and we revealed the mechanism of apo A-IV accumulation in plasma using a rat model. Third, apo B48 is associated with lipid absorption in the intestinal epithelium, but the lymph apo B48 output was not changed during the absorption of mid-chain triglycerides, unlike apo A-IV. Fourth, we showed for the first time that the cerebrospinal apo E level was reduced in early-onset Alzheimer's disease and increased in a late-onset group. Taken together, apolipoproteins show various functions via the regulation of lipid metabolism. We have also studied the effect of cytokines on atherosclerosis using cytokine knockout mice. TNF-α and IL-1ß increased the number and size of atherosclerotic lesions, but IFN-γ attenuated the lesions. Plaque formation is influenced by not only the cholesterol level in plasma but also cytokine levels and other unknown factors. It may be of no merit to give cholesterol-lowering drugs to hypercholesterolemic patients without plaque. It is, thus, strongly expected that a biomarker which can predict the presence of plaque will be developed in the future.


Asunto(s)
Apolipoproteínas/fisiología , Aterosclerosis/etiología , Metabolismo de los Lípidos , Animales , Apolipoproteína A-I , Apolipoproteína B-48 , Apolipoproteínas A , Apolipoproteínas E , Enfermedad de la Arteria Coronaria/etiología , Citocinas/fisiología , Modelos Animales de Enfermedad , Interferón gamma/fisiología , Interleucina-1beta/fisiología , Ratones , Ratas , Factor de Necrosis Tumoral alfa/fisiología
11.
Proc Natl Acad Sci U S A ; 113(4): 830-7, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26699492

RESUMEN

Two specific genetic variants of the apolipoprotein L1 (APOL1) gene are responsible for the high rate of kidney disease in people of recent African ancestry. Expression in cultured cells of these APOL1 risk variants, commonly referred to as G1 and G2, results in significant cytotoxicity. The underlying mechanism of this cytotoxicity is poorly understood. We hypothesized that this cytotoxicity is mediated by APOL1 risk variant-induced dysregulation of intracellular signaling relevant for cell survival. To test this hypothesis, we conditionally expressed WT human APOL1 (G0), the APOL1 G1 variant, or the APOL1 G2 variant in human embryonic kidney cells (T-REx-293) using a tetracycline-mediated (Tet-On) system. We found that expression of either G1 or G2 APOL1 variants increased apparent cell swelling and cell death compared with G0-expressing cells. These manifestations of cytotoxicity were preceded by G1 or G2 APOL1-induced net efflux of intracellular potassium as measured by X-ray fluorescence, resulting in the activation of stress-activated protein kinases (SAPKs), p38 MAPK, and JNK. Prevention of net K(+) efflux inhibited activation of these SAPKs by APOL1 G1 or G2. Furthermore, inhibition of SAPK signaling and inhibition of net K(+) efflux abrogated cytotoxicity associated with expression of APOL1 risk variants. These findings in cell culture raise the possibility that nephrotoxicity of APOL1 risk variants may be mediated by APOL1 risk variant-induced net loss of intracellular K(+) and subsequent induction of stress-activated protein kinase pathways.


Asunto(s)
Apolipoproteínas/genética , Transporte Iónico/genética , Enfermedades Renales/genética , Lipoproteínas HDL/genética , Proteínas Quinasas Activadas por Mitógenos/fisiología , Mutación Missense , Potasio/metabolismo , Sustitución de Aminoácidos , Apolipoproteína L1 , Apolipoproteínas/fisiología , Población Negra/genética , Muerte Celular , Tamaño de la Célula , Receptor gp130 de Citocinas/biosíntesis , Receptor gp130 de Citocinas/genética , Progresión de la Enfermedad , Activación Enzimática , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Enfermedades Renales/etnología , Lipoproteínas HDL/fisiología , Sistema de Señalización de MAP Quinasas , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/metabolismo , Riesgo , Factor de Transcripción STAT3/metabolismo , Transfección
12.
PLoS One ; 10(7): e0134529, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26226615

RESUMEN

Apolipoprotein E (ApoE), an exchangeable apolipoprotein, is necessary for production of infectious Hepatitis C virus (HCV) particles. However, ApoE is not the only liver-expressed apolipoprotein and the role of other apolipoproteins for production of infectious HCV progeny is incompletely defined. Therefore, we quantified mRNA expression of human apolipoproteins in primary human hepatocytes. Subsequently, cDNAs encoding apolipoproteins were expressed in 293T/miR-122 cells to explore if they complement HCV virus production in cells that are non-permissive due to limiting endogenous levels of human apolipoproteins. Primary human hepatocytes expressed high mRNA levels of ApoA1, A2, C1, C3, E, and H. ApoA4, A5, B, D, F, J, L1, L2, L3, L4, L6, M, and O were expressed at intermediate levels, and C2, C4, and L5 were not detected. All members of the ApoA and ApoC family of lipoproteins complemented HCV virus production in HCV transfected 293T/miR-122 cells, albeit with significantly lower efficacy compared with ApoE. In contrast, ApoD expression did not support production of infectious HCV. Specific infectivity of released particles complemented with ApoA family members was significantly lower compared with ApoE. Moreover, the ratio of extracellular to intracellular infectious virus was significantly higher for ApoE compared to ApoA2 and ApoC3. Since apolipoproteins complementing HCV virus production share amphipathic alpha helices as common structural features we altered the two alpha helices of ApoC1. Helix breaking mutations in both ApoC1 helices impaired virus assembly highlighting a critical role of alpha helices in apolipoproteins supporting HCV assembly. In summary, various liver expressed apolipoproteins with amphipathic alpha helices complement HCV virus production in human non liver cells. Differences in the efficiency of virus assembly, the specific infectivity of released particles, and the ratio between extracellular and intracellular infectivity point to distinct characteristics of these apolipoproteins that influence HCV assembly and cell entry. This will guide future research to precisely pinpoint how apolipoproteins function during virus assembly and cell entry.


Asunto(s)
Apolipoproteínas/fisiología , Hepacivirus/fisiología , Hepatocitos/metabolismo , Replicación Viral/fisiología , Línea Celular , Hepacivirus/patogenicidad , Humanos , Virulencia
14.
DNA Cell Biol ; 34(8): 550-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26057873

RESUMEN

To explore the anti-inflammatory effect of apolipoprotein M (apoM) on regulation of tumor necrosis factor-α (TNF-α)-induced expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) and further investigate the molecular mechanism of apoM in this process. We found that TNF-α could decrease expression of apoM and inhibitor of NF-κB-α (IκBα) in HepG2 cells. Overexpression of apoM caused a significant decrease of ICAM-1 and VCAM-1 expression, while it caused a significant increase of IκBα expression in HepG2 cells. Furthermore, the treatment with TNF-α could increase ICAM-1 and VCAM-1 expression, decrease IκBα protein expression, and increase nuclear factor-κB (NF-κB) activity, and these effects were markedly enhanced by small interfering RNA (siRNA)-mediated silencing of apoM in HepG2 cells. Our findings demonstrated that apoM suppressed TNF-α-induced expression of ICAM-1 and VCAM-1 through inhibiting the activity of NF-κB.


Asunto(s)
Apolipoproteínas/fisiología , Molécula 1 de Adhesión Intercelular/genética , Lipocalinas/fisiología , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/genética , Apolipoproteínas M , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
Drug Des Devel Ther ; 9: 2375-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25987835

RESUMEN

Apolipoprotein M (ApoM) is predominantly located in the high-density lipoprotein in human plasma. It has been demonstrated that ApoM expression could be regulated by several crucial nuclear receptors that are involved in the bile acid metabolism. In the present study, by combining gene-silencing experiments, overexpression studies, and chromatin immunoprecipitation assays, we showed that ApoM positively regulated liver receptor homolog-1 (LRH-1) gene expression via direct binding to an LRH-1 promoter region (nucleotides -406/ -197). In addition, we investigated the effects of farnesoid X receptor agonist GW4064 on hepatic ApoM expression in vitro. In HepG2 cell cultures, both mRNA and protein levels of ApoM and LRH-1 were decreased in a time-dependent manner in the presence of 1 µM GW4064, and the inhibition effect was gradually attenuated after 24 hours. In conclusion, our findings present supportive evidence that ApoM is a regulator of human LRH-1 transcription, and further reveal the importance of ApoM as a critical regulator of bile acids metabolism.


Asunto(s)
Apolipoproteínas/fisiología , Lipocalinas/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas M , Ácidos y Sales Biliares/metabolismo , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Células Hep G2 , Humanos , Isoxazoles/farmacología , Lipocalinas/genética , Lipocalinas/metabolismo , Hígado/metabolismo , Plásmidos/genética , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/biosíntesis , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Transfección
17.
Turk J Med Sci ; 45(5): 1004-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26738339

RESUMEN

This review highlight the similarities in the pathogenesis between Alzheimer disease and age-related macular degeneration. All studies published between 1990 and 2014 were reviewed to identify the common pathological pathways. Alzheimer disease and age-related macular degeneration share common features such as vitronectin and amyloid-ß accumulation, increased oxidative stress, and apolipoprotein and complement activation pathways, which are reviewed as histologic and immunologic common features.


Asunto(s)
Enfermedad de Alzheimer/etiología , Degeneración Macular/etiología , Factores de Edad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/fisiología , Apolipoproteínas/fisiología , Activación de Complemento , Humanos , Degeneración Macular/metabolismo , Degeneración Macular/patología , Estrés Oxidativo/fisiología , Vitronectina/fisiología
18.
Proc Natl Acad Sci U S A ; 111(20): E2130-9, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24808134

RESUMEN

ApolipoproteinL1 (APOL1) protects humans and some primates against several African trypanosomes. APOL1 genetic variants strongly associated with kidney disease in African Americans have additional trypanolytic activity against Trypanosoma brucei rhodesiense, the cause of acute African sleeping sickness. We combined genetic, physiological, and biochemical studies to explore coevolution between the APOL1 gene and trypanosomes. We analyzed the APOL1 sequence in modern and archaic humans and baboons along with geographic distribution in present day Africa to understand how the kidney risk variants evolved. Then, we tested Old World monkey, human, and engineered APOL1 variants for their ability to kill human infective trypanosomes in vivo to identify the molecular mechanism whereby human trypanolytic APOL1 variants evade T. brucei rhodesiense virulence factor serum resistance-associated protein (SRA). For one APOL1 kidney risk variant, a two-residue deletion of amino acids 388 and 389 causes a shift in a single lysine residue that mimics the Old World monkey sequence, which augments trypanolytic activity by preventing SRA binding. A second human APOL1 kidney risk allele, with an amino acid substitution that also restores sequence alignment with Old World monkeys, protected against T. brucei rhodesiense due in part to reduced SRA binding. Both APOL1 risk variants induced tissue injury in murine livers, the site of transgenic gene expression. Our study shows that both genetic variants of human APOL1 that protect against T. brucei rhodesiense have recapitulated molecular signatures found in Old World monkeys and raises the possibility that APOL1 variants have broader innate immune activity that extends beyond trypanosomes.


Asunto(s)
Apolipoproteínas/genética , Evolución Biológica , Resistencia a la Enfermedad/genética , Lipoproteínas HDL/genética , Tripanosomiasis Africana/genética , África , Alelos , Animales , Apolipoproteína L1 , Apolipoproteínas/fisiología , Frecuencia de los Genes , Geografía , Haplotipos , Humanos , Lipoproteínas HDL/fisiología , Lisina/genética , Mandrillus , Ratones , Ratones Transgénicos , Modelos Teóricos , Papio/genética , Polimorfismo Genético , Trypanosoma brucei rhodesiense
19.
Best Pract Res Clin Endocrinol Metab ; 28(3): 281-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24840259

RESUMEN

Considerable progress has been achieved in the treatment of dyslipidemias. However, half of cardiovascular events occur in individuals with average or low cholesterol levels and there is still a considerable residual risk with 70% of patients having an event despite statin treatment. In the era of personalized medicine there is increased interest in the incorporation of individual biomarkers in risk score algorithms in order to improve cardiovascular risk stratification followed by the prompt initiation of preventive measures. Since the 2001 third report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation and Treatment on High Blood Cholesterol in Adults (ATP III) several studies have evaluated the prognostic value of lipid related biomarkers such as non-HDL-cholesterol, apolipoprotein B, apolipoprotein B/apolipoprotein A1 ratio, lipoprotein(a), lipoprotein-associated phospholipase A2, and C-reactive protein. This article tries to summarize the most recent results in this area.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/fisiología , Proteína C-Reactiva/fisiología , Dislipidemias/terapia , Lípidos/fisiología , Enfermedades Vasculares/prevención & control , 1-Alquil-2-acetilglicerofosfocolina Esterasa/sangre , Adulto , Apolipoproteínas/sangre , Apolipoproteínas/fisiología , Biomarcadores/sangre , Dislipidemias/sangre , Dislipidemias/complicaciones , Humanos , Lípidos/sangre , Lipoproteínas LDL/sangre , Lipoproteínas LDL/fisiología , Tamaño de la Partícula , Enfermedades Vasculares/sangre , Enfermedades Vasculares/etiología
20.
J Virol ; 88(1): 592-603, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24173214

RESUMEN

Apolipoprotein L1 (APOL1) is a major component of the human innate immune response against African trypanosomes. Although the mechanism of the trypanolytic activity of circulating APOL1 has been recently clarified, the intracellular function(s) of APOL1 in human cells remains poorly defined. Like that of many genes linked to host immunity, APOL1 expression is induced by proinflammatory cytokines gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α). Additionally, IFN-γ-polarized macrophages that potently restrict HIV-1 replication express APOL1, which suggests that APOL1 may contribute to HIV-1 suppression. Here, we report that APOL1 inhibits HIV-1 replication by multiple mechanisms. We found that APOL1 protein targeted HIV-1 Gag for degradation by the endolysosomal pathway. Interestingly, we found that APOL1 stimulated both endocytosis and lysosomal biogenesis by promoting nuclear localization of transcription factor EB (TFEB) and expression of TFEB target genes. Moreover, we demonstrated that APOL1 depletes cellular viral accessory protein Vif, which counteracts the host restriction factor APOBEC3G, via a pathway involving degradation of Vif in lysosomes and by secretion of Vif in microvesicles. As a result of Vif depletion by APOL1, APOBEC3G was not degraded and reduced infectivity of progeny virions. In support of this model, we also showed that endogenous expression of APOL1 in differentiated U937 monocytic cells stimulated with IFN-γ resulted in a reduced production of virus particles. This finding supports the hypothesis that induction of APOL1 contributes to HIV-1 suppression in differentiated monocytes. Deciphering the precise mechanism of APOL1-mediated HIV-1 restriction may facilitate the design of unique therapeutics to target HIV-1 replication.


Asunto(s)
Apolipoproteínas/fisiología , Infecciones por VIH/inmunología , VIH-1/aislamiento & purificación , Inmunidad Innata , Lipoproteínas HDL/fisiología , Apolipoproteína L1 , Diferenciación Celular , Línea Celular , Endocitosis , Citometría de Flujo , Productos del Gen gag/inmunología , VIH-1/fisiología , Humanos , Interferones/inmunología , Macrófagos/inmunología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...