Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Environ Monit Assess ; 196(4): 369, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38489113

RESUMO

Protected areas are typically managed as a network of sites exposed to varying anthropogenic conditions. Managing these networks benefits from monitoring of conditions across sites to help prioritize coordinated efforts. Monitoring marine vessel activity and related underwater radiated noise impacts across a network of protected areas, like the U.S. National Marine Sanctuary system, helps managers ensure the quality of habitats used by a wide range of marine species. Here, we use underwater acoustic detections of vessels to quantify different characteristics of vessel noise at 25 locations within eight marine sanctuaries including the Hawaiian Archipelago and the U.S. east and west coasts. Vessel noise metrics, including temporal presence and sound levels, were paired with Automatic Identification System (AIS) vessel tracking data to derive a suite of robust vessel noise indicators for use across the network of marine protected areas. Network-wide comparisons revealed a spectrum of vessel noise conditions that closely matched AIS vessel traffic composition. Shifts in vessel noise were correlated with the decrease in vessel activity early in the COVID-19 pandemic, and vessel speed reduction management initiatives. Improving our understanding of vessel noise conditions in these protected areas can help direct opportunities for reducing vessel noise, such as establishing and maintaining noise-free periods, enhancing port efficiency, engaging with regional and international vessel quieting initiatives, and leveraging co-benefits of management actions for reducing ocean noise.


Assuntos
Pandemias , Navios , Humanos , Monitoramento Ambiental , Ruído , Acústica , Ecossistema
2.
J Acoust Soc Am ; 153(3): 1710, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-37002102

RESUMO

Marine soundscapes provide the opportunity to non-invasively learn about, monitor, and conserve ecosystems. Some fishes produce sound in chorus, often in association with mating, and there is much to learn about fish choruses and the species producing them. Manually analyzing years of acoustic data is increasingly unfeasible, and is especially challenging with fish chorus, as multiple fish choruses can co-occur in time and frequency and can overlap with vessel noise and other transient sounds. This study proposes an unsupervised automated method, called SoundScape Learning (SSL), to separate fish chorus from soundscape using an integrated technique that makes use of randomized robust principal component analysis (RRPCA), unsupervised clustering, and a neural network. SSL was applied to 14 recording locations off southern and central California and was able to detect a single fish chorus of interest in 5.3 yrs of acoustically diverse soundscapes. Through application of SSL, the chorus of interest was found to be nocturnal, increased in intensity at sunset and sunrise, and was seasonally present from late Spring to late Fall. Further application of SSL will improve understanding of fish behavior, essential habitat, species distribution, and potential human and climate change impacts, and thus allow for protection of vulnerable fish species.


Assuntos
Ecossistema , Som , Animais , Acústica , Peixes , Ruído
3.
Biochemistry (Mosc) ; 87(11): 1252-1259, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36509728

RESUMO

Adiponectin is an adipose tissue hormone, participating in energy metabolism and involved in atherogenesis. Previously, it was found that adiponectin increases expression of the APOA1 (apolipoprotein A-1) gene in hepatocytes, but the mechanisms of this effect remained unexplored. Our aim was to investigate the role of adiponectin receptors AdipoR1/R2, AMP-activated protein kinase (AMPK), nuclear peroxisome proliferator-activated receptor alpha (PPARα) and liver X receptors (LXRs) in mediating the action of adiponectin on hepatic APOA1 expression in human hepatoma HepG2 cells. The level of APOA1 expression was determined by RT-qPCR and ELISA. We showed that the siRNA-mediated knockdown of genes coding for AdipoR1, AdipoR2, AMPK, PPARα, and LXRα and ß prevented adiponectin-induced APOA1 expression in HepG2 cells and demonstrated that interaction of PPARα and LXRs with the APOA1 gene hepatic enhancer is important for the adiponectin-dependent APOA1 transcription. The results of this study point out to the involvement of both types of adiponectin receptors, AMPK, PPARα, and LXRs in the adiponectin-dependent upregulation of the APOA1 expression.


Assuntos
Adiponectina , PPAR alfa , Humanos , PPAR alfa/genética , PPAR alfa/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Receptores X do Fígado/genética , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Células Hep G2 , Apolipoproteína A-I/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Expressão Gênica
4.
European J Org Chem ; 2022(17): e202101278, 2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35910459

RESUMO

Teraryl-based α-helix mimetics have proven to be useful compounds for the inhibition of protein-protein interactions (PPI). We have developed a modular and flexible approach for the synthesis of teraryl-based α-helix mimetics using a benzene core unit featuring two leaving groups of differentiated reactivity in the Pd-catalyzed cross-coupling used for teraryl assembly. In previous publications we have introduced the methodology of 4-iodophenyltriflates decorated with the side chains of some of the proteinogenic amino acids. We herein report the core fragments corresponding to the previously missing amino acids Arg, Asn, Asp, Met, Trp and Tyr. Therefore, our set now encompasses all relevant amino acid analogues with the exception of His. In order to be compatible with the triflate moiety, some of the nucleophilic side chains had to be provided in a protected form to serve as stable building blocks. Additionally, cross-coupling procedures for the assembly of teraryls were investigated.

5.
Mol Cell Biochem ; 448(1-2): 211-223, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29442267

RESUMO

Apolipoprotein A-I (ApoA-I) is the main structural and functional protein component of high-density lipoprotein. ApoA-I has been shown to regulate lipid metabolism and inflammation in macrophages. Recently, we found the moderate expression of endogenous apoA-I in human monocytes and macrophages and showed that pro-inflammatory cytokine tumor necrosis factor α (TNFα) increases apoA-I mRNA and stimulates ApoA-I protein secretion by human monocytes and macrophages. Here, we present data about molecular mechanisms responsible for the TNFα-mediated activation of apoA-I gene in human monocytes and macrophages. This activation depends on JNK and MEK1/2 signaling pathways in human monocytes, whereas inhibition of NFκB, JNK, or p38 blocks an increase of apoA-I gene expression in the macrophages treated with TNFα. Nuclear receptor PPARα is a ligand-dependent regulator of apoA-I gene, whereas LXRs stimulate apoA-I mRNA transcription and ApoA-I protein synthesis and secretion by macrophages. Treatment of human macrophages with PPARα or LXR synthetic ligands as well as knock-down of LXRα, and LXRß by siRNAs interfered with the TNFα-mediated activation of apoA-I gene in human monocytes and macrophages. At the same time, TNFα differently regulated the levels of PPARα, LXRα, and LXRß binding to the apoA-I gene promoter in THP-1 cells. Obtained results suggest a novel tissue-specific mechanism of the TNFα-mediated regulation of apoA-I gene in monocytes and macrophages and show that endogenous ApoA-I might be positively regulated in macrophage during inflammation.


Assuntos
Apolipoproteína A-I/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Receptores X do Fígado/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , NF-kappa B/metabolismo , PPAR alfa/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Humanos , Macrófagos/citologia , Monócitos/citologia , Células THP-1
6.
J Cell Biochem ; 118(2): 382-396, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27404023

RESUMO

Apolipoprotein A-I (ApoA-I) is a key component of high density lipoproteins which possess anti-atherosclerotic and anti-inflammatory properties. Insulin is a crucial mediator of the glucose and lipid metabolism that has been implicated in atherosclerotic and inflammatory processes. Important mediators of insulin signaling such as Liver X Receptors (LXRs) and Forkhead Box A2 (FOXA2) are known to regulate apoA-I expression in liver. Forkhead Box O1 (FOXO1) is a well-known target of insulin signaling and a key mediator of oxidative stress response. Low doses of insulin were shown to activate apoA-I expression in human hepatoma HepG2 cells. However, the detailed mechanisms for these processes are still unknown. We studied the possible involvement of FOXO1, FOXA2, LXRα, and LXRß transcription factors in the insulin-mediated regulation of apoA-I expression. Treatment of HepG2 cells with high doses of insulin (48 h, 100 nM) suppresses apoA-I gene expression. siRNAs against FOXO1, FOXA2, LXRß, or LXRα abrogated this effect. FOXO1 forms a complex with LXRß and insulin treatment impairs FOXO1/LXRß complex binding to hepatic enhancer and triggers its nuclear export. Insulin as well as LXR ligand TO901317 enhance the interaction between FOXA2, LXRα, and hepatic enhancer. These data suggest that high doses of insulin downregulate apoA-I gene expression in HepG2 cells through redistribution of FOXO1/LXRß complex, FOXA2, and LXRα on hepatic enhancer of apoA-I gene. J. Cell. Biochem. 118: 382-396, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Apolipoproteína A-I/biossíntese , Carcinoma Hepatocelular/metabolismo , Regulação para Baixo/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Insulina/farmacologia , Neoplasias Hepáticas/metabolismo , Receptores X do Fígado/metabolismo , Proteínas de Neoplasias/metabolismo , Carcinoma Hepatocelular/patologia , Células Hep G2 , Humanos , Hidrocarbonetos Fluorados/farmacologia , Neoplasias Hepáticas/patologia , Sulfonamidas/farmacologia
7.
J Cell Biochem ; 117(9): 2010-22, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26813964

RESUMO

Apolipoprotein A-I (ApoA-I) is the main anti-atherogenic component of human high-density lipoproteins (HDL). ApoA-I gene expression is regulated by several nuclear receptors, which are the sensors for metabolic changes during development of cardiovascular diseases. Activation of nuclear receptor PPARγ has been shown to impact lipid metabolism as well as inflammation. Here, we have shown that synthetic PPARγ agonist GW1929 decreases both ApoA-I mRNA and protein levels in HepG2 cells and the effect of GW1929 on apoA-I gene transcription depends on PPARγ. PPARγ binds to the sites A and C within the hepatic enhancer of apoA-I gene and the negative regulation of apoA-I gene transcription by PPARγ appears to be realized via the site C (-134 to -119). Ligand activation of PPARγ leads to an increase of LXRß and a decrease of PPARα binding to the apoA-I gene hepatic enhancer in HepG2 cells. GW1929 abolishes the TNFα-mediated decrease of ApoA-I mRNA expression in both HepG2 and Caco-2 cells but does not block TNFα-mediated inhibition of ApoA-I protein secretion by HepG2 cells. These data demonstrate that complex of PPARγ with GW1929 is a negative regulator involved in the control of ApoA-I expression and secretion in human hepatocyte- and enterocyte-like cells. J. Cell. Biochem. 117: 2010-2022, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Apolipoproteína A-I/biossíntese , Regulação para Baixo , Elementos Facilitadores Genéticos , Enterócitos/metabolismo , Hepatócitos/metabolismo , PPAR gama/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Apolipoproteína A-I/genética , Benzofenonas/farmacologia , Células CACO-2 , Células Hep G2 , Humanos , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/genética , Fator de Necrose Tumoral alfa/genética , Tirosina/análogos & derivados , Tirosina/farmacologia
8.
J Biol Chem ; 288(15): 10692-702, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23449984

RESUMO

Nuclear receptors modulate macrophage effector functions, which are imperative for clearance or survival of mycobacterial infection. The adopted orphan nuclear receptor Rev-erbα is a constitutive transcriptional repressor as it lacks AF2 domain and was earlier shown to be present in macrophages. In the present study, we highlight the differences in the relative subcellular localization of Rev-erbα in monocytes and macrophages. The nuclear localization of Rev-erbα in macrophages is subsequent to monocyte differentiation. Expression analysis of Rev-erbα elucidated it to be considerably more expressed in M1 phenotype in comparison with M2. Rev-erbα overexpression augments antimycobacterial properties of macrophage by keeping IL10 in a basal repressed state. Further, promoter analysis revealed that IL10 promoter harbors a Rev-erbα binding site exclusive to humans and higher order primates and not mouse, demonstrating a species barrier in its functionality. This direct gene repression is mediated by recruitment of co-repressors NCoR and HDAC3. In addition, our data elucidate that its overexpression reduced the survival of intracellular pathogen Mycobacterium tuberculosis by enhancing phagosome lysosome maturation, an event resulting from IL10 repression. Thus, these findings suggest that Rev-erbα bestows protection against mycobacterial infection by direct gene repression of IL10 and thus provide a novel target in modulating macrophage microbicidal properties.


Assuntos
Regulação da Expressão Gênica , Interleucina-10/biossíntese , Macrófagos/metabolismo , Mycobacterium tuberculosis/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Elementos de Resposta , Animais , Linhagem Celular , Feminino , Humanos , Interleucina-10/genética , Interleucina-10/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Masculino , Camundongos , Mycobacterium tuberculosis/imunologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/imunologia , Especificidade da Espécie , Tuberculose/genética , Tuberculose/imunologia , Tuberculose/metabolismo , Tuberculose/patologia
9.
J Biol Chem ; 288(3): 1726-38, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23168409

RESUMO

Complement C3 is a pivotal component of three cascades of complement activation. The liver is the main source of C3 in circulation and expression and secretion of C3 by hepatocytes is increased during acute inflammation. However, the mechanism of the regulation of the C3 gene in hepatocytes is not well elucidated. We showed that the C3 gene is the direct target for peroxisome proliferator-activated receptor α (PPARα) in human hepatoma HepG2 cells and mouse liver. Using PPARα siRNA and synthetic PPARα agonist WY-14643 and antagonist MK886 we showed that activation of PPARα results in up-regulation of C3 gene expression and protein secretion by HepG2 cells. The PPAR response element (PPRE), which is able to bind PPARα in vitro and in vivo, was found in the human C3 promoter. PPRE is conserved between human and mouse, and WY-14643 stimulates mouse C3 expression in the liver. TNFα increases C3 gene via NF-κB and, to a lesser extent, MEK1/2 signaling pathways, whereas TNFα-mediated stimulation of C3 protein secretion depends on activation of MEK1/2, p38, and JNK in HepG2 cells. Activation of PPARα abolishes TNFα-mediated up-regulation of C3 gene expression and protein secretion due to interference with NF-κB via PPRE-dependent mechanism in HepG2 cells. TNFα decreases PPARα protein content via NF-κB and MEK1/2 signaling pathways and inhibits PPARα binding with the human C3 promoter in HepG2 cells. These results suggest novel mechanism controlling C3 expression in hepatocytes during acute phase inflammation and demonstrate a crosstalk between PPARα and TNFα in the regulation of complement system.


Assuntos
Complemento C3/genética , Hepatócitos/metabolismo , Inflamação/genética , PPAR alfa/genética , Fator de Necrose Tumoral alfa/farmacologia , Animais , Complemento C3/agonistas , Complemento C3/antagonistas & inibidores , Complemento C3/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/imunologia , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Lipopolissacarídeos , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , PPAR alfa/imunologia , Proliferadores de Peroxissomos/farmacologia , Ligação Proteica , Pirimidinas/farmacologia , Elementos de Resposta , Transdução de Sinais , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Transplant Cell Ther ; 30(2): 191-202, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37967650

RESUMO

Chimeric antigen receptor T cell (CAR-T) therapy has shown rapid, frequent, and deep responses in patients with relapsed/refractory multiple myeloma (RRMM). However, relapse frequently occurs following CAR-T therapy, and the cause of this resistance is not well defined. Among the potential mechanisms of resistance, T cell intrinsic factors may be an important source of failure. Here we used spectral flow cytometry to identify the changes in T cell phenotypes in bone marrow aspirates at different stages of multiple myeloma progression, including cases that relapsed after anti-BCMA CAR-T therapy. We identified completely different T cell phenotypes in RRMM and post CAR-T relapse cases compared to healthy donors and earlier stages of multiple myeloma, novel double-negative CD3+ T cells in RRMM and CAR-T relapsed cases, and differences in CD8 T cell phenotype at the baseline between peripheral blood and bone marrow from healthy donors. We found that the majority of T cells in RRMM patients and significant T cell subsets in post-CAR-T relapsed patients expressed multiple coinhibitory markers, including PD1, TIGIT, 2B4, and KLRG1.


Assuntos
Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Humanos , Mieloma Múltiplo/terapia , Antígeno de Maturação de Linfócitos B/genética , Recidiva Local de Neoplasia , Recidiva , Terapia Baseada em Transplante de Células e Tecidos , Receptores Imunológicos , Lectinas Tipo C
11.
J Soc Cardiovasc Angiogr Interv ; 3(4): 101350, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-39130181

RESUMO

Background: Pulmonary artery wedge pressure (PAWP) can underestimate directly measured pulmonary vein pressure (PVP) as demonstrated in animal studies and human case reports. This concept has not been validated in a larger cohort of pediatric patients with pulmonary vein stenosis (PVS). Methods: Pediatric patients who underwent cardiac catheterization for PVS at a single center from January 1, 2018, to March 31, 2023, were retrospectively reviewed. Mismatch between the PAWP and directly measured PVP or LA pressure was defined as >3 mm Hg. Preintervention angiography was reviewed and percent stenosis calculated. Results: Twenty-six patients met inclusion criteria; 51 lower pulmonary veins (34 left, 17 right) from 42 catheterizations were evaluated. Significant PVS (≥30% stenosis) was seen in 38/51 (75%) veins, and 9/51 (18%) veins had no angiographic narrowing (0% stenosis). PAWP-PVP mismatch occurred in 37/51 (73%) veins with a median difference of 8 mm Hg (IQR, 6-12). Of these, PAWP was equal to LA pressure in 26 instances, all of which had significant PVS (median % stenosis 54 [IQR, 45-60]). Six of the cases with PAWP-PVP mismatch, PVS (range, 41%-70% stenosis), and no PAWP-LA mismatch reported both a proximal and distal segmental PAWP. In all 6 instances, there was no distal PAWP-PVP mismatch (median difference 1 mm Hg [range, 0-3]). Conclusions: In this single-center study of pediatric patients with PVS, PAWP significantly underestimated directly measured PVP in lower pulmonary veins. Balloon wedge or end hole catheter position in a distal lobar segment may more accurately estimate the PVP.

12.
J Biol Chem ; 287(8): 5954-68, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22194611

RESUMO

Complement C3 is a pivotal component of three cascades of complement activation. C3 is expressed in human atherosclerotic lesions and is involved in atherogenesis. However, the mechanism of C3 accumulation in atherosclerotic lesions is not well elucidated. We show that acetylated low density lipoprotein and oxidized low density lipoprotein (oxLDL) increase C3 gene expression and protein secretion by human macrophages. Modified LDL (mLDL)-mediated activation of C3 expression mainly depends on liver X receptor (LXR) and partly on Toll-like receptor 4 (TLR4), whereas C3 secretion is increased due to TLR4 activation by mLDL. LXR agonist TO901317 stimulates C3 gene expression in human monocyte-macrophage cells but not in human hepatoma (HepG2) cells. We find LXR-responsive element inside of the promoter region of the human C3 gene, which binds to LXRß in macrophages but not in HepG2 cells. We show that C3 expression and secretion is decreased in IL-4-treated (M2) and increased in IFNγ/LPS-stimulated (M1) human macrophages as compared with resting macrophages. LXR agonist TO901317 potentiates LPS-induced C3 gene expression and protein secretion in macrophages, whereas oxLDL differently modulates LPS-mediated regulation of C3 in M1 or M2 macrophages. Treatment of human macrophages with anaphylatoxin C3a results in stimulation of C3 transcription and secretion as well as increased oxLDL accumulation and augmented oxLDL-mediated up-regulation of the C3 gene. These data provide a novel mechanism of C3 gene regulation in macrophages and suggest new aspects of cross-talk between mLDL, C3, C3a, and TLR4 during development of atherosclerotic lesions.


Assuntos
Complemento C3/genética , Complemento C3/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Macrófagos/efeitos dos fármacos , Receptores Nucleares Órfãos/metabolismo , Receptor 4 Toll-Like/metabolismo , Sequência de Bases , Transporte Biológico , Complemento C3/biossíntese , Células Hep G2 , Humanos , Lipoproteínas LDL/metabolismo , Receptores X do Fígado , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/metabolismo , NF-kappa B/metabolismo , Receptores Nucleares Órfãos/agonistas , Regiões Promotoras Genéticas/genética , Transcrição Gênica/efeitos dos fármacos
13.
Synthesis (Stuttg) ; 55(15): 2319-2324, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37691879

RESUMO

A new method to prepare 1,4-oxazinone intermediates was developed based on aza-conjugate addition of ß-amino alcohols to electron-deficient alkyne precursors. A tandem intramolecular cycloaddition/cycloreversion reaction sequence was evaluated, leading to the synthesis of the guaipyridine alkaloid natural products rupestine M and L. Starting from (-)-citronellal and thus a known configuration of the C5 stereocenter, a revised absolute configuration of natural rupestine L is suggested based on optical rotation.

14.
Cell Biochem Biophys ; 81(1): 151-160, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36251137

RESUMO

ATP-binding cassette transporter A-I (ABCA1) is an ubiquitously expressed protein whose main function is the transmembrane transport of cholesterol and phospholipids. Synthesis of ABCA1 protein in liver is necessary for high-density lipoprotein (HDL) formation in mammals. Thus, the mechanism of ABCA1 gene expression regulation in hepatocytes are of critical importance. Recently, we have found the insulin-dependent downregulation of other key player in the HDL formation-apolipoprotein A-I gene (J. Cell. Biochem., 2017, 118:382-396). Nothing is known about the role of insulin in the regulation of ABCA1 gene. Here we show for the first time that insulin decreases the mRNA and protein levels of ABCA1 in human hepatoma cell line HepG2. PI3K, p38, MEK1/2, JNK and mTORC1 signaling pathways are involved in the insulin-mediated downregulation of human ABCA1 gene. Transcription factors LXRα, LXRß, FOXO1 and NF-κB are important contributors to this process, while FOXA2 does not regulate ABCA1 gene expression. Insulin causes the decrease in FOXO1, LXRα and LXRß binding to ABCA1 promoter, which is likely the cause of the decrease in the gene expression. Interestingly, the murine ABCA1 gene seems to be not regulated by insulin in hepatocytes (in vitro and in vivo). We suggest that the reason for this discrepancy is the difference in the 5'-regulatory regions of human and murine ABCA1 genes.


Assuntos
Transportador 1 de Cassete de Ligação de ATP , Insulina , Receptores X do Fígado , Receptores Nucleares Órfãos , Animais , Humanos , Camundongos , Transportador 1 de Cassete de Ligação de ATP/genética , Carcinoma Hepatocelular , Linhagem Celular , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Insulina/metabolismo , Neoplasias Hepáticas , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Mamíferos/metabolismo , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo
15.
Liver Transpl ; 17(4): 418-27, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21445925

RESUMO

The transplantation of primary hepatocytes has been shown to augment the function of damaged livers and to bridge patients to liver transplantation. However, primary hepatocytes often have low levels of engraftment and survive for only a short time after transplantation. To explore the potential benefits of using decellularized liver matrix (DLM) as a carrier for hepatocyte transplantation, DLM from whole mouse livers was generated. Human fetal hepatocytes immortalized by telomerase reconstitution (FH-hTERTs) or primary human hepatocytes were infused into the DLM, which was then implanted into the omenta of immunodeficient nonobese diabetic/severe combined immunodeficient/interleukin-2 receptor γ-deficient mice or nonobese diabetic/severe combined immunodeficient/mucopolysaccharidosis type VII mice. The removal of endogenous cellular components and the preservation of the extracellular matrix proteins and vasculature were demonstrated in the resulting DLM. Bioluminescent imaging revealed that FH-hTERTs transduced with a lentiviral vector expressing firefly luciferase survived in the DLM for 8 weeks after peritoneal implantation, whereas the luciferase signal from FH-hTERTs rapidly declined in control mice 3 to 4 weeks after transplantation via splenic injection or omental implantation after Matrigel encapsulation. Furthermore, primary human hepatocytes that were reconstituted in the DLM not only survived 6 weeks after transplantation but also maintained their function, as demonstrated by messenger RNA levels of albumin and cytochrome P450 (CYP) subtypes (CYP3A4, CYP2C9, and CYP1A1) similar to the levels in freshly isolated human primary hepatocytes (hPHs). In contrast, when hPHs were transplanted into mice via splenic injection, they failed to express CYP3A4, although they expressed albumin. In conclusion, DLM provides an excellent environment for long-term survival and maintenance of the hepatocyte phenotype after transplantation.


Assuntos
Transplante de Tecido Fetal/métodos , Hepatócitos/transplante , Fígado/citologia , Animais , Sobrevivência Celular , Células Cultivadas , Hepatócitos/fisiologia , Humanos , Camundongos
16.
Expert Opin Ther Targets ; 25(3): 167-174, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33703985

RESUMO

INTRODUCTION: KRAS mutations drive tumorigenesis by altering cell signaling and the tumor immune microenvironment. Recent studies have shown promise for KRAS-G12C covalent inhibitors, which are advancing rapidly through clinical trials. The sequencing and combination of these agents with other therapies including immune checkpoint blockade (ICB) will benefit from strategies that also address the immune microenvironment to improve durability of response. AREAS COVERED: This paper reviews KRAS signaling and discusses downstream effects on cytokine production and the tumor immune microenvironment. RAS targeted therapy is introduced and perspectives on therapeutic targeting of KRAS-G12C and its immunosuppressive tumor microenvironment are offered. EXPERT OPINION: The availability of KRAS-G12C covalent inhibitors raises hopes for targeting this pervasive oncogene and designing better therapeutic combinations to promote anti-tumor immunity. A comprehensive mechanistic understanding of KRAS immunosuppression is required in order to prioritize agents for clinical trials.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Animais , Antineoplásicos/administração & dosagem , Desenho de Fármacos , Humanos , Inibidores de Checkpoint Imunológico/administração & dosagem , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade Inata , Terapia de Alvo Molecular , Mutação , Neoplasias/genética , Neoplasias/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Microambiente Tumoral/imunologia
17.
Biochem Biophys Res Commun ; 402(3): 477-82, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-20951680

RESUMO

Synthesis of ABCA1 protein in liver is necessary for high-density lipoproteins (HDL) formation in mammals. Nuclear receptor PPARγ is known as activator of ABCA1 expression, but details of PPARγ-mediated regulation of ABCA1 at both transcriptional and post-transcriptional levels in hepatocytes have not still been well elucidated. In this study we have shown, that PPARγ activates ABCA1 gene transcription in human hepatoma cells HepG2 through increasing of LXRß binding with promoter region of ABCA1 gene. Treatment of HepG2 cells with PPARγ agonist GW1929 leads to dissociation of LXRß from ABCA1/LXRß complex and to nuclear translocation of this nuclear receptor resulting in reduction of ABCA1 protein level 24h after treatment. Inhibition of protein kinases MEK1/2 abolishes PPARγ-mediated dissociation of LXRß from ABCA1/LXRß complex, but does not block PPARγ-dependent down-regulation of ABCA1 protein in HepG2 cells. These data suggest that PPARγ may be important for regulation of the level of hepatic ABCA1 protein and indicate the new interplays between PPARγ, LXRß and MEK1/2 in regulation of ABCA1 mRNA and protein expression.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Fígado/metabolismo , PPAR gama/metabolismo , Ativação Transcricional , Transportador 1 de Cassete de Ligação de ATP , Benzofenonas/farmacologia , Núcleo Celular/metabolismo , Regulação para Baixo , Células Hep G2 , Humanos , Receptores X do Fígado , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/metabolismo , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , PPAR gama/agonistas , Regiões Promotoras Genéticas , Biossíntese de Proteínas , Transcrição Gênica , Tirosina/análogos & derivados , Tirosina/farmacologia
18.
Biochem Biophys Res Commun ; 398(2): 224-30, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20599735

RESUMO

Human apolipoprotein A-I (ApoA-I) is a major structural and functional protein component of high-density lipoproteins. The expression of the apolipoprotein A-I gene (apoA-I) in hepatocytes is repressed by pro-inflammatory cytokines such as IL-1beta and TNFalpha. Recently, two novel additional (alternative) promoters for human apoA-I gene have been identified. Nothing is known about the role of alternative promoters in TNFalpha-mediated downregulation of apoA-I gene. In this article we report for the first time about the different effects of TNFalpha on two alternative promoters of human apoA-I gene. Stimulation of HepG2 cells by TNFalpha leads to activation of the distal alternative apoA-I promoter and downregulation of the proximal alternative and the canonical apoA-I promoters. This effect is mediated by weakening of the promoter competition within human apoA-I 5'-regulatory region (apoA-I promoter switching) in the cells treated by TNFalpha. The MEK1/2-ERK1/2 cascade and nuclear receptors PPARalpha and LXRs are important for TNFalpha-mediated apoA-I promoter switching.


Assuntos
Apolipoproteína A-I/genética , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular Tumoral , Humanos , Receptores X do Fígado , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Receptores Nucleares Órfãos/metabolismo , PPAR alfa/metabolismo , Regiões Promotoras Genéticas
19.
Biochemistry ; 48(50): 11950-60, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-19883121

RESUMO

The expression of the apolipoprotein A-I gene (apoA-I) in hepatocytes is repressed by pro-inflammatory cytokines such as IL-1beta and TNFalpha. In this work, we have demonstrated that treatment of HepG2 human hepatoma cells with chemical inhibitors for JNK, p38 protein kinases, and NFkappaB transcription factor abolishes the TNFalpha-mediated inhibition of human apoA-I gene expression in HepG2 cells. In addition, we have shown that TNFalpha decreases also the rate of secretion of apoA-I protein by HepG2 cells, and this effect depends on JNK and p38, but not on NFkappaB and MEK1/2 signaling pathways. The inhibitory effect of TNFalpha has been found to be mediated by the hepatic enhancer of the apoA-I gene. The decrease in the level of human apoA-I gene expression under the impact of TNFalpha appears to be partly mediated by the inhibition of HNF4alpha and PPARalpha gene expression. Treatment of HepG2 cells with PPARalpha antagonist (MK886) or LXR agonist (TO901317) abolishes the TNFalpha-mediated decrease in the level of apoA-I gene expression. PPARalpha agonist (WY-14643) abolishes the negative effect of TNFalpha on apoA-I gene expression in the case of simultaneous inhibition of MEK1/2, although neither inhibition of MEK1/2 nor addition of WY-14643 leads to the blocking of the TNFalpha-mediated decrease in the level of apoA-I gene expression individually. The ligand-dependent regulation of apoA-I gene expression by PPARalpha appears to be affected by the TNFalpha-mediated activation of MEK1/2 kinases, probably through PPARalpha phosphorylation. Treatment of HepG2 cells with PPARalpha and LXR synthetic agonists also blocks the inhibition of apoA-I protein secretion in HepG2 cells under the impact of TNFalpha. A chromatin immunoprecipitation assay demonstrates that TNFalpha leads to a 2-fold decrease in the level of PPARalpha binding with the apoA-I gene hepatic enhancer. At the same time, the level of LXRbeta binding with the apoA-I gene hepatic enhancer is increased 3-fold under the impact of TNFalpha. These results suggest that nuclear receptors HNF4alpha, PPARalpha, and LXRs are involved in the TNFalpha-mediated downregulation of human apoA-I gene expression and apoA-I protein secretion in HepG2 cells.


Assuntos
Apolipoproteína A-I/antagonistas & inibidores , Apolipoproteína A-I/genética , Regulação para Baixo/genética , Fator 4 Nuclear de Hepatócito/fisiologia , Receptores Nucleares Órfãos/fisiologia , PPAR alfa/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Apolipoproteína A-I/biossíntese , Linhagem Celular Tumoral , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Ligantes , Receptores X do Fígado , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/fisiologia , NF-kappa B/fisiologia , Nafenopina/metabolismo , Nafenopina/farmacologia , Pirimidinas/metabolismo , Pirimidinas/farmacologia
20.
Rev Stomatol Chir Maxillofac ; 110(5): 251-5, 2009 Nov.
Artigo em Francês | MEDLINE | ID: mdl-19427008

RESUMO

INTRODUCTION: Implant supported dental prostheses are the most up-to-date solution for edentulous patients. This technique requires and adequate bone quantity and quality. Bone distraction may allow compensating for some bone deficit, especially mandibulary. Few studies have been dedicated to how patients adjusted to this therapy (Int J oral Maxillofac Surg 34 [2005] 238-42, Int J oral Maxillofac Surg 36 [2007] 896-9, Med Oral Pathol Oral Cir Bucal 12 [2007] E225-8). We evaluated the psychological impact of alveolar mandibular distraction. PATIENTS AND METHODS: Between 1999 and 2006, 31 patients aged 27 to 70 years underwent vertical alveolar mandibular distraction. Seventeen patients (54.8%) presented with complications. A questionnaire assessed the psychological impact by using notions used in healthcare psychology: perceived stress, perceived control, and social support. RESULTS: Twenty-three answers (74.2% of operated cases) were studied. In 87% of the cases, patients adjusted well the distraction procedure. Eighty-one percent felt no stress. Fifty-seven percent reported light to moderate pain, and 43% found the treatment painful. Confrontation to adverse events was mentioned only in 13% of the cases. In 17% of the cases, there was a slight alteration of sleep. Fifty-seven percent of the patients managed to forget the presence of the distractor. The most difficult stages were insertion of the distractor (48%) and the activation phase (17%). Seventy-one percent of the patients did not find the protocol restraining. The treatment length was not a problem for 65%. Two patients (9%) found it too long. Ninety-one percent of the patients activated the device on their own, for two (9% of the cases) the surgeon activated the device. Ninety-seven percent of the patients found supervision satisfactory. Medical information helped to adjust well to the procedure in 96% of the cases. Forty-three percent of the patients (10 cases) required specific help during the treatment: family support, attending physician, or surgical team. Among these, 70% considered the medical team's contribution as the most beneficial compared to family support, and 30% reported they were the same. For 96% of the patients, the medical team was the major actor of therapeutic success. Twenty-two percent of the patients considered they were partly responsible for treatment results (perceived internal control). Fifty-two percent of the cases reported they would be ready to undergo another distraction protocol if necessary. DISCUSSION: Alveolar mandibular distraction has only a weak psychological impact. It improves the patient's quality of life. It can be suggested that hope for improvement helped patients to better stand treatment. The information received contributed to better adjust to events. This unpredictable situation does not allow the patient to prepare himself and generates stress. Information in necessary before operating, to let the patient assess his own resources and prepare his own psychological adaptation mechanism. It is through this means that we can speak about true "informed consent or refusal".


Assuntos
Mandíbula/cirurgia , Procedimentos Cirúrgicos Ortognáticos/psicologia , Osteogênese por Distração/psicologia , Adaptação Psicológica , Adulto , Idoso , Humanos , Arcada Edêntula/cirurgia , Pessoa de Meia-Idade , Procedimentos Cirúrgicos Pré-Protéticos Bucais/psicologia , Satisfação do Paciente/estatística & dados numéricos , Qualidade de Vida , Estresse Psicológico , Inquéritos e Questionários
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA