RESUMO
The regulation of the mammalian circadian clock is largely dependent on heredity. In model animals for circadian rhythm studies, C57BL/6 and BALB/c mice exhibit considerable differences in their adaptation to circadian disruption, yet deeper comparisons remain unexplored. Here, we have established embryonic fibroblast cells derived from C57BL/6 mice (MEF) and BALB/c (BALB/3T3) mice, which have been transfected with the Bmal1 promoter-driven luciferase (Bmal1-Luc) reporter gene. Next, dexamethasone was applied for various cyclic stimulations, which revealed that Bmal1 bioluminescence of MEF cells was entrained to 24 to 26 h cycles, whereas BALB/3T3 cells have a wider range (22 to 28 h) with lower amplitudes. Behaviorally, BALB/c mice swiftly adapted to a 6-h advance light/dark cycle, unlike C57BL/6 mice. Furthermore, we found the expression of the circadian rhythm gene Npas2 in BALB/c mice is significantly lower than that in C57BL/6 mice. This observation is consistent with the differentially expressed genes (DEGs) in the intestine and lung tissues of C57BL/6 and BALB/c mice, based on the RNA-seq datasets downloaded from the Gene Expression Omnibus (GEO). In summary, our study uncovers that BALB/c mice possess greater resilience in circadian rhythm than C57BL/6 mice, both cellular and behaviorally, identifying potential genes underlying this difference.
Assuntos
Fatores de Transcrição ARNTL , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Relógios Circadianos , Ritmo Circadiano , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Animais , Camundongos , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Relógios Circadianos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fibroblastos/metabolismo , Fotoperíodo , Comportamento Animal , Dexametasona/farmacologia , Especificidade da Espécie , Adaptação Fisiológica/genética , Células 3T3 BALB , Proteínas do Tecido NervosoRESUMO
Deprivation of sleep (DS) and its effects on circadian rhythm gene expression are not well understood despite their influence on various physiological and psychological processes. This study aimed to elucidate the changes in the expression of circadian rhythm genes following a night of sleep and DS. Their correlation with sleep architecture and physical activity was also examined. The study included 81 participants who underwent polysomnography (PSG) and DS with actigraphy. Blood samples were collected after PSG and DS. Expression levels of brain and muscle ARNT-like 1 (BMAL1), circadian locomotor output cycles kaput (CLOCK), neuronal PAS domain protein 2 (NPAS2), period 1 (PER1), cryptochrome 1 (CRY1) and nuclear receptor subfamily 1 group D member 1 (NR1D1) were analyzed using qRT-PCR. DS decreased the expression of CLOCK and BMAL1 while increasing PER1. PER1 expression correlated positively with total sleep time and non-rapid-eye-movement (NREM) sleep duration and negatively with sleep latency, alpha, beta and delta waves in the O1A2 lead. Physical activity during DS showed positive correlations with CLOCK, BMAL1, and CRY1. The findings highlight the role of PER1 in modulating sleep patterns, suggesting potential targets for managing sleep-related disorders. Further research is essential to deepen the understanding of these relationships and their implications.
Assuntos
Ritmo Circadiano , Privação do Sono , Sono , Humanos , Masculino , Ritmo Circadiano/genética , Feminino , Sono/genética , Sono/fisiologia , Adulto , Privação do Sono/genética , Privação do Sono/metabolismo , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Polissonografia , Criptocromos/genética , Criptocromos/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Regulação da Expressão Gênica , Exercício FísicoRESUMO
Seasonal affective disorder (SAD), also known as winter depression, is a subtype of depression typically manifesting in winter. Typical symptoms of SAD, such as an increased need for sleep and carbohydrate cravings associated with increased appetite and weight, are distinct from those of major depression, and the underlying mechanisms of SAD remain unclear. Although laboratory mice are generally considered non-seasonal animals, we observed depression-like behaviors in melatonin-proficient female CBA/N mice maintained under winter-mimicking conditions. Transcriptome analysis of the brains of CBA/N mice maintained under winter- and summer-mimicking conditions revealed changes in the expression of circadian clock genes, including Arntl (also known as Bmal1). We generated Arntl-deficient, melatonin-proficient CBA/N mice using the speed congenic method to examine the role of Arntl in depressive behavior. The tail suspension test in these mice revealed a depressive phenotype. These results suggested that the circadian clock gene Arntl may be involved in winter depression-like behavior.
Assuntos
Fatores de Transcrição ARNTL , Depressão , Melatonina , Camundongos Endogâmicos CBA , Animais , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Feminino , Camundongos , Melatonina/metabolismo , Depressão/genética , Depressão/metabolismo , Relógios Circadianos/genética , Estações do Ano , Comportamento Animal , Ritmo Circadiano/genéticaRESUMO
BACKGROUND: Biological synchronized rhythmicity is a critical physiological process. The lack of synchronized rhythms, mainly those showing a circadian basis, like sleep, the heart rate (HR) and arterial blood pressure (BP), often leads to several organic challenges, usually associated with adverse outcomes. OBJECTIVES: The aim of the study was to investigate whether the intensive care unit (ICU) environment favors clock genes and cardiorespiratory changes. MATERIAL AND METHODS: A total of 22 critically ill patients (16 males; 72.73%) with a mean age of 60.82 ±20.07 years and well-established cardiovascular conditions were selected from ICU. Blood samples were obtained, and total RNA was isolated and reverse-transcribed into complementary DNA (cDNA). A quantitative polymerase chain reaction (qPCR) was performed to assess the target gene expression levels. The urinary concentration levels of melatonin (MEL) were assessed. The heart rate, BP (systolic - SBP, diastolic - DBP and mean - MBP) and the oxygen saturation (SpO2) levels were assessed as continuous variables. RESULTS: The urinary MEL and Brain and muscle Arnt-like protein-1 (BMAL1) levels were shown to have a non-linear relationship with HR (coefficient (coef): 2.318, p = 0.032; coef: 2.722, p = 0.006, respectively) and SBP (coef: 1.000, p = 0.008; coef: 2.000, p = 0.037, respectively), with an explanatory power of up to 50.3% and 39.7% of the HR and SBP variability, respectively. Melatonin, but not BMAL1, was also shown to have a non-linear relationship with MBP (coef: 1.000, p = 0.007), with an explanatory power of up to 31.3% regarding the MBP variability. The HR and SBP oscillatory dynamics was shown to be related to changes in the genetic expression of BMAL1 and the urinary MEL concentrations. To a lower degree, MEL also impacted the variation of MBP. CONCLUSIONS: Our results suggest that not only are circadian functional matrices crucial for the dynamics of vital parameters in critically ill patients, but also that routinely assessed cardiovascular parameters like HR and BP may constitute important markers for the circadian timing system function. These parameters are easy to assess and have a relevant prognostic value regarding recovery outcomes, as well as the morbidity and mortality rates in ICU.
Assuntos
Fatores de Transcrição ARNTL , Biomarcadores , Pressão Sanguínea , Relógios Circadianos , Estado Terminal , Frequência Cardíaca , Unidades de Terapia Intensiva , Melatonina , Humanos , Masculino , Pessoa de Meia-Idade , Feminino , Melatonina/sangue , Melatonina/metabolismo , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Pressão Sanguínea/fisiologia , Biomarcadores/sangue , Idoso , Saturação de Oxigênio , Ritmo Circadiano/fisiologiaRESUMO
BACKGROUND: Many patients with bipolar disorder (BD) do not respond to or have difficulties tolerating lithium and/or other mood stabilizing agents. There is a need for personalized treatments based on biomarkers in guiding treatment options. The calcium voltage-gated channel CACNA1C is a promising candidate for developing personalized treatments. CACNA1C is implicated in BD by genome-wide association studies and several lines of evidence suggest that targeting L-type calcium channels could be an effective treatment strategy. However, before such individualized treatments can be pursued, biomarkers predicting treatment response need to be developed. METHODS: As a first step in testing the hypothesis that CACNA1C genotype is associated with serum levels of CACNA1C, we conducted ELISA measures on serum samples from 100 subjects with BD and 100 control subjects. RESULTS: We observed significantly higher CACNA1C (p < 0.01) protein levels in subjects with BD. The risk single nucleotide polymorpshism (SNP) (rs11062170) showed functional significance as subjects homozygous for the risk allele (CC) had significantly greater CACNA1C protein levels compared to subjects with one (p = 0.013) or no copies (p = 0.009). We observed higher somatostatin (SST) (p < 0.003) protein levels and lower levels of the clock protein aryl hydrocarbon receptor nuclear translocator-like (ARTNL) (p < 0.03) and stress signaling factor corticotrophin releasing hormone (CRH) (p < 0.001) in BD. SST and period 2 (PER2) protein levels were associated with both alcohol dependence and lithium response. CONCLUSIONS: Our findings represent the first evidence for increased serum levels of CACNA1C in BD. Along with altered levels of SST, ARNTL, and CRH our findings suggest CACNA1C is associated with circadian rhythm and stress response disturbances in BD.
Assuntos
Transtorno Bipolar , Canais de Cálcio Tipo L , Ritmo Circadiano , Polimorfismo de Nucleotídeo Único , Humanos , Transtorno Bipolar/sangue , Transtorno Bipolar/genética , Canais de Cálcio Tipo L/genética , Feminino , Masculino , Adulto , Pessoa de Meia-Idade , Ritmo Circadiano/fisiologia , Genótipo , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/genética , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Somatostatina/sangue , AlelosRESUMO
In mammals, the central circadian oscillator is located in the suprachiasmatic nucleus (SCN). Hypothalamus-pituitary-thyroid axis components exhibit circadian oscillation, regulated by both central clock innervation and intrinsic circadian clocks in the anterior pituitary and thyroid glands. Thyroid disorders alter the rhythmicity of peripheral clocks in a tissue-dependent response; however, whether these effects are influenced by alterations in the master clock remains unknown. This study aimed to characterize the effects of hypothyroidism on the rhythmicity of SCN, body temperature (BT) and metabolism, and the possible mechanisms involved in this signalling. C57BL/6J adult male mice were divided into Control and Hypothyroid groups. Profiles of spontaneous locomotor activity (SLA), BT, oxygen consumption ( V Ì O 2 ${{\dot{V}}_{{{{\mathrm{O}}}_{\mathrm{2}}}}}$ ) and respiratory quotient (RQ) were determined under free-running conditions. Clock gene expression, and neuronal activity of the SCN and medial preoptic nucleus (MPOM) area were investigated in light-dark (LD) conditions. Triiodothyronine (T3) transcriptional regulation of Bmal1 promoter activity was evaluated in GH3-transfected cells. Hypothyroidism delayed the rhythmicity of SLA and BT, and altered the expression of core clock components in the SCN. The activity of SCN neurons and their outputs were also affected, as evidenced by the loss of circadian rhythmicity in V Ì O 2 ${{\dot{V}}_{{{{\mathrm{O}}}_{\mathrm{2}}}}}$ and RQ and alterations in the neuronal activity pattern of MPOM. In GH3 cells, T3 increased Bmal1 promoter activity in a time-dependent manner. Thyroid hormone may act as a temporal cue for the central circadian clock, and the uncoupling of central and peripheral clocks might contribute to a wide range of metabolic and thermoregulatory impairments observed in hypothyroidism. KEY POINTS: Hypothyroidism alters clock gene expression in the suprachiasmatic nucleus (SCN). Thyroid hypofunction alters the phase of spontaneous locomotor activity and body temperature rhythms. Thyroid hormone deficiency alters the daily pattern of SCN and medial preoptic nucleus neuronal activities. Hypothyroidism alterations are extended to daily oscillations of oxygen consumption and metabolism, which might contribute to the development of metabolic syndrome. Triiodothyronine increases Bmal1 promoter activity acting as temporal cue for the central circadian clock.
Assuntos
Fatores de Transcrição ARNTL , Hipotireoidismo , Camundongos Endogâmicos C57BL , Núcleo Supraquiasmático , Tri-Iodotironina , Animais , Masculino , Hipotireoidismo/fisiopatologia , Hipotireoidismo/metabolismo , Hipotireoidismo/genética , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Camundongos , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiologia , Ritmo Circadiano/fisiologia , Temperatura Corporal/fisiologia , Relógios Circadianos/genética , Relógios Circadianos/fisiologia , Regulação da Expressão GênicaRESUMO
BACKGROUND: The role of BMAL1 in various diseases remains unclear, particularly its impact on sepsis-induced acute kidney injury (AKI). This study aims to investigate the role of BMAL1 in sepsis-induced AKI and its potential effects on cell ferroptosis. Initially, we assessed BMAL1 expression levels in mice treated with sepsis-induced AKI (via LPS injection) and in LPS-stimulated renal tubular epithelial cells. Subsequently, we explored the correlation between BMAL1 and ferroptosis using sequencing technology, validating our findings throughout experimental approaches. To further elucidate BMAL1's specific effects on AKI-related ferroptosis, we constructed BMAL1 overexpression models in mice and cells, analysing its impact on AKI and ferroptosis both in vivo and in vitro. Furthermore, using transcriptome sequencing technology, we identified key BMAL1-regulated genes and their associated biological pathways, validating these findings through in vivo and in vitro experiments. RESULTS: Our findings indicate decreased BMAL1 expression in sepsis-induced AKI. BMAL1 overexpression effectively mitigated renal tubular injury by reducing ferroptosis levels in renal tubular epithelial cells. Using transcriptome sequencing and ChIP-qPCR technology, we identified YAP as a target of BMAL1. The overexpression of BMAL1 significantly reduced the transcriptional activity of YAP and inhibited the Hippo signalling pathway. Treatment with the Hippo inhibitor Verteporfin (VP) reversed the BMAL1-downregulation-induced damage. Additionally, our study revealed that YAP positively regulates ACSL4 gene expression and its downstream signalling pathways. CONCLUSION: This study demonstrates that BMAL1 overexpression alleviates renal tubular epithelial cell injury and ferroptosis by inhibiting YAP expression and the Hippo pathway, thereby exerting protective effects in sepsis-induced AKI. These findings underscore the therapeutic potential of targeting BMAL1 in managing sepsis-induced AKI.
Assuntos
Fatores de Transcrição ARNTL , Injúria Renal Aguda , Ferroptose , Camundongos Endogâmicos C57BL , Sepse , Animais , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/etiologia , Sepse/complicações , Sepse/metabolismo , Camundongos , Masculino , Linhagem Celular , Humanos , Modelos Animais de Doenças , Transdução de Sinais , Proteínas de Sinalização YAP , Células Epiteliais/metabolismo , LipopolissacarídeosRESUMO
Disruption of circadian rhythms contributes to deficits in cognitive functions during aging. Up to date, the biochemical, molecular and chronobiological bases of such deterioration have not been completely elucidated. Here, we aim: 1) to investigate the endogenous nature of 24 h-rhythms of antioxidant defenses, oxidative stress, clock's, and neurotrophic factors expression, in the rat temporal cortex (TC), and 2) to study the consequences of aging on the circadian organization of those factors. We observed a circadian organization of antioxidant enzymes activity, lipoperoxidation and the clock, BMAL1 and RORa, proteins, in the TC of young rats. Such temporal organization suggests the existence of a two-way communication among clock transcription factors and antioxidant defenses. This might generate the rhythmic and circadian expression of Bdnf and Rc3 genes involved in the TC-depending cognitive function. Noteworthy, such circadian organization disappears in the TC of aged rats. Aging also reduces glutathione peroxidase activity and expression, and it increases lipid peroxidation, throughout a 24 h-period. An increased oxidative stress makes the cellular redox environment change into an oxidative status which alters the endogenous clock activity and disrupts the circadian organization of, at least part, of the molecular basis of the synaptic plasticity in the TC.
Assuntos
Fatores de Transcrição ARNTL , Envelhecimento , Proteínas CLOCK , Ritmo Circadiano , Peroxidação de Lipídeos , Estresse Oxidativo , Ratos Wistar , Lobo Temporal , Animais , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Masculino , Ritmo Circadiano/fisiologia , Lobo Temporal/metabolismo , Proteínas CLOCK/metabolismo , Proteínas CLOCK/genética , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Ratos , Estresse Oxidativo/fisiologia , Peroxidação de Lipídeos/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Antioxidantes/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Peroxidase/genética , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/genética , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Expressão GênicaRESUMO
BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is a primary histological type of esophageal carcinoma with high morbidity. Aryl hydrocarbon receptor nuclear translocator-like (ARNTL) is a circadian clock gene associated with the progression of multiple tumors. However, its roles and mechanisms in ESCC remain unknown. METHODS: ARNTL expression was analyzed using TCGA database and detected using qRT-PCR, and ARNTL-related pathways were analyzed through GSEA. Cell functional behaviors were assessed in vitro by measuring cell viability, proliferation, and apoptosis. Cell growth in the murine model was investigated through xenograft model and immunofluorescence assays of PCNA and Ki67. The downstream targets of ARNTL were analyzed through sequencing and identified via luciferase report, ChIP, and RNA pull-down analyses. Dual-specificity protein phosphatase-1 (DUSP1) expression was analyzed using GEO datasets and measured using qRT-PCR and western blotting. Protein expression was examined via western blotting. RESULTS: ARNTL expression was decreased in esophageal carcinoma and associated with histological types, and elevated expression of ARNTL repressed ESCC cell viability and proliferation and facilitated cell apoptosis. ARNTL upregulation reduced tumor cell growth in murine models and decreased PCNA and Ki67 levels. Furthermore, DUSP1 was downregulated upon ARNTL silencing in ESCC. ARNTL could bind and positively regulate DUSP1 transcription. Additionally, DUSP1 silencing reversed the influences of ARNTL upregulation on cell viability, proliferation, and apoptosis in ESCC cells. ARNTL attenuated the activation of the ERK signaling by decreasing ERK phosphorylation through upregulation of DUSP1. CONCLUSION: ARNTL hinders cell growth and contributes to cell apoptosis by inactivating ERK signaling through transcriptional upregulation of DUSP1 in ESCC.
⢠ARNTL is differentially expressed in ESCC and associated with cell apoptosis.⢠ARNTL augments cell apoptosis.⢠ARNTL increases DUSP1 transcription.⢠ARNTL inhibits activation of the ERK signaling by upregulating DUSP1.⢠DUSP1 silencing reverses the effects of ARNTL in esophageal squamous cell carcinoma.
Assuntos
Fatores de Transcrição ARNTL , Progressão da Doença , Fosfatase 1 de Especificidade Dupla , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Sistema de Sinalização das MAP Quinases , Animais , Humanos , Masculino , Camundongos , Apoptose , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Fosfatase 1 de Especificidade Dupla/metabolismo , Fosfatase 1 de Especificidade Dupla/genética , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Endogâmicos BALB CRESUMO
Aim: This study investigated the role of lncRNA LINC01232 in ferroptosis of colorectal cancer (CRC).Materials & methods: Real time quantitative polymerase chain reaction or western blot experiments were performed to examine relevant mRNAs and proteins expression. The kit assays evaluated malondialdehyde, iron, Fe2+ and glutathione levels. ROS levels were verified by flow cytometry. Chromatin immunoprecipitation and RNA immunoprecipitation analysis monitored the correlation among LINC01232, H3K27ac, p300 and ARNTL2.Results: LINC01232 or ARNTL2 knockdown facilitated erastin-induced ferroptosis. The interaction between LINC01232 and p300 resulted in the enhancement of H3K27ac levels at ARNTL2 promoter to promote ARNTL2 transcriptional activity. ARNTL2 overexpression reversed the promoting effect of LINC01232 knockdown on ferroptosis.Conclusion: LINC01232 inhibited the ferroptosis in CRC by epigenetically upregulating the transcriptional activity of ARNTL2.
Colorectal cancer (CRC) is a malignant disease of the digestive tract that occurs worldwide, which has high morbidity and mortality but has not effective targeted therapy. Ferroptosis has emerged as a new target for treating CRC since its proposed in 2012. Long noncoding RNAs are noncoding RNAs with a length greater than 200 nucleotides and their role in ferroptosis of cancer cells has attracted more and more attention in recent years. Herein, our study explored the effect of long noncoding RNA LINC01232 on CRC progression. This research exhibited the relationship between LINC01232 and the ferroptosis at the occurrence and development of CRC, which is expected to provide a potential therapeutic target for the treatment of CRC.
Assuntos
Fatores de Transcrição ARNTL , Neoplasias Colorretais , Ferroptose , RNA Longo não Codificante , Ativação Transcricional , Humanos , Ferroptose/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , RNA Longo não Codificante/genética , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Histonas/metabolismo , Proteína p300 Associada a E1A/metabolismo , Proteína p300 Associada a E1A/genética , Regulação Neoplásica da Expressão Gênica , Linhagem Celular TumoralRESUMO
To explore the action mechanism of berberine in improving adipocytic insulin resistance(IR) by mediating brain and muscle arnt-like 1(BMAL1): circadian locomotor output cycles kaput(CLOCK) complex and regulating glucose and lipid metabolism. After the IR-3T3-L1 adipocyte model was established by dexamethasone induction for 96 h, 0.5, 1, 5, 10, and 20 µmol·L~(-1) berberine was administered for 24 h. The glucose oxidase method and cell counting kit-8(CCK-8) were used to detect extracellular glucose content and cell viability, respectively. The triglyceride(TG) and glycerol contents were detected by enzyme colorimetry. Oil red O staining was used to detect lipid droplets, and fluorescence staining was used to detect Ca~(2+), mitochondrial structure, and reactive oxygen species(ROS). Adiponectin(ADPN), BMAL1, CLOCK, hormone-sensitive triglyceride lipase(HSL), carbohydrate-response element-binding protein(ChREBP), sterol regulatory element-binding protein 1C(SREBP-1C), peroxisome proliferator-activated receptor γ coactivator 1α(PGC1α), carnitine palmitoyl transferase 1α(CPT1α), and peroxisome proliferator-activated receptor α(PPARα) were detected by Western blot(WB). Moreover, the nuclear localization of BMAL1 was detected by immunofluorescence. In addition, 20 µmol·L~(-1) CLK8 inhibitor was added to detect glucose consumption and BMAL1/ChREBP/PPARα protein. The results showed that berberine increased glucose consumption in IR-3T3-L1 adipocytes without affecting cell viability and reduced TG content. In addition, 5 µmol·L~(-1) berberine increased glycerol content and reduced lipid droplet accumulation due to enhanced lipolysis, while 10 µmol·L~(-1) berberine did not affect glycerol content, and fewer lipid droplets were observed due to enhanced lipolysis and glycerol utilization. Berberine improved mitochondrial function by reducing intracellular Ca~(2+) and ROS in IR-3T3-L1 adipocytes and upregulated PGC1α to improve the mitochondrial structure. The results also showed that berberine elevated ADPN to increase the insulin sensitivity of IR-3T3-L1 adipocytes, upregulated peripheral rhythm-related proteins BMAL1 and CLOCK, and strengthened the nuclear localization of BMAL1. In addition, berberine increased key lipolysis protein and lipid oxidation rate-limiting enzyme CPT1α and downregulated the key protein of TG synthesis, SREBP-1C. Moreover, ChREBP and PPARα in IR-3T3-L1 adipocytes were upregula-ted. All the above results suggested that berberine may transform glucose into lipids to enhance the hypoglycemic effect. By considering that CLK8 specifically inhibited the CLOCK acylation to modify BMAL1 and form complex, the results showed that the addition of CLK8 to the berberine group reduced glucose consumption, which suggested that berberine upregulated the formation of BMAL1:CLOCK complex to improve glucose metabolism. The addition of CLK8 to the berberine group upregulated BMAL1 but downregulated ChREBP and PPARα, which suggested that berberine mediated BMAL1:CLOCK complex for the regulation of glucose and lipid metabo-lism to improve adipocytic IR.
Assuntos
Células 3T3-L1 , Fatores de Transcrição ARNTL , Adipócitos , Berberina , Proteínas CLOCK , Glucose , Resistência à Insulina , Metabolismo dos Lipídeos , Animais , Camundongos , Metabolismo dos Lipídeos/efeitos dos fármacos , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Berberina/farmacologia , Adipócitos/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/citologia , Glucose/metabolismo , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Triglicerídeos/metabolismoRESUMO
Purpose: Bone loss is a common complication of type 2 diabetes mellitus (T2DM). Circadian rhythms play a significant role in T2DM and bone remodeling. Eldecalcitol (ED-71), a novel active vitamin D analog, has shown promise in ameliorating T2DM. We aimed to investigate whether the circadian rhythm coregulator BMAL1 mediates the anti-osteoporotic effect of ED-71 in T2DM and its associated mechanisms. Methods: A T2DM mouse model was established using high-fat diet (HDF) and streptozotocin (STZ) injection, and blood glucose levels were monitored weekly. HE staining, Masson staining, and Micro-CT were performed to assess the changes in bone mass. IHC staining and IF staining were used to detect osteoblast status and BMAL1 expression and RT-qPCR was applied to detect the change of oxidative stress factors. In vitro, high glucose (HG) stimulation was used to simulate the cell environment in T2DM. RT-qPCR, Western blot, IF, ALP staining and AR staining were used to detect osteogenic differentiation and SIRT1/GSK3ß signaling pathway. DCFH-DA staining was used to detect reactive oxygen species (ROS) levels. Results: ED-71 increased bone mass and promoted osteogenesis in T2DM mice. Moreover, ED-71 inhibited oxidative stress and promoted BMAL1 expression in osteoblasts The addition of STL1267, an agonist of the BMAL1 transcriptional repressor protein REV-ERB, reversed the inhibitory effect of ED-71 on oxidative stress and the promotional effect on osteogenic differentiation. In addition, ED-71 facilitated SIRT1 expression and reduced GSK3ß activity. The inhibition of SIRT1 with EX527 partially attenuated ED-71's effects, whereas the GSK3ß inhibitor LiCl further enhanced ED-71's positive effects on BMAL1 expression. Conclusion: ED-71 ameliorates bone loss in T2DM by upregulating the circadian rhythm coregulator BMAL1 and promoting osteogenesis through inhibition of oxidative stress. The SIRT1/GSK3ß signaling pathway is involved in the regulation of BMAL1.
Assuntos
Fatores de Transcrição ARNTL , Ritmo Circadiano , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Camundongos Endogâmicos C57BL , Osteogênese , Regulação para Cima , Animais , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Camundongos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Osteogênese/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Masculino , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Ritmo Circadiano/efeitos dos fármacos , Estreptozocina , Vitamina D/farmacologia , Vitamina D/análogos & derivados , Dieta Hiperlipídica , Células CultivadasRESUMO
Nobiletin has been reported to protect against obesity-related metabolic disorders by enhancing the circadian rhythm; however its effects on lipid metabolism in adipose tissue are unclear. In this study, mice were fed with high-fat diet (HFD) for four weeks firstly and gavaged with 50 or 200 mg/kg bodyweight/day nobiletin at Zeitgeber time (ZT) 4 for another four weeks while still receiving HFD. At the end of the 8-week experimental period, the mice were sacrificed at ZT4 or ZT8 on the same day. Mature 3T3-L1 adipocytes were treated with nobiletin in the presence or absence of siBmal1, siRora, siRorc, SR8278 or SR9009. Nobiletin reduced the weight of white adipose tissue (WAT) and the size of adipocytes in WAT. At ZT4, nobiletin decreased the TG, TC and LDL-c levels and increased serum FFA level and glucose tolerance. Nobiletin triggered the lipolysis of mesenteric and epididymal WAT at both ZT4 and ZT16. Nobiletin increased the level of RORγ at ZT16, that of BMAL1 and PPARγ at ZT4, and that of ATGL at both ZT4 and ZT16. Nobiletin increased lipolysis and ATGL levels in 3T3-L1 adipocytes in Bmal1- or Rora/c- dependent manner. Dual luciferase assay indicated that nobiletin enhanced the transcriptional activation of RORα/γ on Atgl promoter and decreased the repression of RORα/γ on PPARγ-binding PPRE. Promoter deletion analysis indicated that nobiletin inhibited the suppression of PPARγ-mediated Atgl transcription by RORα/γ. Taken together, nobiletin elevated lipolysis in WAT by increasing ATGL levels through activating the transcriptional activity of RORα/γ and decreasing the repression of RORα/γ on PPARγ-binding PPRE.
Assuntos
Células 3T3-L1 , Tecido Adiposo Branco , Relógios Circadianos , Flavonas , Lipólise , Camundongos Endogâmicos C57BL , Animais , Flavonas/farmacologia , Lipólise/efeitos dos fármacos , Camundongos , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Masculino , Relógios Circadianos/efeitos dos fármacos , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Dieta Hiperlipídica/efeitos adversos , PPAR gama/metabolismo , PPAR gama/genética , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Lipase/metabolismo , Obesidade/metabolismo , Obesidade/tratamento farmacológico , Aciltransferases , Membro 3 do Grupo F da Subfamília 1 de Receptores NuclearesRESUMO
BACKGROUND: Basic helix-loop-helix ARNT like 2 (ARNTL2) is a transcription factor that controls the circadian rhythm. Amounts of studies have demonstrated the carcinogenic function of ARNTL2 in human malignant tumors albeit the underlying mechanisms remain poorly understood. We aimed to study the significance of ARNTL2 in bladder cancer (BLCA). METHODS: Immunohistochemical staining, immunoblotting and the database from TCGA were used to analyze the clinical relevance of ARNTL2, enolase 1 (ENO1) and solute carrier family 31 member 1 (SLC31A1) in BLCA. The function of ARNTL2 was explored by cell proliferation assay, apoptosis, colony formation and xenografted tumorigenesis. The molecular mechanisms of ARNTL2-driving BLCA development were investigated by RT-qPCR, immunoblotting and luciferase assays. Glycolysis was checked by measuring glucose consumption and lactate production. ENO1 activity was assessed by using indicated assay kit. RESULTS: Overexpression of ARNTL2 facilitates the proliferation and tumorigenesis of BLCA cells through suppression of apoptosis and enhancement of glycolysis. Up-regulation of SLC31A1, ENO1, and enhancement of SLC31A1-mediated ENO1 activity were critical for ARNTL2-triggered glycolysis and malignant growth in BLCA cells. ARNTL2 was positively correlated with SLC31A1 and ENO1 in BLCA patients. High expression of ARNTL2, SLC31A1 or ENO1 predicted the poor prognosis of BLCA patients. Depletion of SLC31A1 and inhibition of glycolysis completely blunted the growth ability of BLCA cells. CONCLUSION: In summary, ARNTL2 facilitates the progression of BLCA via activating ENO1-mediated glycolysis in a SLC31A1-independent and -dependent manner. Inhibiting SLC31A1 and glycolysis may be an aspirational approach for the treatment of BLCA patients with overexpression of ARNTL2.
Assuntos
Fatores de Transcrição ARNTL , Proliferação de Células , Proteínas de Ligação a DNA , Glicólise , Fosfopiruvato Hidratase , Proteínas Supressoras de Tumor , Neoplasias da Bexiga Urinária , Animais , Feminino , Humanos , Masculino , Camundongos , Apoptose , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Biomarcadores Tumorais , Carcinogênese/metabolismo , Carcinogênese/genética , Linhagem Celular Tumoral , Progressão da Doença , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfopiruvato Hidratase/metabolismo , Fosfopiruvato Hidratase/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/genética , Transportador de Cobre 1/genética , Transportador de Cobre 1/metabolismoRESUMO
Serum iron has long been thought to exhibit diurnal variation and is subsequently considered an unreliable biomarker of systemic iron status. Circadian regulation (endogenous ~24-h periodic oscillation of a biologic function) governs many critical physiologic processes. It is unknown whether serum iron levels are regulated by circadian machinery; likewise, the circadian nature of key players of iron homeostasis is unstudied. Here we show that serum iron, transferrin saturation (TSAT), hepatic transferrin receptor (TFR1) gene (Tfrc) expression, and erythropoietic activity exhibit circadian rhythms. Daily oscillations of serum iron, TSAT, hepatic Tfrc expression, and erythropoietic activity are maintained in mice housed in constant darkness, where oscillation reflects an endogenous circadian period. Oscillations of serum iron, TSAT, hepatic Tfrc, and erythropoietic activity were ablated when circadian machinery was disrupted in Bmal1 knockout mice. Interestingly, we find that circadian oscillations of erythropoietic activity and hepatic Tfrc expression are maintained in opposing phase, likely allowing for optimized usage and storage of serum iron whilst maintaining adequate serum levels and TSAT. This study provides the first confirmatory evidence that serum iron is circadian regulated, discerns circadian rhythms of TSAT, a widely used clinical marker of iron status, and uncovers liver-specific circadian regulation of TFR1, a major player in cellular iron uptake.
Assuntos
Ritmo Circadiano , Eritropoese , Ferro , Fígado , Camundongos Knockout , Receptores da Transferrina , Transferrina , Animais , Receptores da Transferrina/genética , Receptores da Transferrina/sangue , Ferro/metabolismo , Ferro/sangue , Fígado/metabolismo , Camundongos , Transferrina/metabolismo , Fatores de Transcrição ARNTL/genética , Masculino , Camundongos Endogâmicos C57BL , Regulação da Expressão GênicaRESUMO
Many types of neurons exhibit a daily rhythm of intrinsic excitability. Here, we present a protocol for assessing circadian regulation of dentate granule cell excitability using a mouse model for conditional knockout of the molecular clock protein BMAL1. We describe steps for obtaining healthy oblique horizontal slices that contain the hippocampus and measuring intrinsic excitability and synaptic potentials by combining whole-cell patch-clamp recordings and perforant-path electric stimulation. We then detail procedures for validating single-cell genetic deletion of Bmal1 by immunohistochemistry. For complete details on the use and execution of this protocol, please refer to Gonzalez et al.1.
Assuntos
Ritmo Circadiano , Giro Denteado , Animais , Camundongos , Giro Denteado/citologia , Giro Denteado/metabolismo , Giro Denteado/fisiologia , Ritmo Circadiano/fisiologia , Técnicas de Patch-Clamp/métodos , Neurônios/metabolismo , Neurônios/fisiologia , Neurônios/citologia , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Camundongos Knockout , MasculinoRESUMO
The transcriptional response to hypoxia is temporally regulated, yet the molecular underpinnings and physiological implications are unknown. We examined the roles of hepatic Bmal1 and Hif1α in the circadian response to hypoxia in mice. We found that the majority of the transcriptional response to hypoxia is dependent on either Bmal1 or Hif1α, through shared and distinct roles that are daytime determined. We further show that hypoxia-inducible factor (HIF)1α accumulation upon hypoxia is temporally regulated and Bmal1 dependent. Unexpectedly, mice lacking both hepatic Bmal1 and Hif1α are hypoxemic and exhibit increased mortality upon hypoxic exposure in a daytime-dependent manner. These mice display mild liver dysfunction with pulmonary vasodilation likely due to extracellular signaling regulated kinase (ERK) activation, endothelial nitric oxide synthase, and nitric oxide accumulation in lungs, suggestive of hepatopulmonary syndrome. Our findings indicate that hepatic BMAL1 and HIF1α are key time-dependent regulators of the hypoxic response and can provide molecular insights into the pathophysiology of hepatopulmonary syndrome.
Assuntos
Fatores de Transcrição ARNTL , Síndrome Hepatopulmonar , Subunidade alfa do Fator 1 Induzível por Hipóxia , Hipóxia , Fígado , Animais , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fígado/metabolismo , Camundongos , Hipóxia/metabolismo , Síndrome Hepatopulmonar/metabolismo , Camundongos Knockout , Camundongos Endogâmicos C57BL , Masculino , Ritmo Circadiano , Pulmão/metabolismoRESUMO
INTRODUCTION: This study aimed to investigate the relationship between obstructive sleep apnea (OSA), circadian rhythms, and individual sleep-wake preferences, as measured by chronotype, and to assess the association between circadian clock gene expression and subjective sleep-related variables. METHODS: A total of 184 individuals were recruited, underwent polysomnography (PSG), and completed questionnaires including a chronotype questionnaire (CQ), insomnia severity index (ISI), and Epworth sleepiness scale (ESS). Blood samples were collected in the evening before and morning after PSG. Gene expression analysis included BMAL1, CLOCK, PER1, CRY1, NPAS2, and NR1D1. RESULTS: In the OSA group, the subjective amplitude (AM score of CQ) positively correlated with all circadian clock genes in the morning (R ≥ 0.230 and p < 0.05 for each one), while the morningness-eveningness (ME score of CQ) was only associated with the evening BMAL1 level (R = 0.192; p = 0.044). In healthy controls, insomnia severity correlated with evening expression of BMAL1, PER1, and CRY1. CONCLUSIONS: The findings highlight the complex interplay between OSA, circadian rhythms, and sleep-related variables, suggesting potential determinants of morning chronotype in OSA and implicating disrupted circadian clock function in subjective feelings of energy throughout the day. Further research is warranted to elucidate underlying mechanisms and guide personalized management strategies.
Assuntos
Relógios Circadianos , Ritmo Circadiano , Apneia Obstrutiva do Sono , Distúrbios do Início e da Manutenção do Sono , Humanos , Masculino , Distúrbios do Início e da Manutenção do Sono/genética , Distúrbios do Início e da Manutenção do Sono/metabolismo , Feminino , Apneia Obstrutiva do Sono/genética , Apneia Obstrutiva do Sono/fisiopatologia , Apneia Obstrutiva do Sono/metabolismo , Pessoa de Meia-Idade , Relógios Circadianos/genética , Adulto , Ritmo Circadiano/genética , Polissonografia , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Regulação da Expressão Gênica , Sonolência , Inquéritos e Questionários , Cronotipo , CriptocromosRESUMO
Biological processes throughout the body are orchestrated in time through the regulation of local circadian clocks. The retina is among the most metabolically active tissues, with demands depending greatly on the light/dark cycle. Most cell types within the rodent retina are known to express the circadian clock; however, retinal clock expression in humans has not previously been localized. Moreover, the effect of local circadian clock dysfunction on retinal homeostasis is incompletely understood. The current study indicated an age-dependent decline in circadian clock gene and protein expression in the human retina. An animal model of targeted Bmal1 deficiency was used to identify the circadian clock of the retinal Müller glia as essential for neuronal survival, vascular integrity, and retinal function. These results suggest a potential role for the local retinal circadian clock within the Müller glia in age-related retinal disease and retinal degeneration.
Assuntos
Sobrevivência Celular , Relógios Circadianos , Células Ependimogliais , Homeostase , Retina , Relógios Circadianos/fisiologia , Relógios Circadianos/genética , Animais , Células Ependimogliais/metabolismo , Homeostase/fisiologia , Retina/metabolismo , Retina/patologia , Humanos , Sobrevivência Celular/fisiologia , Camundongos , Masculino , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Feminino , Pessoa de Meia-Idade , Adulto , Idoso , Degeneração Retiniana/patologia , Degeneração Retiniana/metabolismo , Neurônios/metabolismo , Neurônios/patologiaRESUMO
Depression is associated with dysregulated circadian rhythms, but the role of intrinsic clocks in mood-controlling brain regions remains poorly understood. We found increased circadian negative loop and decreased positive clock regulators expression in the medial prefrontal cortex (mPFC) of a mouse model of depression, and a subsequent clock countermodulation by the rapid antidepressant ketamine. Selective Bmal1KO in CaMK2a excitatory neurons revealed that the functional mPFC clock is an essential factor for the development of a depression-like phenotype and ketamine effects. Per2 silencing in mPFC produced antidepressant-like effects, while REV-ERB agonism enhanced the depression-like phenotype and suppressed ketamine action. Pharmacological potentiation of clock positive modulator ROR elicited antidepressant-like effects, upregulating plasticity protein Homer1a, synaptic AMPA receptors expression and plasticity-related slow wave activity specifically in the mPFC. Our data demonstrate a critical role for mPFC molecular clock in regulating depression-like behavior and the therapeutic potential of clock pharmacological manipulations influencing glutamatergic-dependent plasticity.