Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38562905

ABSTRACT

Epidemiological studies have shown that circadian rhythm disruption (CRD) is associated with the risk of breast cancer. However, the role of CRD in mammary gland morphology and aggressive basal mammary tumorigenesis and the molecular mechanisms underlying CRD and cancer risk remain unknown. To investigate the effect of CRD on aggressive tumorigenesis, a genetically engineered mouse model that recapitulates the human basal type of breast cancer was used for this study. The effect of CRD on mammary gland morphology was investigated using wild-type mice model. The impact of CRD on the tumor microenvironment was investigated using the tumors from LD12:12 and CRD mice via scRNA seq. ScRNA seq was substantiated by multiplexing immunostaining, flow cytometry, and realtime PCR. The effect of LILRB4 immunotherapy on CRD-induced tumorigenesis was also investigated. Here we identified the impact of CRD on basal tumorigenesis and mammary gland morphology and identified the role of LILRB4 on CRD-induced lung metastasis. We found that chronic CRD disrupted mouse mammary gland morphology and increased tumor burden, and lung metastasis and induced an immunosuppressive tumor microenvironment by enhancing LILRB4a expression. Moreover, CRD increased the M2-macrophage and regulatory T-cell populations but decreased the M1-macrophage populations. Furthermore, targeted immunotherapy against LILRB4 reduced CRD-induced immunosuppressive microenvironment and lung metastasis. These findings identify and implicate LILRB4a as a link between CRD and aggressive mammary tumorigenesis. This study also establishes the potential role of the targeted LILRB4a immunotherapy as an inhibitor of CRD-induced lung metastasis.

2.
J Hepatol ; 80(2): 282-292, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37890720

ABSTRACT

BACKGROUND & AIMS: Chronic circadian dysfunction increases the risk of non-alcoholic fatty liver disease (NAFLD)-related hepatocellular carcinoma (HCC), but the underlying mechanisms and direct relevance to human HCC have not been established. In this study, we aimed to determine whether chronic circadian dysregulation can drive NAFLD-related carcinogenesis from human hepatocytes and human HCC progression. METHODS: Chronic jet lag of mice with humanized livers induces spontaneous NAFLD-related HCCs from human hepatocytes. The clinical relevance of this model was analysed by biomarker, pathological/histological, genetic, RNA sequencing, metabolomic, and integrated bioinformatic analyses. RESULTS: Circadian dysfunction induces glucose intolerance, NAFLD-associated human HCCs, and human HCC metastasis independent of diet in a humanized mouse model. The deregulated transcriptomes in necrotic-inflammatory humanized livers and HCCs bear a striking resemblance to those of human non-alcoholic steatohepatitis (NASH), cirrhosis, and HCC. Stable circadian entrainment of hosts rhythmically paces NASH and HCC transcriptomes to decrease HCC incidence and prevent HCC metastasis. Circadian disruption directly reprogrammes NASH and HCC transcriptomes to drive a rapid progression from hepatocarcinogenesis to HCC metastasis. Human hepatocyte and tumour transcripts are clearly distinguishable from mouse transcripts in non-parenchymal cells and tumour stroma, and display dynamic changes in metabolism, inflammation, angiogenesis, and oncogenic signalling in NASH, progressing to hepatocyte malignant transformation and immunosuppressive tumour stroma in HCCs. Metabolomic analysis defines specific bile acids as prognostic biomarkers that change dynamically during hepatocarcinogenesis and in response to circadian disruption at all disease stages. CONCLUSION: Chronic circadian dysfunction is independently carcinogenic to human hepatocytes. Mice with humanized livers provide a powerful preclinical model for studying the impact of the necrotic-inflammatory liver environment and neuroendocrine circadian dysfunction on hepatocarcinogenesis and anti-HCC therapy. IMPACT AND IMPLICATIONS: Human epidemiological studies have linked chronic circadian dysfunction to increased hepatocellular carcinoma (HCC) risk, but direct evidence that circadian dysfunction is a human carcinogen has not been established. Here we show that circadian dysfunction induces non-alcoholic steatohepatitis (NASH)-related carcinogenesis from human hepatocytes in a murine humanized liver model, following the same molecular and pathologic pathways observed in human patients. The gene expression signatures of humanized HCC transcriptomes from circadian-disrupted mice closely match those of human HCC with the poorest prognostic outcomes, while those from stably circadian entrained mice match those from human HCC with the best prognostic outcomes. Our studies establish a new model for defining the mechanism of NASH-related HCC and highlight the importance of circadian biology in HCC prevention and treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Animals , Mice , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Liver/pathology , Disease Models, Animal , Carcinogenesis/metabolism , Carcinogens/metabolism
3.
iScience ; 25(12): 105546, 2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36465103

ABSTRACT

During evolution, humans are acclimatized to the stresses of natural radiation and circadian rhythmicity. Radiosensitivity of mammalian cells varies in the circadian period and adaptive radioprotection can be induced by pre-exposure to low-level radiation (LDR). It is unclear, however, if clock proteins participate in signaling LDR radioprotection. Herein, we demonstrate that radiosensitivity is increased in mice with the deficient Period 2 gene (Per2def) due to impaired DNA repair and mitochondrial function in progenitor bone marrow hematopoietic stem cells and monocytes. Per2 induction and radioprotection are also identified in LDR-treated Per2wt mouse cells and in human skin (HK18) and breast (MCF-10A) epithelial cells. LDR-boosted PER2 interacts with pGSK3ß(S9) which activates ß-catenin and the LEF/TCF mediated gene transcription including Per2 and genes involved in DNA repair and mitochondrial functions. This study demonstrates that PER2 plays an active role in LDR adaptive radioprotection via PER2/pGSK3ß/ß-catenin/Per2 loop, a potential target for protecting normal cells from radiation injury.

4.
Nat Commun ; 9(1): 4349, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341289

ABSTRACT

Hepatocyte nuclear factor 4 alpha (HNF4α) is a master regulator of liver-specific gene expression with potent tumor suppressor activity, yet many liver tumors express HNF4α. This study reveals that P1-HNF4α, the predominant isoform expressed in the adult liver, inhibits expression of tumor promoting genes in a circadian manner. In contrast, an additional isoform of HNF4α, driven by an alternative promoter (P2-HNF4α), is induced in HNF4α-positive human hepatocellular carcinoma (HCC). P2-HNF4α represses the circadian clock gene ARNTL (BMAL1), which is robustly expressed in healthy hepatocytes, and causes nuclear to cytoplasmic re-localization of P1-HNF4α. We reveal mechanisms underlying the incompatibility of BMAL1 and P2-HNF4α in HCC, and demonstrate that forced expression of BMAL1 in HNF4α-positive HCC prevents the growth of tumors in vivo. These data suggest that manipulation of the circadian clock in HNF4α-positive HCC could be a tractable strategy to inhibit tumor growth and progression in the liver.


Subject(s)
ARNTL Transcription Factors/metabolism , Carcinoma, Hepatocellular/metabolism , Hepatocyte Nuclear Factor 4/physiology , Liver Neoplasms/metabolism , ARNTL Transcription Factors/genetics , Active Transport, Cell Nucleus , Carcinoma, Hepatocellular/pathology , Circadian Clocks , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Liver Neoplasms/pathology , Protein Isoforms/physiology
5.
Cancer Cell ; 30(6): 909-924, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27889186

ABSTRACT

Chronic jet lag induces spontaneous hepatocellular carcinoma (HCC) in wild-type mice following a mechanism very similar to that observed in obese humans. The process initiates with non-alcoholic fatty liver disease (NAFLD) that progresses to steatohepatitis and fibrosis before HCC detection. This pathophysiological pathway is driven by jet-lag-induced genome-wide gene deregulation and global liver metabolic dysfunction, with nuclear receptor-controlled cholesterol/bile acid and xenobiotic metabolism among the top deregulated pathways. Ablation of farnesoid X receptor dramatically increases enterohepatic bile acid levels and jet-lag-induced HCC, while loss of constitutive androstane receptor (CAR), a well-known liver tumor promoter that mediates toxic bile acid signaling, inhibits NAFLD-induced hepatocarcinogenesis. Circadian disruption activates CAR by promoting cholestasis, peripheral clock disruption, and sympathetic dysfunction.


Subject(s)
Carcinoma, Hepatocellular/genetics , Circadian Clocks , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Non-alcoholic Fatty Liver Disease/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Constitutive Androstane Receptor , Disease Models, Animal , Gene Expression Regulation , Genetic Predisposition to Disease , Homeostasis , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Metabolome , Mice , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Cytoplasmic and Nuclear/genetics
6.
J Biol Rhythms ; 31(5): 443-60, 2016 10.
Article in English | MEDLINE | ID: mdl-27432117

ABSTRACT

Circadian rhythmicity is a fundamental process that synchronizes behavioral cues with metabolic homeostasis. Disruption of daily cycles due to jet lag or shift work results in severe physiological consequences including advanced aging, metabolic syndrome, and even cancer. Our understanding of the molecular clock, which is regulated by intricate positive feedforward and negative feedback loops, has expanded to include an important metabolic transcriptional coregulator, Steroid Receptor Coactivator-2 (SRC-2), that regulates both the central clock of the suprachiasmatic nucleus (SCN) and peripheral clocks including the liver. We hypothesized that an environmental uncoupling of the light-dark phases, termed chronic circadian disruption (CCD), would lead to pathology similar to the genetic circadian disruption observed with loss of SRC-2 We found that CCD and ablation of SRC-2 in mice led to a common comorbidity of metabolic syndrome also found in humans with circadian disruption, non-alcoholic fatty liver disease (NAFLD). The combination of SRC-2(-/-) and CCD results in a more robust phenotype that correlates with human non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) gene signatures. Either CCD or SRC-2 ablation produces an advanced aging phenotype leading to increased mortality consistent with other circadian mutant mouse models. Collectively, our studies demonstrate that SRC-2 provides an essential link between the behavioral activities influenced by light cues and the metabolic homeostasis maintained by the liver.


Subject(s)
Aging , Liver/pathology , Nuclear Receptor Coactivator 2/genetics , Nuclear Receptor Coactivator 2/physiology , Animals , Carcinoma, Hepatocellular/genetics , Circadian Clocks , Circadian Rhythm/physiology , Disease Models, Animal , Humans , Liver/metabolism , Liver Neoplasms/genetics , Mice , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/physiopathology , Nuclear Receptor Coactivator 2/deficiency , Period Circadian Proteins/genetics , Photoperiod , Suprachiasmatic Nucleus/physiology
7.
Mol Endocrinol ; 30(9): 988-95, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27427832

ABSTRACT

Circadian rhythm regulates multiple metabolic processes and in turn is readily entrained by feeding-fasting cycles. However, the molecular mechanisms by which the peripheral clock senses nutrition availability remain largely unknown. Bile acids are under circadian control and also increase postprandially, serving as regulators of the fed state in the liver. Here, we show that nuclear receptor Small Heterodimer Partner (SHP), a regulator of bile acid metabolism, impacts the endogenous peripheral clock by directly regulating Bmal1. Bmal1-dependent gene expression is altered in Shp knockout mice, and liver clock adaptation is delayed in Shp knockout mice upon restricted feeding. These results identify SHP as a potential mediator connecting nutrient signaling with the circadian clock.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Cells, Cultured , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/genetics , Signal Transduction/physiology
8.
Cell Metab ; 22(3): 448-59, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26166747

ABSTRACT

Circadian disruption is associated with obesity, implicating the central clock in body weight control. Our comprehensive screen of wild-type and three circadian mutant mouse models, with or without chronic jet lag, shows that distinct genetic and physiologic interventions differentially disrupt overall energy homeostasis and Leptin signaling. We found that BMAL1/CLOCK generates circadian rhythm of C/EBPα-mediated leptin transcription in adipose. Per and Cry mutant mice show similar disruption of peripheral clock and deregulation of leptin in fat, but opposite body weight and composition phenotypes that correlate with their distinct patterns of POMC neuron deregulation in the arcuate nucleus. Chronic jet lag is sufficient to disrupt the endogenous adipose clock and also induce central Leptin resistance in wild-type mice. Thus, coupling of the central and peripheral clocks controls Leptin endocrine feedback homeostasis. We propose that Leptin resistance, a hallmark of obesity in humans, plays a key role in circadian dysfunction-induced obesity and metabolic syndromes.


Subject(s)
Adipose Tissue/metabolism , Circadian Clocks , Leptin/metabolism , Obesity/metabolism , Animals , Base Sequence , Body Weight , CLOCK Proteins/metabolism , Circadian Rhythm , Energy Metabolism , Leptin/blood , Leptin/genetics , Melanocortins/metabolism , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/genetics , Signal Transduction , Transcriptional Activation
9.
Ann Med ; 46(4): 208-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24901356

ABSTRACT

Humans as diurnal beings are active during the day and rest at night. This daily oscillation of behavior and physiology is driven by an endogenous circadian clock not environmental cues. In modern societies, changes in lifestyle have led to a frequent disruption of the endogenous circadian homeostasis leading to increased risk of various diseases including cancer. The clock is operated by the feedback loops of circadian genes and controls daily physiology by coupling cell proliferation and metabolism, DNA damage repair, and apoptosis in peripheral tissues with physical activity, energy homeostasis, immune and neuroendocrine functions at the organismal level. Recent studies have revealed that defects in circadian genes due to targeted gene ablation in animal models or single nucleotide polymorphism, deletion, deregulation and/or epigenetic silencing in humans are closely associated with increased risk of cancer. In addition, disruption of circadian rhythm can disrupt the molecular clock in peripheral tissues in the absence of circadian gene mutations. Circadian disruption has recently been recognized as an independent cancer risk factor. Further study of the mechanism of clock-controlled tumor suppression will have a significant impact on human health by improving the efficiencies of cancer prevention and treatment.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/genetics , Neoplasms/genetics , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Genetic Variation , Homeostasis/physiology , Humans , Mutation , Neoplasms/pathology , Risk Factors
10.
Cell Rep ; 6(4): 633-45, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24529706

ABSTRACT

Synchrony of the mammalian circadian clock is achieved by complex transcriptional and translational feedback loops centered on the BMAL1:CLOCK heterodimer. Modulation of circadian feedback loops is essential for maintaining rhythmicity, yet the role of transcriptional coactivators in driving BMAL1:CLOCK transcriptional networks is largely unexplored. Here, we show diurnal hepatic steroid receptor coactivator 2 (SRC-2) recruitment to the genome that extensively overlaps with the BMAL1 cistrome during the light phase, targeting genes that enrich for circadian and metabolic processes. Notably, SRC-2 ablation impairs wheel-running behavior, alters circadian gene expression in several peripheral tissues, alters the rhythmicity of the hepatic metabolome, and deregulates the synchronization of cell-autonomous metabolites. We identify SRC-2 as a potent coregulator of BMAL1:CLOCK and find that SRC-2 targets itself with BMAL1:CLOCK in a feedforward loop. Collectively, our data suggest that SRC-2 is a transcriptional coactivator of the BMAL1:CLOCK oscillators and establish SRC-2 as a critical positive regulator of the mammalian circadian clock.


Subject(s)
Circadian Rhythm , Metabolome , Nuclear Receptor Coactivator 2/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Liver/metabolism , Male , Mice , Nuclear Receptor Coactivator 2/genetics , Organ Specificity , Transcriptome
11.
Prog Mol Biol Transl Sci ; 119: 221-82, 2013.
Article in English | MEDLINE | ID: mdl-23899600

ABSTRACT

Most aspects of mammalian function display circadian rhythms driven by an endogenous clock. The circadian clock is operated by genes and comprises a central clock in the brain that responds to environmental cues and controls subordinate clocks in peripheral tissues via circadian output pathways. The central and peripheral clocks coordinately generate rhythmic gene expression in a tissue-specific manner in vivo to couple diverse physiological and behavioral processes to periodic changes in the environment. However, with the industrialization of the world, activities that disrupt endogenous homeostasis with external circadian cues have increased. This change in lifestyle has been linked to an increased risk of diseases in all aspects of human health, including cancer. Studies in humans and animal models have revealed that cancer development in vivo is closely associated with the loss of circadian homeostasis in energy balance, immune function, and aging, which are supported by cellular functions important for tumor suppression including cell proliferation, senescence, metabolism, and DNA damage response. The clock controls these cellular functions both locally in cells of peripheral tissues and at the organismal level via extracellular signaling. Thus, the hierarchical mammalian circadian clock provides a unique system to study carcinogenesis as a deregulated physiological process in vivo. The asynchrony between host and malignant tissues in cell proliferation and metabolism also provides new and exciting options for novel anticancer therapies.


Subject(s)
Circadian Clocks , Neoplasms/physiopathology , Humans , Neoplasms/therapy
12.
Islets ; 3(6): 381-8, 2011.
Article in English | MEDLINE | ID: mdl-22045262

ABSTRACT

The circadian clock has been shown to regulate metabolic homeostasis. Mice with a deletion of Bmal1, a key component of the core molecular clock, develop hyperglycemia and hypoinsulinemia, suggesting ß-cell dysfunction. However, the underlying mechanisms are not fully known. In this study, we investigated the mechanisms underlying the regulation of ß-cell function by Bmal1. We studied ß-cell function in global Bmal1-/- mice, in vivo and in isolated islets ex vivo, as well as in rat insulinoma cell lines with shRNA-mediated Bmal1 knockdown. Global Bmal1-/- mice develop diabetes secondary to a significant impairment in glucose-stimulated insulin secretion (GSIS). There is a blunting of GSIS in both isolated Bmal1-/- islets and in Bmal1 knockdown cells, as compared to controls, suggesting that this is secondary to a loss of cell-autonomous effect of Bmal1. In contrast to previous studies, in these Bmal1-/- mice on a C57Bl/6 background, the loss of stimulated insulin secretion, interestingly, is with glucose but not to other depolarizing secretagogues, suggesting that events downstream of membrane depolarization are largely normal in Bmal1-/- islets. This defect in GSIS occurs as a result increased mitochondrial uncoupling with consequent impairment of glucose-induced mitochondrial potential generation and ATP synthesis, due to an upregulation of Ucp2. Inhibition of Ucp2, in isolated islets, leads to a rescue of the glucose-induced ATP production and insulin secretion in Bmal1-/- islets. Thus, Bmal1 regulates mitochondrial energy metabolism to maintain normal GSIS and its disruption leads to diabetes due to a loss of GSIS.


Subject(s)
ARNTL Transcription Factors/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , Cell Line, Tumor , Immunohistochemistry , Insulin Secretion , Ion Channels/metabolism , Islets of Langerhans/cytology , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Rats , Uncoupling Protein 2 , Up-Regulation/physiology
13.
PLoS One ; 5(6): e10995, 2010 Jun 07.
Article in English | MEDLINE | ID: mdl-20539819

ABSTRACT

BACKGROUND: Cell proliferation in all rapidly renewing mammalian tissues follows a circadian rhythm that is often disrupted in advanced-stage tumors. Epidemiologic studies have revealed a clear link between disruption of circadian rhythms and cancer development in humans. Mice lacking the circadian genes Period1 and 2 (Per) or Cryptochrome1 and 2 (Cry) are deficient in cell cycle regulation and Per2 mutant mice are cancer-prone. However, it remains unclear how circadian rhythm in cell proliferation is generated in vivo and why disruption of circadian rhythm may lead to tumorigenesis. METHODOLOGY/PRINCIPAL FINDINGS: Mice lacking Per1 and 2, Cry1 and 2, or one copy of Bmal1, all show increased spontaneous and radiation-induced tumor development. The neoplastic growth of Per-mutant somatic cells is not controlled cell-autonomously but is dependent upon extracellular mitogenic signals. Among the circadian output pathways, the rhythmic sympathetic signaling plays a key role in the central-peripheral timing mechanism that simultaneously activates the cell cycle clock via AP1-controlled Myc induction and p53 via peripheral clock-controlled ATM activation. Jet-lag promptly desynchronizes the central clock-SNS-peripheral clock axis, abolishes the peripheral clock-dependent ATM activation, and activates myc oncogenic potential, leading to tumor development in the same organ systems in wild-type and circadian gene-mutant mice. CONCLUSIONS/SIGNIFICANCE: Tumor suppression in vivo is a clock-controlled physiological function. The central circadian clock paces extracellular mitogenic signals that drive peripheral clock-controlled expression of key cell cycle and tumor suppressor genes to generate a circadian rhythm in cell proliferation. Frequent disruption of circadian rhythm is an important tumor promoting factor.


Subject(s)
Circadian Rhythm/genetics , Homeostasis , Neoplasms, Experimental/pathology , Signal Transduction , Animals , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/physiopathology , Sympathetic Nervous System/metabolism
14.
PLoS One ; 4(8): e6843, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19718444

ABSTRACT

BACKGROUND: Bile acids are potentially toxic compounds and their levels of hepatic production, uptake and export are tightly regulated by many inputs, including circadian rhythm. We tested the impact of disrupting the peripheral circadian clock on integral steps of bile acid homeostasis. METHODOLOGY/PRINCIPAL FINDINGS: Both restricted feeding, which phase shifts peripheral clocks, and genetic ablation in Per1(-/-)/Per2(-/-) (PERDKO) mice disrupted normal bile acid control and resulted in hepatic cholestasis. Restricted feeding caused a dramatic, transient elevation in hepatic bile acid levels that was associated with activation of the xenobiotic receptors CAR and PXR and elevated serum aspartate aminotransferase (AST), indicative of liver damage. In the PERDKO mice, serum bile acid levels were elevated and the circadian expression of key bile acid synthesis and transport genes, including Cyp7A1 and NTCP, was lost. This was associated with blunted expression of a primary clock output, the transcription factor DBP, which transactivates the promoters of both genes. CONCLUSIONS/SIGNIFICANCE: We conclude that disruption of the circadian clock results in dysregulation of bile acid homeostasis that mimics cholestatic disease.


Subject(s)
Bile Acids and Salts/physiology , Circadian Rhythm , Homeostasis , Animals , Aspartate Aminotransferases/genetics , Liver/metabolism , Mice , Mice, Knockout , Period Circadian Proteins/genetics , Period Circadian Proteins/physiology
15.
Prog Brain Res ; 153: 177-88, 2006.
Article in English | MEDLINE | ID: mdl-16876575

ABSTRACT

Mice with circadian gene Period and Cryptochrome mutations develop high bone mass early in life. Such a phenotype is accompanied by an increase in osteoblast numbers in mutant bone and cannot be corrected by leptin intracerebroventricular infusion. Thus, the molecular clock plays a key role in leptin-mediated sympathetic regulation of bone formation. Indeed, we found that leptin-dependent sympathetic signaling induces the expression of AP1 and circadian genes in bone and in osteoblasts with similar kinetics, and these two pathways play opposite roles in controlling c-myc expression. Mutations in the Period 1 and 2 genes result in uncontrolled c-myc signaling, overexpression of G1 cyclins, and increased osteoblast proliferation and bone-formation parameters. These results indicate that the role of leptin-dependent sympathetic signaling in bone formation is achieved through regulating two antagonistic pathways in osteoblasts.


Subject(s)
Circadian Rhythm/physiology , Leptin/physiology , Osteogenesis/physiology , Animals , Cell Cycle Proteins , Circadian Rhythm/genetics , Humans , Mice , Mice, Mutant Strains , Models, Biological , Nuclear Proteins , Osteogenesis/genetics , Period Circadian Proteins , Signal Transduction/physiology , Transcription Factors
16.
Cell ; 122(5): 803-15, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16143109

ABSTRACT

The hormone leptin is a regulator of bone remodeling, a homeostatic function maintaining bone mass constant. Mice lacking molecular-clock components (Per and Cry), or lacking Per genes in osteoblasts, display high bone mass, suggesting that bone remodeling may also be subject to circadian regulation. Moreover, Per-deficient mice experience a paradoxical increase in bone mass following leptin intracerebroventricular infusion. Thus, clock genes may mediate the leptin-dependent sympathetic regulation of bone formation. We show that expression of clock genes in osteoblasts is regulated by the sympathetic nervous system and leptin. Clock genes mediate the antiproliferative function of sympathetic signaling by inhibiting G1 cyclin expression. Partially antagonizing this inhibitory loop, leptin also upregulates AP-1 gene expression, which promotes cyclin D1 expression, osteoblast proliferation, and bone formation. Thus, leptin determines the extent of bone formation by modulating, via sympathetic signaling, osteoblast proliferation through two antagonistic pathways, one of which involves the molecular clock.


Subject(s)
Bone Remodeling/drug effects , Circadian Rhythm/physiology , Leptin/pharmacology , Animals , Bone Density/drug effects , Bone Density/physiology , Bone Remodeling/physiology , Cell Cycle Proteins , Cell Proliferation/drug effects , Circadian Rhythm/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Leptin/metabolism , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/physiology , Period Circadian Proteins , Phenotype , Signal Transduction/drug effects , Signal Transduction/physiology , Stromal Cells/drug effects , Stromal Cells/physiology , Sympathetic Nervous System/metabolism , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology
17.
Nat Rev Cancer ; 3(5): 350-61, 2003 May.
Article in English | MEDLINE | ID: mdl-12724733

ABSTRACT

The circadian rhythms are daily oscillations in various biological processes that are regulated by an endogenous clock. Disruption of these rhythms has been associated with cancer in humans. One of the cellular processes that is regulated by circadian rhythm is cell proliferation, which often shows asynchrony between normal and malignant tissues. This asynchrony highlights the importance of the circadian clock in tumour suppression in vivo and is one of the theoretical foundations for cancer chronotherapy. Investigation of the mechanisms by which the circadian clock controls cell proliferation and other cellular functions might lead to new therapeutic targets.


Subject(s)
Biological Clocks , Circadian Rhythm , Neoplasms/prevention & control , Animals , Eye Proteins/physiology , Humans , Neoplasms/etiology , Neoplasms/therapy , Proto-Oncogene Proteins c-myc/physiology , Suprachiasmatic Nucleus/physiology , Tumor Suppressor Protein p53/physiology
18.
Cell ; 111(1): 41-50, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12372299

ABSTRACT

The Period2 gene plays a key role in controlling circadian rhythm in mice. We report here that mice deficient in the mPer2 gene are cancer prone. After gamma radiation, these mice show a marked increase in tumor development and reduced apoptosis in thymocytes. The core circadian genes are induced by gamma radiation in wild-type mice but not in mPer2 mutant mice. Temporal expression of genes involved in cell cycle regulation and tumor suppression, such as Cyclin D1, Cyclin A, Mdm-2, and Gadd45alpha, is deregulated in mPer2 mutant mice. In particular, the transcription of c-myc is controlled directly by circadian regulators and is deregulated in the mPer2 mutant. Our studies suggest that the mPer2 gene functions in tumor suppression by regulating DNA damage-responsive pathways.


Subject(s)
Cell Cycle Proteins , DNA Damage , Neoplasms, Experimental/genetics , Nuclear Proteins/genetics , Nuclear Proteins/physiology , ARNTL Transcription Factors , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Division , Cyclin A/genetics , Cyclin D1/genetics , Dimerization , Flow Cytometry , Gamma Rays , Genetic Predisposition to Disease , Mice , Mice, Inbred C57BL , Mutation , Nerve Tissue Proteins/metabolism , Period Circadian Proteins , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Proto-Oncogene Proteins c-myc/metabolism , Thymus Gland/cytology , Time Factors , Transcription Factors/metabolism , Transfection , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...