Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Plant Physiol ; 195(2): 1461-1474, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38431527

RESUMEN

Black goji berry (Lycium ruthenicum Murray) contains a rich source of health-promoting anthocyanins which are used in herbal medicine and nutraceutical foods in China. A natural variant producing white berries allowed us to identify two key genes involved in the regulation of anthocyanin biosynthesis in goji berries: one encoding a MYB transcription factor (LrAN2-like) and one encoding a basic helix-loop-helix (bHLH) transcription factor (LrAN1b). We previously found that LrAN1b expression was lost in the white berry variant, but the molecular basis for this phenotype was unknown. Here, we identified the molecular mechanism for loss of anthocyanins in white goji berries. In white goji, the LrAN1b promoter region has a 229 bp deletion that removes three MYB-binding elements and one bHLH-binding element, which are key to its expression. Complementation of the white goji berry LrAN1b allele with the LrAN1b promoter restored pigmentation. Virus-induced gene silencing of LrAN1b in black goji berry reduced fruit anthocyanin biosynthesis. Molecular analyses showed that LrAN2-like and another bHLH transcription factor LrJAF13 can activate LrAN1b by binding directly to the MYB-recognizing element and bHLH-recognizing element of its promoter-deletion region. LrAN1b expression is enhanced by the interaction of LrAN2-like with LrJAF13 and the WD40 protein LrAN11. LrAN2-like and LrAN11 interact with either LrJAF13 or LrAN1b to form two MYB-bHLH-WD40 complexes, which hierarchically regulate anthocyanin biosynthesis in black goji berry. This study on a natural variant builds a comprehensive anthocyanin regulatory network that may be manipulated to tailor goji berry traits.


Asunto(s)
Antocianinas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Frutas , Regulación de la Expresión Génica de las Plantas , Lycium , Proteínas de Plantas , Regiones Promotoras Genéticas , Antocianinas/biosíntesis , Antocianinas/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Frutas/genética , Frutas/metabolismo , Lycium/genética , Lycium/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Eliminación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
BMC Plant Biol ; 24(1): 441, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38778301

RESUMEN

BACKGROUND: Goji (Lycium barbarum L.) is a perennial deciduous shrub widely distributed in arid and semiarid regions of Northwest China. It is highly valued for its medicinal and functional properties. Most goji varieties are naturally self-incompatible, posing challenges in breeding and cultivation. Self-incompatibility is a complex genetic trait, with ongoing debates regarding the number of self-incompatible loci. To date, no genetic mappings has been conducted for S loci or other loci related to self-incompatibility in goji. RESULTS: We used genome resequencing to create a high-resolution map for detecting de novo single-nucleotide polymorphisms (SNP) in goji. We focused on 229 F1 individuals from self-compatible '13-19' and self-incompatible 'new 9' varieties. Subsequently, we conducted a quantitative trait locus (QTL) analysis on traits associated with self-compatibility in goji berries. The genetic map consisted of 249,327 SNPs distributed across 12 linkage groups (LGs), spanning a total distance of 1243.74 cM, with an average interval of 0.002 cM. Phenotypic data related to self-incompatibility, such as average fruit weight, fruit rate, compatibility index, and comparable compatibility index after self-pollination and geitonogamy, were collected for the years 2021-2022, as well as for an extra year representing the mean data from 2021 to 2022 (2021/22). A total of 43 significant QTL, corresponding to multiple traits were identified, accounting for more than 11% of the observed phenotypic variation. Notably, a specific QTL on chromosome 2 consistently appeared across different years, irrespective of the relationship between self-pollination and geitonogamy. Within the localization interval, 1180 genes were annotated, including Lba02g01102 (annotated as an S-RNase gene), which showed pistil-specific expression. Cloning of S-RNase genes revealed that the parents had two different S-RNase alleles, namely S1S11 and S2S8. S-genotype identification of the F1 population indicated segregation of the four S-alleles from the parents in the offspring, with the type of S-RNase gene significantly associated with self-compatibility. CONCLUSIONS: In summary, our study provides valuable insights into the genetic mechanism underlying self-compatibility in goji berries. This highlights the importance of further positional cloning investigations and emphasizes the importance of integration of marker-assisted selection in goji breeding programs.


Asunto(s)
Mapeo Cromosómico , Frutas , Lycium , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Lycium/genética , Lycium/fisiología , Frutas/genética , Frutas/fisiología , Autoincompatibilidad en las Plantas con Flores/genética , Fenotipo , China
3.
Am J Physiol Gastrointest Liver Physiol ; 324(1): G38-G50, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36283963

RESUMEN

Pregnancy induces reprogramming of maternal physiology to support fetal development and growth. Maternal hepatocytes undergo hypertrophy and hyperplasia to drive maternal liver growth and alter their gene expression profiles simultaneously. This study aimed to further understand maternal hepatocyte adaptation to pregnancy. Timed pregnancies were generated in mice. In a nonpregnant state, most hepatocytes expressed Cd133, α-fetal protein (Afp) and epithelial cell adhesion molecule (Epcam) mRNAs, whereas overall, at the protein level, they exhibited a CD133-/AFP- phenotype; however, pericentral hepatocytes were EpCAM+. As pregnancy advanced, although most maternal hepatocytes retained Cd133, Afp, and Epcam mRNA expression, they generally displayed a phenotype of CD133+/AFP+, and EpCAM protein expression was switched from pericentral to periportal maternal hepatocytes. In addition, we found that the Hippo/yes-associated protein (YAP) pathway does not respond to pregnancy. Yap1 gene deletion specifically in maternal hepatocytes did not affect maternal liver growth or metabolic zonation. However, the absence of Yap1 gene eliminated CD133 protein expression without interfering with Cd133 transcript expression in maternal livers. We demonstrated that maternal hepatocytes acquire heterogeneous and dynamic developmental phenotypes, resembling fetal hepatocytes, partially via YAP1 through a posttranscriptional mechanism. Moreover, maternal liver is a new source of AFP. In addition, maternal liver grows and maintains its metabolic zonation independent of the Hippo/YAP1 pathway. Our findings revealed a novel and gestation-dependent phenotypic plasticity in adult hepatocytes.NEW & NOTEWORTHY We found that maternal hepatocytes exhibit developmental phenotypes in a temporal and spatial manner, similarly to fetal hepatocytes. They acquire this new property partially via yes-associated protein 1.


Asunto(s)
Proteínas Señalizadoras YAP , alfa-Fetoproteínas , Embarazo , Femenino , Ratones , Animales , Molécula de Adhesión Celular Epitelial/genética , alfa-Fetoproteínas/genética , Hepatocitos/metabolismo , Hígado/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción/metabolismo , Fenotipo
4.
Am J Physiol Gastrointest Liver Physiol ; 321(4): G389-G399, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34431407

RESUMEN

After partial hepatectomy (PH), the majority of remnant hepatocytes synchronously enter and rhythmically progress through the cell cycle for three major rounds to regain lost liver mass. Whether and how the circadian clock core component Bmal1 modulates this process remains elusive. We performed PH on Bmal1+/+ and hepatocyte-specific Bmal1 knockout (Bmal1hep-/-) mice and compared the initiation and progression of the hepatocyte cell cycle. After PH, Bmal1+/+ hepatocytes exhibited three major waves of nuclear DNA synthesis. In contrast, in Bmal1hep-/- hepatocytes, the first wave of nuclear DNA synthesis was delayed by 12 h, and the third such wave was lost. Following PH, Bmal1+/+ hepatocytes underwent three major waves of mitosis, whereas Bmal1hep-/- hepatocytes fully abolished mitotic oscillation. These Bmal1-dependent disruptions in the rhythmicity of hepatocyte cell cycle after PH were accompanied by suppressed expression peaks of a group of cell cycle components and regulators and dysregulated activation patterns of mitogenic signaling molecules c-Met and epidermal growth factor receptor. Moreover, Bmal1+/+ hepatocytes rhythmically accumulated fat as they expanded following PH, whereas this phenomenon was largely inhibited in Bmal1hep-/- hepatocytes. In addition, during late stages of liver regrowth, Bmal1 absence in hepatocytes caused the activation of redox sensor Nrf2, suggesting an oxidative stress state in regenerated liver tissue. Collectively, we demonstrated that during liver regeneration, Bmal1 partially modulates the oscillation of S-phase progression, fully controls the rhythmicity of M-phase advancement, and largely governs fluctuations in fat metabolism in replicating hepatocytes, as well as eventually determines the redox state of regenerated livers.NEW & NOTEWORTHY We demonstrated that Bmal1 centrally controls the synchronicity and rhythmicity of the cell cycle and lipid accumulation in replicating hepatocytes during liver regeneration. Bmal1 plays these roles, at least in part, by ensuring formation of the expression peaks of cell cycle components and regulators, as well as the timing and levels of activation of mitogenic signaling molecules.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Ciclo Celular , Proliferación Celular , Ritmo Circadiano , Hepatocitos/metabolismo , Regeneración Hepática , Factores de Transcripción ARNTL/genética , Animales , Receptores ErbB/metabolismo , Hepatocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Mitosis , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal
5.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G772-G780, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32003603

RESUMEN

Liver resection induces robust liver regrowth or regeneration to compensate for the lost tissue mass. In a clinical setting, pregnant women may need liver resection without terminating pregnancy in some cases. However, how pregnancy affects maternal liver regeneration remains elusive. We performed 70% partial hepatectomy (PH) in nonpregnant mice and gestation day 14 mice, and histologically and molecularly compared their liver regrowth during the next 4 days. We found that compared with the nonpregnant state, pregnancy altered the molecular programs driving hepatocyte replication, indicated by enhanced activities of epidermal growth factor receptor and STAT5A, reduced activities of cMet and p70S6K, decreased production of IL-6, TNFα, and hepatocyte growth factor, suppressed cyclin D1 expression, increased cyclin A1 expression, and early activated cyclin A2 expression. As a result, pregnancy allowed the remnant hepatocytes to enter the cell cycle at least 12 h earlier, increased hepatic fat accumulation, and enhanced hepatocyte mitosis. Consequently, pregnancy ameliorated maternal liver regeneration following PH. In addition, a report showed that maternal liver regrowth after PH is driven mainly by hepatocyte hypertrophy rather than hyperplasia during the second half of gestation in young adult mice. In contrast, we demonstrate that maternal liver relies mainly on hepatocyte hyperplasia instead of hypertrophy to restore the lost mass after PH. Overall, we demonstrate that pregnancy facilitates maternal liver regeneration likely via triggering an early onset of hepatocyte replication, accumulating excessive liver fat, and promoting hepatocyte mitosis. The results from our current studies enable us to gain more insights into how maternal liver regeneration progresses during gestation.NEW & NOTEWORTHY We demonstrate that pregnancy may generate positive effects on maternal liver regeneration following partial hepatectomy, which are manifested by early entry of the cell cycle of remnant hepatocytes, increased hepatic fat accumulation, enhanced hepatocyte mitosis, and overall accelerated liver regrowth.


Asunto(s)
Hepatectomía , Regeneración Hepática/fisiología , Animales , Peso Corporal , Femenino , Hígado/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Mitosis , Tamaño de los Órganos , Embarazo
6.
Molecules ; 24(21)2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31661883

RESUMEN

The yield and quality of goji (Lycium barbarum L.) fruit are heavily dependent on fertilizer, especially the availability of nitrogen, phosphorus, and potassium (N, P, and K, respectively). In this study, we performed a metabolomic analysis of the response of goji berry to nitrogen fertilizer levels using an Ultra Performance Liquid Chromatography-Electrospray Ionization-Tandem Mass Spectrometry (UPLC-ESI-MS/MS) method. There was no significant difference in the fruit yield or the commodity grade between N0 (42.5 g/plant), N1 (85 g/plant), and N2 (127.5 g/plant). The primary nutrients of the goji berry changed with an increasing nitrogen fertilization. Comparative metabolomic profiling of three nitrogen levels resulted in the identification of 612 metabolites, including amino acids, flavonoids, carbohydrates, organic acids, and lipids/alcohols, among others, of which 53 metabolites (lipids, fatty acids, organic acids, and phenolamides) demonstrated significant changes. These results provide new insights into the molecular mechanisms of the relationship between yield and quality of goji berry and nitrogen fertilizer.


Asunto(s)
Fertilizantes , Frutas/metabolismo , Lycium/metabolismo , Metabolómica , Antioxidantes/química , Cromatografía Líquida de Alta Presión , Frutas/efectos de los fármacos , Lycium/efectos de los fármacos , Nitrógeno/farmacología , Extractos Vegetales/metabolismo , Espectrometría de Masas en Tándem
7.
J Org Chem ; 83(12): 6762-6768, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768006

RESUMEN

Efficient syntheses of 3,3-difluorooxindoles and 3-fluorooxindoles via fluorination of hydrazonoindolin-2-one with Selectfluor are reported. Under different solvent conditions, this method produced 3,3-difluorooxindoles and 3-fluorooxindoles selectively. The broad substrate scope and mild reaction conditions make this transformation a valuable method in drug discovery and development.

8.
J Pharmacol Exp Ther ; 358(1): 14-21, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27189962

RESUMEN

Transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates multiple biologic processes, including hepatic lipid metabolism. Estrogen exerts actions affecting energy homeostasis, including a liver fat-lowering effect. Increasing evidence indicates the crosstalk between these two molecules. The aim of this study was to evaluate whether Nrf2 modulates estrogen signaling in hepatic lipid metabolism. Nonalcoholic fatty liver disease (NAFLD) was induced in wild-type and Nrf2-null mice fed a high-fat diet and the liver fat-lowering effect of exogenous estrogen was subsequently assessed. We found that exogenous estrogen eliminated 49% and 90% of hepatic triglycerides in wild-type and Nrf2-null mice with NAFLD, respectively. This observation demonstrates that Nrf2 signaling is antagonistic to estrogen signaling in hepatic fat metabolism; thus, Nrf2 absence results in striking amplification of the liver fat-lowering effect of estrogen. In addition, we found the association of trefoil factor 3 and fatty acid binding protein 5 with the liver fat-lowering effect of estrogen. In summary, we identified Nrf2 as a novel and potent inhibitor of estrogen signaling in hepatic lipid metabolism. Our finding may provide a potential strategy to treat NAFLD by dually targeting Nrf2 and estrogen signaling.


Asunto(s)
Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Factor 2 Relacionado con NF-E2/deficiencia , Proteínas de Neoplasias/metabolismo , Animales , Western Blotting , Dieta Alta en Grasa , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Am J Physiol Gastrointest Liver Physiol ; 308(4): G262-8, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25524062

RESUMEN

The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates various cellular activities, including redox balance, detoxification, metabolism, autophagy, proliferation, and apoptosis. Several studies have demonstrated that Nrf2 regulates hepatocyte proliferation during liver regeneration. The aim of this study was to investigate how Nrf2 modulates the cell cycle of replicating hepatocytes in regenerating livers. Wild-type and Nrf2 null mice were subjected to 2/3 partial hepatectomy (PH) and killed at multiple time points for various analyses. Nrf2 null mice exhibited delayed liver regrowth, although the lost liver mass was eventually restored 7 days after PH. Nrf2 deficiency did not affect the number of hepatocytes entering the cell cycle but did delay hepatocyte mitosis. Mechanistically, the lack of Nrf2 resulted in increased mRNA and protein levels of hepatic cyclin A2 when the remaining hepatocytes were replicating in response to PH. Moreover, Nrf2 deficiency in regenerating livers caused dysregulation of Wee1, Cdc2, and cyclin B1 mRNA and protein expression, leading to decreased Cdc2 activity. Thus, Nrf2 is required for timely M phase entry of replicating hepatocytes by ensuring proper regulation of cyclin A2 and the Wee1/Cdc2/cyclin B1 pathway during liver regeneration.


Asunto(s)
División Celular , Hepatocitos/metabolismo , Regeneración Hepática , Hígado/metabolismo , Mitosis , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ciclina A2/genética , Ciclina A2/metabolismo , Ciclina B1/genética , Ciclina B1/metabolismo , Regulación de la Expresión Génica , Hepatectomía , Hepatocitos/patología , Cinética , Hígado/patología , Hígado/cirugía , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/metabolismo
10.
J Cell Sci ; 126(Pt 7): 1618-25, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23418358

RESUMEN

Pregnancy induces widespread adaptive responses in maternal organ systems including the liver. The maternal liver exhibits significant growth by increasing the number and size of hepatocytes, by largely unknown mechanisms. Nrf2 mediates cellular defense against oxidative stress and inflammation and also regulates liver regeneration. To determine whether Nrf2 is involved in the regulation of maternal hepatic adaptations to pregnancy, we assessed the proliferation and size of maternal hepatocytes and the associated molecular events in wild-type and Nrf2-null mice at various stages of gestation. We found that wild-type maternal hepatocytes underwent proliferation and size reduction during the first half, and size increase without overt replication during the second half, of pregnancy. Although pregnancy decreased Nrf2 activity in the maternal liver, Nrf2 deficiency caused a delay in maternal hepatocyte proliferation, concomitant with dysregulation of the activation of Cyclin D1, E1, and, more significantly, A2. Remarkably, as a result of Nrf2 absence, the maternal hepatocytes were largely prevented from reducing their sizes during the first half of pregnancy, which was associated with an increase in mTOR activation. During the second half of pregnancy, maternal hepatocytes of both genotypes showed continuous volume increase accompanied by persistent activation of mTOR. However, the lack of Nrf2 resulted in dysregulation of the activation of the mTOR upstream regulator AKT1 and the mTOR target p70SK6 and thus disruption of the AKT1/mTOR/p70S6K pathway, which is known to control cell size. This suggests an mTOR-dependent and AKT1- and p70S6K-independent compensatory mechanism when Nrf2 is deficient. In summary, our study demonstrates that Nrf2 is required for normal maternal hepatic adjustments to pregnancy by ensuring proper regulation of the number and size of maternal hepatocytes.


Asunto(s)
Hígado/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Western Blotting , Ciclina A2/metabolismo , Ciclina D1/metabolismo , Ciclina E/metabolismo , Femenino , Hepatocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Proteínas Oncogénicas/metabolismo , Embarazo
11.
Am J Pathol ; 184(4): 1152-66, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24655377

RESUMEN

Activin A, a member of the transforming growth factor-ß superfamily, provides pleiotropic regulation of fibrosis and inflammation. We aimed at determining whether selective inhibition of activin A would provide a regenerative benefit. The introduction of activin A into normal muscle increased the expression of inflammatory and muscle atrophy genes Tnf, Tnfrsf12a, Trim63, and Fbxo32 by 3.5-, 10-, 2-, and 4-fold, respectively. The data indicate a sensitive response of muscle to activin A. Two hours after cardiotoxin-induced muscle damage, local activin A protein expression increased by threefold to ninefold. Neutralization of activin A with a specific monoclonal antibody in this muscle injury model decreased the muscle protein levels of lymphotoxin α and Il17a by 32% and 42%, respectively. Muscle histopathological features showed that activin A antibody-treated mice displayed an increase in muscle degradation, with the concomitant 9.2-fold elevation in F4/80-positive cells 3 days after injury. At the same time, the number of Pax7/Myod1-positive cells also increased, indicative of potentiated muscle precursor activation. Ultimately, activin A inhibition resulted in rapid recovery of muscle contractile properties indicated by a restoration of maximum and specific force. In summary, selective inhibition of activin A with a monoclonal antibody in muscle injury leads to the early onset of tissue degradation and subsequent enhanced myogenesis, thereby accelerating muscle repair and functional recovery.


Asunto(s)
Activinas/antagonistas & inhibidores , Contracción Muscular/fisiología , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Animales , Electroporación , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunohistoquímica , Ratones Endogámicos C57BL , Regeneración/fisiología , Transcriptoma
12.
J Pharmacol Exp Ther ; 349(2): 355-71, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24627466

RESUMEN

Follistatin (FST) is a member of the tissue growth factor ß family and is a secreted glycoprotein that antagonizes many members of the family, including activin A, growth differentiation factor 11, and myostatin. The objective of this study was to explore the use of an engineered follistatin therapeutic created by fusing FST315 lacking heparin binding activity to the N terminus of a murine IgG1 Fc (FST315-ΔHBS-Fc) as a systemic therapeutic agent in models of muscle injury. Systemic administration of this molecule was found to increase body weight and lean muscle mass after weekly administration in normal mice. Subsequently, we tested this agent in several models of muscle injury, which were chosen based on their severity of damage and their ability to reflect clinical settings. FST315-ΔHBS-Fc treatment proved to be a potent inducer of muscle remodeling and regeneration. FST315-ΔHBS-Fc induced improvements in muscle repair after injury/atrophy by modulating the early inflammatory phase allowing for increased macrophage density, and Pax7-positive cells leading to an accelerated restoration of myofibers and muscle function. Collectively, these data demonstrate the benefits of a therapeutically viable form of FST that can be leveraged as an alternate means of ameliorating muscle regeneration.


Asunto(s)
Folistatina/farmacología , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Músculo Esquelético/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Regeneración , Animales , Folistatina/genética , Ratones , Músculo Esquelético/fisiología , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética
13.
Plant Physiol Biochem ; 206: 108285, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38145586

RESUMEN

Stomata are ports that facilitate gas and water vapor exchange during plant photosynthesis and transpiration. Stomatal development is strictly regulated by endogenous hormone. Jasmonate, an important signal that modulates multiple physiological processes in plants, has been found to negatively regulate stomatal development in Arabidopsis thaliana, yet the molecular mechanisms underlying stomata development signaling remain to be understood. Jasmonate ZIM-domain (JAZ) proteins are the members of TIFY family and the key component of JA signaling pathway. Its function in stomatal development is unclear to data. Here, we screened out 24 TIFY family members against the genome of Lycium, and identified a JAZ member by combination analyses of evolutionary tree, cis-elements in promoter and gene expression patterns. Overexpression of this gene (LrJAZ2) in Lycium ruthenicum and Arabidopsis thaliana indicated LrJAZ2 negatively regulates stomatal development. Microscopic observations revealed that overexpression of LrJAZ2 negatively regulated stomatal development by decreasing stomatal density and index, which may lead to lower leaf transpiration rates. Transcriptome data indicated the overexpression of LrJAZ2 up-regulated the stomatal related genes such as LrERL2, LrPYL4, and down-regulated the LrSPCH. Collectively, our study found that LrJAZ2 is a key gene in stomatal development regulation in L. ruthenicum and provided new insights into the regulation of stomatal development.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Lycium , Arabidopsis/genética , Arabidopsis/metabolismo , Lycium/genética , Ciclopentanos/metabolismo , Oxilipinas/metabolismo , Regulación de la Expresión Génica de las Plantas , Proteínas de Arabidopsis/genética
14.
Hepatol Res ; 43(8): 876-89, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23279269

RESUMEN

AIM: Partial hepatectomy (PH) induces robust hepatic regenerative and metabolic responses that are considered to be triggered by humoral factors. The aim of the study was to identify plasma protein factors that potentially trigger or reflect the body's immediate-early responses to liver mass reduction. METHODS: Male C57BL/6 mice were subjected to sham operation, 70% PH or 90% PH. Blood was collected from the inferior vena cava at 20, 60 and 180 min after surgery. RESULTS: Using a label-free quantitative mass spectrometry-based proteomics approach, we identified 399 proteins exhibiting significant changes in plasma expression between any two groups. Of the 399 proteins, 167 proteins had multiple unique sequences and high peptide ID confidence (>90%) and were defined as priority 1 proteins. A group of plasma proteins largely associated with metabolism is enriched after 70% PH. Among the plasma proteins that respond to 90% PH are a dominant group of proteins that are also associated with metabolism and one known cytokine (platelet factor 4). Ninety percent PH and 70% PH induces similar changes in plasma protein profile. CONCLUSION: Our findings enable us to gain insight into the immediate-early response of plasma proteins to liver mass loss. Our data support the notion that increased metabolic demands of the body after massive liver mass loss may function as a sensor that calibrates hepatic regenerative response.

15.
Biochem Pharmacol ; 214: 115668, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37364623

RESUMEN

Activins are a subgroup of the TGFß superfamily of growth and differentiation factors, dimeric in nature and consisting of two inhibin beta subunits linked via a disulfide bridge. Canonical activin signaling occurs through Smad2/3, with negative feedback initiated by Smad6/7 following signal transduction, which binds activin type I receptor preventing phosphorylation of Smad2/3 and activation of downstream signaling. In addition to Smad6/7, other inhibitors of activin signaling have been identified as well, including inhibins (dimers of an inhibin alpha and beta subunit), BAMBI, Cripto, follistatin, and follistatin-like 3 (fstl3). To date, activins A, B, AB, C, and E have been identified and isolated in mammals, with activin A and B having the most characterization of biological activity. Activin A has been implicated as a regulator of several important functions of liver biology, including hepatocyte proliferation and apoptosis, ECM production, and liver regeneration; the role of other subunits of activin in liver physiology are less understood. There is mounting data to suggest a link between dysregulation of activins contributing to various hepatic diseases such as inflammation, fibrosis, and hepatocellular carcinoma, and emerging studies demonstrating the protective and regenerative effects of inhibiting activins in mouse models of liver disease. Due to their importance in liver biology, activins demonstrate utility as a therapeutic target for the treatment of hepatic diseases such as cirrhosis, NASH, NAFLD, and HCC; further research regarding activins may provide diagnostic or therapeutic opportunity for those suffering from various liver diseases.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Ratones , Animales , Folistatina , Activinas/fisiología , Receptores de Activinas , Mamíferos
16.
Food Chem ; 428: 136797, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37418879

RESUMEN

Fresh-eating walnuts are perishable and become mildewed during shelf life, limiting their sales span. The effects of chlorine dioxide (ClO2) alone and its combination with walnut green husk extract (WGHE) on shelf stored fresh walnuts were investigated to develop a pollution-free preservative for the produce. The initial development of mildew incidence was delayed by both treatments under 25 °C, whereas, WGHE + ClO2 acted more effectively than ClO2 under 5 °C. The WGHE + ClO2 treatment presented superior effects on improving moisture, soluble sugar and total phenol content, alleviating loss of oil and unsaturated fatty acid and delaying peroxide value increase of walnut kernels at both temperatures. Both treatments inhibited the activities of three lipolytic enzymes and two oxidases at 25 °C and 5 °C, WGHE + ClO2 acted more effectively at 5 °C. The results guide the combined application of WGHE with ClO2 on shelf preservation of fresh walnut.


Asunto(s)
Juglans , Antioxidantes/farmacología , Óxidos/farmacología , Extractos Vegetales/farmacología , Cloro
17.
Biomedicines ; 11(7)2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37509548

RESUMEN

Patients with end-stage liver disease exhibit progressive skeletal muscle atrophy, highlighting a negative crosstalk between the injured liver and muscle. Our study was to determine whether TGFß ligands function as the mediators. Acute or chronic liver injury was induced by a single or repeated administration of carbon tetrachloride. Skeletal muscle injury and repair was induced by intramuscular injection of cardiotoxin. Activin type IIB receptor (ActRIIB) ligands and growth differentiation factor 8 (Gdf8) were neutralized with ActRIIB-Fc fusion protein and a Gdf8-specific antibody, respectively. We found that acute hepatic injury induced rapid and adverse responses in muscle, which was blunted by neutralizing ActRIIB ligands. Chronic liver injury caused muscle atrophy and repair defects, which were prevented or reversed by inactivating ActRIIB ligands. Furthermore, we found that pericentral hepatocytes produce excessive Gdf8 in injured mouse liver and cirrhotic human liver. Specific inactivation of Gdf8 prevented liver injury-induced muscle atrophy, similar to neutralization of ActRIIB ligands. Inhibition of Gdf8 also reversed muscle atrophy in a treatment paradigm following chronic liver injury. Direct injection of exogenous Gdf8 protein into muscle along with acute focal muscle injury recapitulated similar dysregulated muscle regeneration as that observed with liver injury. The results indicate that injured liver negatively communicate with the muscle largely via Gdf8. Unexpectedly, inactivation of Gdf8 simultaneously ameliorated liver fibrosis in mice following chronic liver injury. In vitro, Gdf8 induced human hepatic stellate (LX-2) cells to form a septa-like structure and stimulated expression of profibrotic factors. Our findings identified Gdf8 as a novel hepatomyokine contributing to injured liver-muscle negative crosstalk along with liver injury progression.

18.
Cell Mol Gastroenterol Hepatol ; 13(1): 35-55, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34438112

RESUMEN

BACKGROUND & AIMS: Maternal liver shows robust adaptations to pregnancy to accommodate the metabolic needs of the developing and growing placenta and fetus by largely unknown mechanisms. We found that Ascl1, a gene encoding a basic helix-loop-helix transcription factor essential for neuronal development, is highly activated in maternal hepatocytes during the second half of gestation in mice. METHODS: To investigate whether and how Ascl1 plays a pregnancy-dependent role, we deleted the Ascl1 gene specifically in maternal hepatocytes from midgestation until term. RESULTS: As a result, we identified multiple Ascl1-dependent phenotypes. Maternal livers lacking Ascl1 showed aberrant hepatocyte structure, increased hepatocyte proliferation, enlarged hepatocyte size, reduced albumin production, and increased release of liver enzymes, indicating maternal liver dysfunction. Simultaneously, maternal pancreas and spleen and the placenta showed marked overgrowth; and the maternal ceca microbiome showed alterations in relative abundance of several bacterial subpopulations. Moreover, litters born from maternal hepatic Ascl1-deficient dams experienced abnormal postnatal growth after weaning, implying an adverse pregnancy outcome. Mechanistically, we found that maternal hepatocytes deficient for Ascl1 showed robust activation of insulin-like growth factor 2 expression, which may contribute to the Ascl1-dependent phenotypes widespread in maternal and uteroplacental compartments. CONCLUSIONS: In summary, we show that maternal liver, via activating Ascl1 expression, modulates the adaptations of maternal organs and the growth of the placenta to maintain a healthy pregnancy. Our studies show that Ascl1 is a novel and critical regulator of the physiology of pregnancy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Hígado , Embarazo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Femenino , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hígado/fisiología , Ratones
19.
PLoS One ; 17(6): e0269383, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35696363

RESUMEN

The transcription factor Nrf2 modulates the initiation and progression of a number of diseases including liver disorders. We evaluated whether Nrf2 mediates hepatic adaptive responses to cholestasis. Wild-type and Nrf2-null mice were subjected to bile duct ligation (BDL) or a sham operation. As cholestasis progressed to day 15 post-BDL, hepatocytes in the wild-type mice exhibited a tendency to dedifferentiate, indicated by the very weak expression of hepatic progenitor markers: CD133 and tumor necrosis factor-like weak induced apoptosis receptor (Fn14). During the same period, Nrf2 deficiency augmented this tendency, manifested by higher CD133 expression, earlier, stronger, and continuous induction of Fn14 expression, and markedly reduced albumin production. Remarkably, as cholestasis advanced to the late stage (40 days after BDL), hepatocytes in the wild-type mice exhibited a Fn14+ phenotype and strikingly upregulated the expression of deleted in malignant brain tumor 1 (DMBT1), a protein essential for epithelial differentiation during development. In contrast, at this stage, hepatocytes in the Nrf2-null mice entirely inhibited the upregulation of DMBT1 expression, displayed a strong CD133+/Fn14+ phenotype indicative of severe dedifferentiation, and persistently reduced albumin production. We revealed that Nrf2 maintains hepatocytes in the differentiated state potentially via the increased activity of the Nrf2/DMBT1 pathway during cholestasis.


Asunto(s)
Colestasis Extrahepática , Colestasis , Factor 2 Relacionado con NF-E2/metabolismo , Albúminas/metabolismo , Animales , Conductos Biliares/patología , Diferenciación Celular , Colestasis/metabolismo , Colestasis Extrahepática/patología , Hepatocitos/metabolismo , Ligadura , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Hepatol Commun ; 6(10): 2812-2826, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35866567

RESUMEN

The role of activin B, a transforming growth factor ß (TGFß) superfamily cytokine, in liver health and disease is largely unknown. We aimed to investigate whether activin B modulates liver fibrogenesis. Liver and serum activin B, along with its analog activin A, were analyzed in patients with liver fibrosis from different etiologies and in mouse acute and chronic liver injury models. Activin B, activin A, or both was immunologically neutralized in mice with progressive or established carbon tetrachloride (CCl4 )-induced liver fibrosis. Hepatic and circulating activin B was increased in human patients with liver fibrosis caused by several liver diseases. In mice, hepatic and circulating activin B exhibited persistent elevation following the onset of several types of liver injury, whereas activin A displayed transient increases. The results revealed a close correlation of activin B with liver injury regardless of etiology and species. Injured hepatocytes produced excessive activin B. Neutralizing activin B largely prevented, as well as improved, CCl4 -induced liver fibrosis, which was augmented by co-neutralizing activin A. Mechanistically, activin B mediated the activation of c-Jun-N-terminal kinase (JNK), the induction of inducible nitric oxide synthase (iNOS) expression, and the maintenance of poly (ADP-ribose) polymerase 1 (PARP1) expression in injured livers. Moreover, activin B directly induced a profibrotic expression profile in hepatic stellate cells (HSCs) and stimulated these cells to form a septa structure. Conclusions: We demonstrate that activin B, cooperating with activin A, mediates the activation or expression of JNK, iNOS, and PARP1 and the activation of HSCs, driving the initiation and progression of liver fibrosis.


Asunto(s)
Tetracloruro de Carbono , Ribosa , Activinas , Adenosina Difosfato/efectos adversos , Animales , Tetracloruro de Carbono/toxicidad , Humanos , Cirrosis Hepática/inducido químicamente , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ribosa/efectos adversos , Factor de Crecimiento Transformador beta/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA