Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
N Engl J Med ; 387(13): 1196-1206, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36170501

ABSTRACT

BACKGROUND: B-cell maturation antigen (BCMA)-directed chimeric antigen receptor (CAR) T-cell therapies have generated responses in patients with advanced myeloma, but relapses are common. G protein-coupled receptor, class C, group 5, member D (GPRC5D) has been identified as an immunotherapeutic target in multiple myeloma. Preclinical studies have shown the efficacy of GPRC5D-targeted CAR T cells, including activity in a BCMA antigen escape model. METHODS: In this phase 1 dose-escalation study, we administered a GPRC5D-targeted CAR T-cell therapy (MCARH109) at four dose levels to patients with heavily pretreated multiple myeloma, including patients with relapse after BCMA CAR T-cell therapy. RESULTS: A total of 17 patients were enrolled and received MCARH109 therapy. The maximum tolerated dose was identified at 150×106 CAR T cells. At the 450×106 CAR T-cell dose, 1 patient had grade 4 cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome (ICANS), and 2 patients had a grade 3 cerebellar disorder of unclear cause. No cerebellar disorder, ICANS of any grade, or cytokine release syndrome of grade 3 or higher occurred in the 12 patients who received doses of 25×106 to 150×106 cells. A response was reported in 71% of the patients in the entire cohort and in 58% of those who received doses of 25×106 to 150×106 cells. The patients who had a response included those who had received previous BCMA therapies; responses were observed in 7 of 10 such patients in the entire cohort and in 3 of 6 such patients who received 25×106 to 150×106 cells. CONCLUSIONS: The results of this study of a GPRC5D-targeted CAR T-cell therapy (MCARH109) confirm that GPRC5D is an active immunotherapeutic target in multiple myeloma. (Funded by Juno Therapeutics/Bristol Myers Squibb; ClinicalTrials.gov number, NCT04555551.).


Subject(s)
Immunotherapy, Adoptive , Multiple Myeloma , Receptors, Chimeric Antigen , Receptors, G-Protein-Coupled , B-Cell Maturation Antigen/therapeutic use , Cytokine Release Syndrome/etiology , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/etiology , Receptors, Chimeric Antigen/therapeutic use , Receptors, G-Protein-Coupled/therapeutic use , T-Lymphocytes
2.
Mol Ther Methods Clin Dev ; 22: 377-387, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34514029

ABSTRACT

With the US Food and Drug Administration (FDA) approval of four CD19- and one BCMA-targeted chimeric antigen receptor (CAR) therapy for B cell malignancies, CAR T cell therapy has finally reached the status of a medicinal product. The successful manufacturing of autologous CAR T cell products is a key requirement for this promising treatment modality. By analyzing the composition of 214 apheresis products from 210 subjects across eight disease indications, we found that high CD14+ cell content poses a challenge for manufacturing CAR T cells, especially in patients with non-Hodgkin's lymphoma and multiple myeloma caused by the non-specific phagocytosis of the magnetic beads used to activate CD3+ T cells. We demonstrated that monocyte depletion via rapid plastic surface adhesion significantly reduces the CD14+ monocyte content in the apheresis products and simultaneously boosts the CD3+ content. We established a 40% CD14+ threshold for the stratification of apheresis products across nine clinical trials and demonstrated the effectiveness of this procedure by comparing manufacturing runs in two phase 1 clinical trials. Our study suggests that CD14+ content should be monitored in apheresis products, and that the manufacturing of CAR T cells should incorporate a step that lessens the CD14+ cell content in apheresis products containing more than 40% to maximize the production success.

3.
Cancer Discov ; 11(11): 2748-2763, 2021 11.
Article in English | MEDLINE | ID: mdl-34266984

ABSTRACT

Malignant pleural diseases, comprising metastatic lung and breast cancers and malignant pleural mesothelioma (MPM), are aggressive solid tumors with poor therapeutic response. We developed and conducted a first-in-human, phase I study of regionally delivered, autologous, mesothelin-targeted chimeric antigen receptor (CAR) T-cell therapy. Intrapleural administration of 0.3M to 60M CAR T cells/kg in 27 patients (25 with MPM) was safe and well tolerated. CAR T cells were detected in peripheral blood for >100 days in 39% of patients. Following our demonstration that PD-1 blockade enhances CAR T-cell function in mice, 18 patients with MPM also received pembrolizumab safely. Among those patients, median overall survival from CAR T-cell infusion was 23.9 months (1-year overall survival, 83%). Stable disease was sustained for ≥6 months in 8 patients; 2 exhibited complete metabolic response on PET scan. Combination immunotherapy with CAR T cells and PD-1 blockade agents should be further evaluated in patients with solid tumors. SIGNIFICANCE: Regional delivery of mesothelin-targeted CAR T-cell therapy followed by pembrolizumab administration is feasible, safe, and demonstrates evidence of antitumor efficacy in patients with malignant pleural diseases. Our data support the investigation of combination immunotherapy with CAR T cells and PD-1 blockade agents in solid tumors.See related commentary by Aldea et al., p. 2674.This article is highlighted in the In This Issue feature, p. 2659.


Subject(s)
Mesothelioma , Pleural Diseases , Antibodies, Monoclonal, Humanized , Humans , Immunotherapy, Adoptive , Mesothelin , Mesothelioma/drug therapy
4.
Endocrinology ; 155(2): 485-501, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24248466

ABSTRACT

The aging process causes an increase in percent body fat, but the mechanism remains unclear. In the present study we examined the impact of aging on brown adipose tissue (BAT) thermogenic activity as potential cause for the increase in adiposity. We show that aging is associated with interscapular BAT morphologic abnormalities and thermogenic dysfunction. In vitro experiments revealed that brown adipocyte differentiation is defective in aged mice. Interscapular brown tissue in aged mice is progressively populated by adipocytes bearing white morphologic characteristics. Aged mice fail to mobilize intracellular fuel reserves from brown adipocytes and exhibit deficiency in homeothermy. Our results suggest a role for orexin (OX) signaling in the regulation of thermogenesis during aging. Brown fat dysfunction and age-related assimilation of fat mass were accelerated in mice in which OX-producing neurons were ablated. Conversely, OX injections in old mice increased multilocular morphology, increased core body temperature, improved cold tolerance, and reduced adiposity. These results argue that BAT can be targeted for interventions to reverse age-associated increase in fat mass.


Subject(s)
Adipocytes/drug effects , Adipose Tissue, Brown/drug effects , Aging/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Thermogenesis/drug effects , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adiposity/drug effects , Adiposity/physiology , Aging/metabolism , Animals , Energy Metabolism/drug effects , Energy Metabolism/physiology , Male , Mice , Orexins , Thermogenesis/physiology
5.
Ann N Y Acad Sci ; 1302: 11-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24111875

ABSTRACT

Nutrition plays a dominant role in human adaptation. Biological traits conferring these adaptations are of considerable significance. Within an obesogenic environment, there is considerable variation among individuals in their susceptibility to weight gain. Some individuals rapidly gain weight, whereas others remain lean without any conscious effort, suggesting that obesity pathogenesis may not be centered on just the primal feeding behavior. The ability of certain individuals to subconsciously resist obesity reveals adaptive calorie-burning mechanisms that may promote fitness. Here, we review a fat-burning mechanism that is turned on by the brain hormone orexin during high-caloric food consumption. Remarkably, the same hormone also induces feeding, and its levels correlate with lean body mass in both rodents and humans. Intriguingly, loss of orexin prevents thermogenic energy expenditure while inducing obesity in the face of hypophagia. Thus, orexin is a unique neuropeptide that promotes both feeding and energy expenditure, conferring resistance to weight gain. Mechanisms that safely augment orexin signaling may have potential in antiobesity therapeutics.


Subject(s)
Adipose Tissue, Brown/physiology , Feeding Behavior/physiology , Obesity/etiology , Thermogenesis , Adipose Tissue, Brown/metabolism , Adiposity/genetics , Animals , Energy Metabolism/genetics , Energy Metabolism/physiology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Narcolepsy/etiology , Narcolepsy/genetics , Narcolepsy/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Neuropeptides/physiology , Obesity/genetics , Obesity/metabolism , Orexins , Sleep/genetics , Sleep/physiology , Thermogenesis/drug effects , Wakefulness/genetics , Wakefulness/physiology
6.
Endocrinology ; 154(11): 3990-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002035

ABSTRACT

Orexins are a pair of hypothalamic neuropeptides that were discovered in the late 1990s and named initially for their ability to promote feeding. Subsequent studies have revealed the importance of orexins to a variety of physiological functions, including brown fat thermogenesis, sleep/wake cycles, physical activity, and cognition. We aim to elucidate the various roles of orexins and discuss how these multiple functions are interlinked. We explain that although the unique dual roles of orexins in increasing feeding while concomitantly elevating energy expenditure appear counterproductive, they are necessary for physiological scenarios during which simultaneous stimulation of energy expenditure and feeding occur, namely diet-induced thermogenesis and arousal from hibernation. The position of orexins at the interface between sleep/wake cycles, energy homeostasis, and environmental factors has important implications in the treatment of obesity.


Subject(s)
Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Adipose Tissue, Brown/physiology , Arousal/physiology , Eating , Humans , Orexins , Sleep/physiology
7.
Adipocyte ; 1(1): 58-63, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-23700511

ABSTRACT

Orexin A (OX) is a small excitatory neuropeptide hormone that stimulates feeding, wakefulness and energy expenditure via a pair of G-coupled protein receptors, namely orexin receptor-1 (OXR1) and orexin receptor-2 (OXR2). OX-deficient mice are sensitive to obesity despite being hypophagic. The obesogenic effect of OX-deletion is due to brown adipose tissue (BAT) dysfunction, a defect that originates during fetal growth. Brown preadipocytes in OX-null mice display undifferentiated histological appearance and fail to support both diet- and cold-induced thermogenesis. We show that the OXR1-null mice phenocopies the differentiation defect observed in the ligand-null mice indicating that OXR1 relays OX's differentiation and thermogenic function. Consistent with this, OX fails to induce differentiation in cultured OXR1-null preadipocytes. Thus, OX signaling via OXR1 constitutes an important thermoregulatory mechanism that defends against cold and obesity.

8.
Cell Metab ; 14(4): 478-90, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21982708

ABSTRACT

Orexin (OX) neuropeptides stimulate feeding and arousal. Deficiency of orexin is implicated in narcolepsy, a disease associated with obesity, paradoxically in the face of reduced food intake. Here, we show that obesity in orexin-null mice is associated with impaired brown adipose tissue (BAT) thermogenesis. Failure of thermogenesis in OX-null mice is due to inability of brown preadipocytes to differentiate. The differentiation defect in OX-null neonates is circumvented by OX injections to OX-null dams. In vitro, OX, triggers the full differentiation program in mesenchymal progenitor stem cells, embryonic fibroblasts and brown preadipocytes via p38 mitogen activated protein (MAP) kinase and bone morphogenetic protein receptor-1a (BMPR1A)-dependent Smad1/5 signaling. Our study suggests that obesity associated with OX depletion is linked to brown-fat hypoactivity, which leads to dampening of energy expenditure. Thus, orexin plays an integral role in adaptive thermogenesis and body weight regulation via effects on BAT differentiation and function.


Subject(s)
Adipose Tissue, Brown/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Adipose Tissue, Brown/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Differentiation , Cell Line , Energy Metabolism/physiology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Obesity/etiology , Orexins , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Thermogenesis/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Circ Res ; 108(4): 437-45, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21233454

ABSTRACT

RATIONALE: Despite overwhelming evidence of the importance of circadian rhythms in cardiovascular health and disease, little is known regarding the circadian regulation of intracellular signaling pathways controlling cardiac function and remodeling. OBJECTIVE: To assess circadian changes in processes dependent on the protein phosphatase calcineurin, relative to changes in phosphorylation of cardiac proteins, in normal, hypertrophic, and failing hearts. METHODS AND RESULTS: We found evidence of large circadian oscillations in calcineurin-dependent activities in the left ventricle of healthy C57BL/6 mice. Calcineurin-dependent transcript levels and nuclear occupancy of the NFAT (nuclear factor of activated T cells) regularly fluctuated as much as 20-fold over the course of a day, peaking in the morning when mice enter a period of rest. Phosphorylation of the protein phosphatase 1 inhibitor 1 (I-1), a direct calcineurin substrate, and phospholamban, an indirect target, oscillated directly out of phase with calcineurin-dependent signaling. Using a surgical model of cardiac pressure overload, we found that although calcineurin-dependent activities were markedly elevated, the circadian pattern of activation was maintained, whereas, oscillations in phospholamban and I-1 phosphorylation were lost. Changes in the expression of fetal gene markers of heart failure did not mirror the rhythm in calcineurin/NFAT activation, suggesting that these may not be direct transcriptional target genes. Cardiac function in mice subjected to pressure overload was significantly lower in the morning than in the evening when assessed by echocardiography. CONCLUSIONS: Normal, opposing circadian oscillations in calcineurin-dependent activities and phosphorylation of proteins that regulate contractility are disrupted in heart failure.


Subject(s)
Calcineurin/physiology , Circadian Rhythm/physiology , Heart Failure/metabolism , Hemodynamics/physiology , Proteins/metabolism , Signal Transduction/physiology , Stress, Physiological/physiology , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Heart Failure/physiopathology , Heart Ventricles/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/metabolism , NFATC Transcription Factors/metabolism , Phosphorylation/physiology , Protein Phosphatase 1/metabolism
10.
J Neurosci ; 29(24): 7820-32, 2009 Jun 17.
Article in English | MEDLINE | ID: mdl-19535594

ABSTRACT

Although it is a widely studied psychiatric syndrome, major depressive disorder remains a poorly understood illness, especially with regard to the disconnect between treatment initiation and the delayed onset of clinical improvement. We have recently validated chronic social defeat stress in mice as a model in which a depression-like phenotype is reversed by chronic, but not acute, antidepressant administration. Here, we use chromatin immunoprecipitation (ChIP)-chip assays--ChIP followed by genome wide promoter array analyses--to study the effects of chronic defeat stress on chromatin regulation in the mouse nucleus accumbens (NAc), a key brain reward region implicated in depression. Our results demonstrate that chronic defeat stress causes widespread and long-lasting changes in gene regulation, including alterations in repressive histone methylation and in phospho-CREB (cAMP response element-binding protein) binding, in the NAc. We then show similarities and differences in this regulation to that observed in another mouse model of depression, prolonged adult social isolation. In the social defeat model, we observed further that many of the stress-induced changes in gene expression are reversed by chronic imipramine treatment, and that resilient mice-those resistant to the deleterious effects of defeat stress-show patterns of chromatin regulation in the NAc that overlap dramatically with those seen with imipramine treatment. These findings provide new insight into the molecular basis of depression-like symptoms and the mechanisms by which antidepressants exert their delayed clinical efficacy. They also raise the novel idea that certain individuals resistant to stress may naturally mount antidepressant-like adaptations in response to chronic stress.


Subject(s)
Antidepressive Agents, Tricyclic/therapeutic use , Chromatin/drug effects , Depression/drug therapy , Depression/pathology , Imipramine/therapeutic use , Nucleus Accumbens/ultrastructure , Animals , Behavior, Animal , CREB-Binding Protein/metabolism , Chromatin Immunoprecipitation/methods , Disease Models, Animal , Dominance-Subordination , Gene Expression/drug effects , Gene Expression/physiology , Genome-Wide Association Study/methods , Histones/metabolism , Male , Methylation/drug effects , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Social Isolation
11.
Neuron ; 62(3): 335-48, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19447090

ABSTRACT

Changes in gene expression contribute to the long-lasting regulation of the brain's reward circuitry seen in drug addiction; however, the specific genes regulated and the transcriptional mechanisms underlying such regulation remain poorly understood. Here, we used chromatin immunoprecipitation coupled with promoter microarray analysis to characterize genome-wide chromatin changes in the mouse nucleus accumbens, a crucial brain reward region, after repeated cocaine administration. Our findings reveal several interesting principles of gene regulation by cocaine and of the role of DeltaFosB and CREB, two prominent cocaine-induced transcription factors, in this brain region. The findings also provide comprehensive insight into the molecular pathways regulated by cocaine-including a new role for sirtuins (Sirt1 and Sirt2)-which are induced in the nucleus accumbens by cocaine and, in turn, dramatically enhance the behavioral effects of the drug.


Subject(s)
Chromatin/drug effects , Cocaine/pharmacology , Gene Expression Regulation/drug effects , Nucleus Accumbens/drug effects , Sirtuins/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Dopamine Uptake Inhibitors/pharmacology , Feedback, Physiological/drug effects , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Reinforcement, Psychology , Signal Transduction/drug effects , Sirtuins/drug effects
12.
J Biol Chem ; 284(16): 10409-21, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19203999

ABSTRACT

Cardiac hypertrophy develops in response to a variety of cardiovascular stresses and results in activation of numerous signaling cascades and proteins. In the present study, we demonstrate that cytoglobin is a stress-responsive hemoprotein in the hypoxia-induced hypertrophic myocardium and it is transcriptionally regulated by calcineurin-dependent transcription factors. The cytoglobin transcript level is abundantly expressed in the adult heart and in response to hypoxia cytoglobin expression is markedly up-regulated within the hypoxia-induced hypertrophic heart. To define the molecular mechanism resulting in the induction of cytoglobin, we undertook a transcriptional analysis of the 5' upstream regulatory region of the cytoglobin gene. Evolutionarily conserved binding elements for transcription factors HIF-1, AP-1, and NFAT are located within the upstream region of the cytoglobin gene. Transcriptional assays demonstrated that calcineurin activity modulates cytoglobin transcription. Increased calcineurin activity enhances the ability of NFAT and AP-1 to bind to the putative cytoglobin promoter, especially under hypoxic conditions. In addition, inhibition of calcineurin, NFAT, and/or AP-1 activities decreases endogenous cytoglobin transcript and protein levels. Thus, the regulation of cytoglobin transcription by calcineurin-dependent transcription factors suggests that cytoglobin may have a functional role in calcium-dependent events accompanying cardiac remodeling.


Subject(s)
Calcineurin/metabolism , Globins/metabolism , Hypoxia , Myocytes, Cardiac/physiology , Transcriptional Activation , Animals , Base Sequence , Calcineurin/genetics , Cell Line , Cytoglobin , Globins/genetics , Humans , Hypertrophy, Left Ventricular/metabolism , Hypoxia/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Myocytes, Cardiac/cytology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Response Elements/genetics , Sequence Alignment
13.
Mol Biosyst ; 4(11): 1116-25, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18931787

ABSTRACT

Recent analysis of a Gal4 mutant (Gap71) carrying three point mutations (S22D, K23Q and K25F) in its DNA-binding domain (DBD), has demonstrated that it cannot occupy GAL promoters efficiently in cells and that it is not mono-ubiquitylated, suggesting a functional link between this modification and stable DNA binding in cells. The mechanistic underpinning of this phenotype is that this protein is hypersensitive to a newly discovered activity of the proteasomal ATPases--their ability to actively dissociate transcription factor-DNA complexes after direct interaction with the activation domain. In this paper, we examine the roles of each of the three point mutations contained in Gap71 individually. These experiments have revealed that serine 22 is a site of phosphorylation in the Gal4 DBD and that lysine 23 is essential for S22 phosphorylation, possibly acting as part of the kinase recognition site. Mutation of either residue blocks Gal4 DBD phosphorylation, its subsequent ubiquitylation and compromises the ability of the activator to bind promoter DNA in vivo. These data represent the first report of an essential phosphorylation event that is critical for the activity of this paradigmatic transcription factor.


Subject(s)
DNA/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Binding Sites , DNA-Binding Proteins , HeLa Cells , Humans , Lysine/genetics , Lysine/metabolism , Models, Molecular , Phosphorylation , Point Mutation , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Serine/genetics , Serine/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Ubiquitination
14.
Neuron ; 59(4): 621-33, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18760698

ABSTRACT

Repeated exposure to cocaine causes sensitized behavioral responses and increased dendritic spines on medium spiny neurons of the nucleus accumbens (NAc). We find that cocaine regulates myocyte enhancer factor 2 (MEF2) transcription factors to control these two processes in vivo. Cocaine suppresses striatal MEF2 activity in part through a mechanism involving cAMP, the regulator of calmodulin signaling (RCS), and calcineurin. We show that reducing MEF2 activity in the NAc in vivo is required for the cocaine-induced increases in dendritic spine density. Surprisingly, we find that increasing MEF2 activity in the NAc, which blocks the cocaine-induced increase in dendritic spine density, enhances sensitized behavioral responses to cocaine. Together, our findings implicate MEF2 as a key regulator of structural synapse plasticity and sensitized responses to cocaine and suggest that reducing MEF2 activity (and increasing spine density) in NAc may be a compensatory mechanism to limit long-lasting maladaptive behavioral responses to cocaine.


Subject(s)
Cocaine/pharmacology , Dendritic Spines/drug effects , Dopamine Uptake Inhibitors/pharmacology , Myogenic Regulatory Factors/metabolism , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Adaptation, Physiological/drug effects , Animals , Behavior, Animal/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclin-Dependent Kinases/drug effects , Down-Regulation , Drug Administration Schedule , Gene Expression Profiling , MEF2 Transcription Factors , Male , Mice , Mice, Inbred C57BL , Neostriatum/cytology , Neostriatum/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Synapses/drug effects
15.
Genes Dev ; 21(22): 2995-3005, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006690

ABSTRACT

Orexin A and Orexin B (also known as hypocretins) are neuropeptides that bind two related G-coupled protein receptors (OXR1 and OXR2) and thus induce wakefulness, food consumption, and locomotion. Conversely, deletion of the orexin gene in mice produces a condition similar to canine and human narcolepsy. Despite the central importance of the orexin system in regulating wakefulness and feeding behavior, little is known about the downstream signaling mechanisms that achieve these effects. In this study, genomics techniques are used to probe this question and reveal that orexin activates the hypoxia-inducible factor 1 (HIF-1), a heterodimeric transcription factor whose pathogenic role in stimulating angiogenesis in hypoxic tumors has been the focus of intense investigation. Orexin-stimulated HIF-1 activity is due to both increased HIF-1alpha gene transcription and a down-regulation of von Hippel-Lindau (VHL), the E3 ubiquitin ligase that mediates the turnover of HIF-1 via the ubiquitin-proteasome pathway. Orexin-mediated activation of HIF-1 results in increased glucose uptake and higher glycolytic activity, as expected from studies of hypoxic cells. However, orexin receptor-expressing cells somehow override the HIF-1-mediated preference for funneling pyruvate into anaerobic glycolysis and instead favor ATP production through the tricarboxylic acid cycle and oxidative phosphorylation. These findings implicate HIF-1 as an important transcription factor in the hormone-mediated regulation of hunger and wakefulness.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Adenosine Triphosphate/analysis , Animals , Cell Culture Techniques , Cell Line , Chromatin Immunoprecipitation , Cobalt/pharmacology , Culture Media, Serum-Free , Genes, Reporter , Glucose/metabolism , Humans , Hypothalamus/cytology , Kidney/cytology , Lactic Acid/analysis , Luciferases, Firefly/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins , Orexin Receptors , Orexins , Organ Culture Techniques , Plasmids , Proteins/metabolism , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/analysis , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Transfection
16.
J Comb Chem ; 9(4): 592-600, 2007.
Article in English | MEDLINE | ID: mdl-17530904

ABSTRACT

Synthetic molecules capable of activating the expression of specific genes are of great interest as tools for biological research and, potentially, as a novel class of pharmaceutical agents. It has been demonstrated previously that such synthetic transcription factor mimics (STFMs) can be constructed by connecting a sequence-specific DNA-binding module to a molecule capable of binding to the transcriptional machinery via a suitable linker. These chimeras mimic the two basic properties of native transcription factors, which are able to recognize a promoter sequence specifically and to recruit the transcriptional machinery to that promoter. However, none of the compounds of this type reported to date have been shown to function in living cells. We report here the first example of a cell-permeable STFM that activates the transcription of a reporter gene in mammalian cells. The compound is composed of a cell-permeable coactivator-binding peptoid fused to a DNA-binding hairpin polyamide. The peptoid was identified by screening a combinatorial library of approximately 50,000 compounds for binding to the KIX domain of the CREB-binding protein (CBP), a mammalian transcription coactivator. When incubated with cultured HeLa cells carrying a luciferase reporter plasmid bearing several hairpin polyamide-binding sites, a 5-fold increase in luciferase expression was observed. These experiments set the stage for the identification of hairpin polyamide-peptoid conjugates that are targeted to native genes.


Subject(s)
Biomimetic Materials/chemical synthesis , Biomimetic Materials/pharmacology , Cell Membrane Permeability/drug effects , Drug Design , Transcription Factors/chemistry , Transcription Factors/pharmacology , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/isolation & purification , Cell Line , Cell Survival , Cricetinae , Databases, Protein , Humans , Molecular Structure , Nylons/chemistry , Peptoids/chemical synthesis , Peptoids/chemistry , Peptoids/isolation & purification , Transcription, Genetic/genetics
18.
Genes Dev ; 21(1): 112-23, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17167105

ABSTRACT

Recent studies have shown that the intersection between transcription and proteins involved in the ubiquitin-proteasome pathway encompasses both proteolytic and nonproteolytic functions. Examples of the latter type include evidence that monoubiquitylation of some transcriptional activators stimulates their activity. In addition, the proteasomal ATPases are recruited to many active promoters through binding to activators and play an important, nonproteolytic role in promoter escape and elongation. In this study, we report the discovery of a new nonproteolytic activity of the proteasome (specifically the proteasomal ATPases): the active destabilization of activator-promoter complexes. This reaction depends on the presence of an activation domain and ATP. Destabilization is inhibited in vitro and in vivo if the protein is monoubiquitylated or if ubiquitin is genetically fused to the activator. The fact that monoubiquitylated activator is resistant to the "stripping" activity of the proteasomal ATPases may explain, in part, why some activators require this modification in order to function efficiently.


Subject(s)
Adenosine Triphosphatases/metabolism , Gene Expression Regulation, Fungal , Promoter Regions, Genetic/genetics , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/metabolism , Blotting, Western , Chromatin Immunoprecipitation , DNA-Binding Proteins , HeLa Cells , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , TATA Box/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation
19.
Cell ; 127(2): 261-4, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-17055428

ABSTRACT

Transcriptional activators need to be modulated and eventually switched off after the initial event that triggers their activation. Here, we discuss how ubiquitination of activators and their proteasome-mediated turnover are crucial steps in this process.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Trans-Activators/metabolism , Transcriptional Activation , Ubiquitin/metabolism , Animals , Humans , Models, Genetic , Time Factors , Transcription, Genetic/physiology
20.
J Biol Chem ; 281(37): 27346-55, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16837462

ABSTRACT

It has recently become clear that various aspects of nucleic acid metabolism and the ubiquitin-proteasome pathway intersect in several direct and important ways. To begin to assess the scope of some of these activities in the yeast Saccharomyces cerevisiae, we assessed the physical and functional association of proteasomal proteins from both the 20 S core and 19 S regulatory particles with approximately 6400 yeast genes. Genome-wide chromatin immunoprecipitation analyses revealed that proteasome substituents are associated with the majority of yeast genes. Many of these associations correlated strongly with expression levels and the presence of RNA polymerase II. Although the data support the presence of the intact 26 S proteasome on most genes, several hundred yeast genes were cross-linked to either the 20 or 19 S complex but not both, consistent with some degree of independent function for the proteasomal subcomplexes.


Subject(s)
Chromatin/chemistry , Gene Expression Regulation, Fungal , Proteasome Endopeptidase Complex/chemistry , Transcription, Genetic , Biochemistry/methods , Carbon/chemistry , Chromatin Immunoprecipitation , RNA Polymerase II/chemistry , Saccharomyces cerevisiae/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...