Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nucl Med Biol ; 112-113: 20-30, 2022.
Article in English | MEDLINE | ID: mdl-35763877

ABSTRACT

Extracellular vesicles (EVs) have garnered increasing interest as delivery vehicles for multiple classes of therapeutics based on their role as mediators in an important, natural intercellular communication system. We recently described a platform to allow the design, production and in vivo study of human EVs with specific properties (drug or tropism modifiers). This article seeks to compare and expand upon historical biodistribution and kinetic data by comparing systemically and compartmentally administered labeled engineered EVs using in vivo and ex vivo techniques. METHODS: EVs were surface-labeled to high radiochemical purity and specific activity with 89Zirconium deferoxamine ([89Zr]Zr-DFO) and/or cy7-scrambled antisense oligonucleotide (Cy7-ExoASOscr), or luminally loaded with GFP for in vivo tracking in rodents and non-human primates (NHPs). Positron Emission Tomography (PET) and subsequent immunohistochemistry (IHC) and autoradiography (ARG) cross-validation enabled assessment of the anatomical and cellular distribution of labeled EVs both spatially and temporally. RESULTS: Over time, systemic administration of engineered EVs distributed preferentially to the liver and spleen (Intravenous, IV), gastrointestinal tract and lymph nodes (Intraperitoneal, IP) and local/regional lymph nodes (Subcutaneous, SC). Immunostaining of dissected organs displaying PET signal revealed co-localization of an EV marker (PTGFRN) with a subset of macrophage markers (CD206, F4/80, IBA1). Compartmental dosing into NHP cerebrospinal fluid (CSF) resulted in a heterogenous distribution of labeled EVs depending upon whether the route was intrathecal (ITH), intracisterna magna (ICM) or intracerebroventricular (ICV), compared to the homogeneous distribution observed in rodents. Thus anatomically, ITH administration in NHP revealed meningeal distribution along the neuraxis to the base of the skull. In contrast ICM and ICV dosing resulted in meningeal distribution around the skull and to the cervical and thoracic spinal column. Further characterization using IHC shows uptake in a subset of meningeal macrophages. CONCLUSIONS: The present studies provide a comprehensive assessment of the fate of robustly and reproducibly labeled engineered EVs across several mammalian species. The in vivo distribution was observed to be both spatially and temporally dependent upon the route of administration providing insight into potential targeting opportunities for engineered EVs carrying a therapeutic payload.


Subject(s)
Extracellular Vesicles , Zirconium , Animals , Cell Line, Tumor , Deferoxamine/chemistry , Mammals , Oligonucleotides, Antisense , Positron-Emission Tomography/methods , Radioisotopes/chemistry , Tissue Distribution , Zirconium/chemistry
2.
Sci Adv ; 8(7): eabj7002, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35179953

ABSTRACT

Effectiveness of checkpoint immunotherapy in cancer can be undermined by immunosuppressive tumor-associated macrophages (TAMs) with an M2 phenotype. Reprogramming TAMs toward a proinflammatory M1 phenotype is a novel approach to induce antitumor immunity. The M2 phenotype is controlled by key transcription factors such as signal transducer and activator of transcription 6 (STAT6), which have been "undruggable" selectively in TAMs. We describe an engineered exosome therapeutic candidate delivering an antisense oligonucleotide (ASO) targeting STAT6 (exoASO-STAT6), which selectively silences STAT6 expression in TAMs. In syngeneic models of colorectal cancer and hepatocellular carcinoma, exoASO-STAT6 monotherapy results in >90% tumor growth inhibition and 50 to 80% complete remissions. Administration of exoASO-STAT6 leads to induction of nitric oxide synthase 2 (NOS2), an M1 macrophage marker, resulting in remodeling of the tumor microenvironment and generation of a CD8 T cell-mediated adaptive immune response. Collectively, exoASO-STAT6 represents the first platform targeting transcription factors in TAMs in a highly selective manner.


Subject(s)
Exosomes , Neoplasms , Exosomes/genetics , Exosomes/metabolism , Humans , Macrophages/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Tumor Microenvironment/genetics , Tumor-Associated Macrophages
3.
Commun Biol ; 4(1): 497, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888863

ABSTRACT

Cyclic dinucleotide (CDN) agonists of the STimulator of InterferoN Genes (STING) pathway have shown immune activation and tumor clearance in pre-clinical models. However, CDNs administered intratumorally also promote STING activation leading to direct cytotoxicity of many cell types in the tumor microenvironment (TME), systemic inflammation due to rapid tumor extravasation of the CDN, and immune ablation in the TME. These result in a failure to establish immunological memory. ExoSTING, an engineered extracellular vesicle (EV) exogenously loaded with CDN, enhances the potency of CDN and preferentially activates antigen presenting cells in the TME. Following intratumoral injection, exoSTING was retained within the tumor, enhanced local Th1 responses and recruitment of CD8+ T cells, and generated systemic anti-tumor immunity to the tumor. ExoSTING at therapeutically active doses did not induce systemic inflammatory cytokines, resulting in an enhanced therapeutic window. ExoSTING is a novel, differentiated therapeutic candidate that leverages the natural biology of EVs to enhance the activity of CDNs.


Subject(s)
Extracellular Vesicles/physiology , Immunologic Surveillance , Tumor Microenvironment/physiology , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
J Am Chem Soc ; 143(12): 4714-4724, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33739832

ABSTRACT

Prodrugs engineered for preferential activation in diseased versus normal tissues offer immense potential to improve the therapeutic indexes (TIs) of preclinical and clinical-stage active pharmaceutical ingredients that either cannot be developed otherwise or whose efficacy or tolerability it is highly desirable to improve. Such approaches, however, often suffer from trial-and-error design, precluding predictive synthesis and optimization. Here, using bromodomain and extra-terminal (BET) protein inhibitors (BETi)-a class of epigenetic regulators with proven anticancer potential but clinical development hindered in large part by narrow TIs-we introduce a macromolecular prodrug platform that overcomes these challenges. Through tuning of traceless linkers appended to a "bottlebrush prodrug" scaffold, we demonstrate correlation of in vitro prodrug activation kinetics with in vivo tumor pharmacokinetics, enabling the predictive design of novel BETi prodrugs with enhanced antitumor efficacies and devoid of dose-limiting toxicities in a syngeneic triple-negative breast cancer murine model. This work may have immediate clinical implications, introducing a platform for predictive prodrug design and potentially overcoming hurdles in drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Prodrugs/pharmacology , Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Molecular Structure , Prodrugs/chemical synthesis , Prodrugs/chemistry , Proteins/metabolism
5.
Mol Ther ; 29(5): 1729-1743, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33484965

ABSTRACT

Extracellular vesicles (EVs) are an important intercellular communication system facilitating the transfer of macromolecules between cells. Delivery of exogenous cargo tethered to the EV surface or packaged inside the lumen are key strategies for generating therapeutic EVs. We identified two "scaffold" proteins, PTGFRN and BASP1, that are preferentially sorted into EVs and enable high-density surface display and luminal loading of a wide range of molecules, including cytokines, antibody fragments, RNA binding proteins, vaccine antigens, Cas9, and members of the TNF superfamily. Molecules were loaded into EVs at high density and exhibited potent in vitro activity when fused to full-length or truncated forms of PTGFRN or BASP1. Furthermore, these engineered EVs retained pharmacodynamic activity in a variety of animal models. This engineering platform provides a simple approach to functionalize EVs with topologically diverse macromolecules and represents a significant advance toward unlocking the therapeutic potential of EVs.


Subject(s)
Extracellular Vesicles/transplantation , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proteins/administration & dosage , Repressor Proteins/metabolism , Animals , Cell Communication , Drug Delivery Systems , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Female , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Repressor Proteins/genetics
6.
Mol Cancer Ther ; 20(3): 523-534, 2021 03.
Article in English | MEDLINE | ID: mdl-33443094

ABSTRACT

The promise of IL12 as a cancer treatment has yet to be fulfilled with multiple tested approaches being limited by unwanted systemic exposure and unpredictable pharmacology. To address these limitations, we generated exoIL12, a novel, engineered exosome therapeutic that displays functional IL12 on the surface of an exosome. IL12 exosomal surface expression was achieved via fusion to the abundant exosomal surface protein PTGFRN resulting in equivalent potency in vitro to recombinant IL12 (rIL12) as demonstrated by IFNγ production. Following intratumoral injection, exoIL12 exhibited prolonged tumor retention and greater antitumor activity than rIL12. Moreover, exoIL12 was significantly more potent than rIL12 in tumor growth inhibition. In the MC38 model, complete responses were observed in 63% of mice treated with exoIL12; in contrast, rIL12 resulted in 0% complete responses at an equivalent IL12 dose. This correlated with dose-dependent increases in tumor antigen-specific CD8+ T cells. Rechallenge studies of exoIL12 complete responder mice showed no tumor regrowth, and depletion of CD8+ T cells completely abrogated antitumor activity of exoIL12. Following intratumoral administration, exoIL12 exhibited 10-fold higher intratumoral exposure than rIL12 and prolonged IFNγ production up to 48 hours. Retained local pharmacology of exoIL12 was further confirmed using subcutaneous injections in nonhuman primates. This work demonstrates that tumor-restricted pharmacology of exoIL12 results in superior in vivo efficacy and immune memory without systemic IL12 exposure and related toxicity. ExoIL12 is a novel cancer therapeutic candidate that overcomes key limitations of rIL12 and thereby creates a therapeutic window for this potent cytokine.


Subject(s)
Exosomes/metabolism , Interleukin-12/metabolism , Animals , Disease Models, Animal , Female , Humans , Macaca fascicularis , Mice
7.
Mol Psychiatry ; 24(11): 1641-1654, 2019 11.
Article in English | MEDLINE | ID: mdl-31481758

ABSTRACT

Although the underlying neurobiology of major mental illness (MMI) remains unknown, emerging evidence implicates a role for oligodendrocyte-myelin abnormalities. Here, we took advantage of a large family carrying a balanced t(1;11) translocation, which substantially increases risk of MMI, to undertake both diffusion tensor imaging and cellular studies to evaluate the consequences of the t(1;11) translocation on white matter structural integrity and oligodendrocyte-myelin biology. This translocation disrupts among others the DISC1 gene which plays a crucial role in brain development. We show that translocation-carrying patients display significant disruption of  white matter integrity compared with familial controls. At a cellular level, we observe dysregulation of key pathways controlling oligodendrocyte development and morphogenesis in induced pluripotent stem cell (iPSC) derived case oligodendrocytes. This is associated with reduced proliferation and a stunted morphology in vitro. Further, myelin internodes in a humanized mouse model that recapitulates the human translocation as well as after transplantation of t(1;11) oligodendrocyte progenitors were significantly reduced when  compared with controls. Thus we provide evidence that the t(1;11) translocation has biological effects at both the systems and cellular level that together suggest oligodendrocyte-myelin dysfunction.


Subject(s)
Myelin Sheath/metabolism , Oligodendroglia/metabolism , Translocation, Genetic/genetics , Adult , Animals , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 11/genetics , Diffusion Tensor Imaging/methods , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mental Disorders/genetics , Mice , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , White Matter/metabolism , White Matter/physiology
8.
J Lipid Res ; 60(4): 758-766, 2019 04.
Article in English | MEDLINE | ID: mdl-30696699

ABSTRACT

Eicosanoids are lipid-mediator molecules with key roles in inflammatory skin diseases, such as psoriasis. Eicosanoids are released close to the source of inflammation, where they elicit local pleiotropic effects and dysregulations. Monitoring inflammatory mediators directly in skin lesions could provide new insights and therapeutic possibilities. Here, we analyzed dermal interstitial fluid samples obtained by dermal open-flow microperfusion in a rat model of skin inflammation. We developed a solid-phase extraction ultra-HPLC/MS/MS method to reliably and precisely analyze small-volume samples and quantified 11 eicosanoids [thromboxane B2, prostaglandin (PG) E2, PGD2, PGF2α, leukotriene B4, 15-HETE, 12-HETE, 5-HETE, 12-hydroxyeicosapentaenoic acid, 13-HODE, and 17-hydroxydocosahexaenoic acid]. Our method achieved a median intraday precision of approximately 5% and interday precision of approximately 8%. All calibration curves showed excellent linearity between 0.01 and 50 ng/ml (R2 > 0.980). In the rat model, eicosanoids were significantly increased in imiquimod-treated inflamed skin sites compared with untreated control sites. Oral treatment with an anti-inflammatory glucocorticoid decreased eicosanoid concentrations. These results show that a combination of tissue-specific sampling with LC/MS analytics is well suited for analyzing small sample volumes from minimally invasive sampling methods such as open-flow microperfusion or microdialysis to study local inflammation and the effect of treatments in skin diseases.


Subject(s)
Disease Models, Animal , Eicosanoids/analysis , Inflammation/metabolism , Skin Diseases/metabolism , Skin/chemistry , Solid Phase Extraction , Animals , Anti-Inflammatory Agents/pharmacology , Chromatography, High Pressure Liquid , Eicosanoids/antagonists & inhibitors , Eicosanoids/metabolism , Glucocorticoids/pharmacology , Inflammation/drug therapy , Male , Rats , Rats, Sprague-Dawley , Skin/metabolism , Skin Diseases/drug therapy , Tandem Mass Spectrometry
9.
Transl Psychiatry ; 8(1): 184, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30190480

ABSTRACT

The neuromodulatory gene DISC1 is disrupted by a t(1;11) translocation that is highly penetrant for schizophrenia and affective disorders, but how this translocation affects DISC1 function is incompletely understood. N-methyl-D-aspartate receptors (NMDAR) play a central role in synaptic plasticity and cognition, and are implicated in the pathophysiology of schizophrenia through genetic and functional studies. We show that the NMDAR subunit GluN2B complexes with DISC1-associated trafficking factor TRAK1, while DISC1 interacts with the GluN1 subunit and regulates dendritic NMDAR motility in cultured mouse neurons. Moreover, in the first mutant mouse that models DISC1 disruption by the translocation, the pool of NMDAR transport vesicles and surface/synaptic NMDAR expression are increased. Since NMDAR cell surface/synaptic expression is tightly regulated to ensure correct function, these changes in the mutant mouse are likely to affect NMDAR signalling and synaptic plasticity. Consistent with these observations, RNASeq analysis of the translocation carrier-derived human neurons indicates abnormalities of excitatory synapses and vesicle dynamics. RNASeq analysis of the human neurons also identifies many differentially expressed genes previously highlighted as putative schizophrenia and/or depression risk factors through large-scale genome-wide association and copy number variant studies, indicating that the translocation triggers common disease pathways that are shared with unrelated psychiatric patients. Altogether, our findings suggest that translocation-induced disease mechanisms are likely to be relevant to mental illness in general, and that such disease mechanisms include altered NMDAR dynamics and excitatory synapse function. This could contribute to the cognitive disorders displayed by translocation carriers.


Subject(s)
Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Translocation, Genetic , Adaptor Proteins, Vesicular Transport , Animals , Carrier Proteins/genetics , Genome-Wide Association Study , Humans , Mice , Models, Animal , Mood Disorders/genetics , Mutation , Nerve Tissue Proteins/genetics , Neuronal Plasticity , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Sequence Analysis, RNA , Synapses/metabolism
10.
Nat Biomed Eng ; 2(11): 822-830, 2018 11.
Article in English | MEDLINE | ID: mdl-30918745

ABSTRACT

At present there are no drugs for the treatment of chronic liver fibrosis that have been approved by the Food and Drug administration of the United States. Telmisartan, a small-molecule antihypertensive drug, displays antifibrotic activity, but its clinical use is limited because it causes systemic hypotension. Here, we report the scalable and convergent synthesis of macromolecular telmisartan prodrugs optimized for preferential release in diseased liver tissue. We optimized the release of active telmisartan in fibrotic liver to be depot-like (that is, a constant therapeutic concentration) through the molecular design of telmisartan brush-arm star polymers, and show that these lead to improved efficacy and to the avoidance of dose-limiting hypotension in both metabolically and chemically induced mouse models of hepatic fibrosis, as determined by histopathology, enzyme levels in the liver, intact-tissue protein markers, hepatocyte necrosis protection, and gene-expression analyses. In rats and dogs, the prodrugs are retained long-term in liver tissue and have a well-tolerated safety profile. Our findings support the further development of telmisartan prodrugs that enable infrequent dosing in the treatment of liver fibrosis.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Drug Design , Liver Cirrhosis/drug therapy , Prodrugs/therapeutic use , Telmisartan/therapeutic use , Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Animals , Carbon Tetrachloride/toxicity , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Half-Life , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred BALB C , Polymers/chemistry , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats , Rats, Sprague-Dawley , Telmisartan/chemistry
11.
Nat Biomed Eng ; 2(9): 707, 2018 Sep.
Article in English | MEDLINE | ID: mdl-31015683

ABSTRACT

In the version of this Article originally published, the author Peter Blume-Jensen was not denoted as a corresponding author; this has now been amended and the author's email address has been added. The 'Correspondence and requests for materials' statement was similarly affected and has now been updated with the author's initials 'P.B-J.'

12.
J Biomol Screen ; 19(2): 232-41, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23896687

ABSTRACT

Numerous assays have been developed to investigate the interactions between G-protein-coupled receptors (GPCRs) and their ligands since GPCRs are key therapeutic targets. Reporter-based assays using the cAMP response element (CRE) coupled with bioluminescence from a luciferase reporter have been used extensively in vitro with high-throughput screens (HTS) of large chemical compound libraries. We have generated a transgenic mouse model (CRE luc) with a luciferase reporter under the control of a synthetic promoter that contains several CREs, which supports real-time bioimaging of GPCR ligand activity in whole animals, tissues, or primary cells. In the CRE luc model, GPCR signaling through the cAMP pathway can be detected from the target GPCR that is in a native cellular environment with a full complement of associated receptors and membrane constituents. Multiple independent lines have been produced by random integration of the transgene, resulting in tissue expression profiles covering the major organs. The goal of the CRE luc model is to accelerate the transition from HTS to profiling of GPCR small-molecule leads in preclinical animal disease models, as well as define the mechanism of action of GPCR drugs in three experimental formats: primary cells, tissue homogenates, and whole animals.


Subject(s)
Ligands , Receptors, G-Protein-Coupled/metabolism , Small Molecule Libraries/chemistry , Animals , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Luminescent Measurements , Mice , Mice, Transgenic , Promoter Regions, Genetic , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/isolation & purification , Transcriptome , Transfection
13.
J Neurosci ; 32(30): 10331-7, 2012 Jul 25.
Article in English | MEDLINE | ID: mdl-22836266

ABSTRACT

The cannabinoid receptor 1 (CB(1)R) is required for body weight homeostasis and normal gastrointestinal motility. However, the specific cell types expressing CB(1)R that regulate these physiological functions are unknown. CB(1)R is widely expressed, including in neurons of the parasympathetic branches of the autonomic nervous system. The vagus nerve has been implicated in the regulation of several aspects of metabolism and energy balance (e.g., food intake and glucose balance), and gastrointestinal functions including motility. To directly test the relevance of CB(1)R in neurons of the vagus nerve on metabolic homeostasis and gastrointestinal motility, we generated and characterized mice lacking CB(1)R in afferent and efferent branches of the vagus nerve (Cnr1(flox/flox); Phox2b-Cre mice). On a chow or on a high-fat diet, Cnr1(flox/flox); Phox2b-Cre mice have similar body weight, food intake, energy expenditure, and glycemia compared with Cnr1(flox/flox) control mice. Also, fasting-induced hyperphagia and after acute or chronic pharmacological treatment with SR141716 [N-piperidino-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-3-pyrazole carboxamide] (CB(1)R inverse agonist) paradigms, mutants display normal body weight and food intake. Interestingly, Cnr1(flox/flox); Phox2b-Cre mice have increased gastrointestinal motility compared with controls. These results unveil CB(1)R in the vagus nerve as a key component underlying normal gastrointestinal motility.


Subject(s)
Body Weight/genetics , Gastrointestinal Motility/genetics , Homeostasis/genetics , Receptor, Cannabinoid, CB1/metabolism , Vagus Nerve/metabolism , Animals , Body Weight/drug effects , Diet, High-Fat , Eating/drug effects , Eating/genetics , Food Deprivation , Gastrointestinal Motility/drug effects , Homeostasis/drug effects , Hyperphagia/genetics , Hyperphagia/metabolism , Mice , Mice, Transgenic , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Rimonabant
14.
Obesity (Silver Spring) ; 19(10): 1923-34, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21799481

ABSTRACT

Cannabinoid receptor CB1 is expressed abundantly in the brain and presumably in the peripheral tissues responsible for energy metabolism. It is unclear if the antiobesity effects of rimonabant, a CB1 antagonist, are mediated through the central or the peripheral CB1 receptors. To address this question, we generated transgenic mice with central nervous system (CNS)-specific knockdown (KD) of CB1, by expressing an artificial microRNA (AMIR) under the control of the neuronal Thy1.2 promoter. In the mutant mice, CB1 expression was reduced in the brain and spinal cord, whereas no change was observed in the superior cervical ganglia (SCG), sympathetic trunk, enteric nervous system, and pancreatic ganglia. In contrast to the neuronal tissues, CB1 was undetectable in the brown adipose tissue (BAT) or the liver. Consistent with the selective loss of central CB1, agonist-induced hypothermia was attenuated in the mutant mice, but the agonist-induced delay of gastrointestinal transit (GIT), a primarily peripheral nervous system-mediated effect, was not. Compared to wild-type (WT) littermates, the mutant mice displayed reduced body weight (BW), adiposity, and feeding efficiency, and when fed a high-fat diet (HFD), showed decreased plasma insulin, leptin, cholesterol, and triglyceride levels, and elevated adiponectin levels. Furthermore, the therapeutic effects of rimonabant on food intake (FI), BW, and serum parameters were markedly reduced and correlated with the degree of CB1 KD. Thus, KD of CB1 in the CNS recapitulates the metabolic phenotype of CB1 knockout (KO) mice and diminishes rimonabant's efficacy, indicating that blockade of central CB1 is required for rimonabant's antiobesity actions.


Subject(s)
Anti-Obesity Agents/pharmacology , Body Weight/drug effects , Central Nervous System/metabolism , Energy Intake/drug effects , Obesity/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Adiponectin/blood , Adiposity/drug effects , Adiposity/genetics , Animals , Anti-Obesity Agents/therapeutic use , Biomarkers/blood , Body Weight/genetics , Central Nervous System/drug effects , Cholesterol/blood , Diet, High-Fat/adverse effects , Energy Intake/genetics , Gastrointestinal Transit/physiology , Hypothermia/prevention & control , Insulin/blood , Leptin/blood , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs , Mutation , Obesity/drug therapy , Obesity/genetics , Peripheral Nervous System/drug effects , Peripheral Nervous System/metabolism , Phenotype , Piperidines/therapeutic use , Promoter Regions, Genetic , Pyrazoles/therapeutic use , Receptor, Cannabinoid, CB1/genetics , Rimonabant , Triglycerides/blood
15.
Nature ; 461(7263): 495-500, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19741607

ABSTRACT

In epithelial tissues, the lineage relationship between normal progenitor cells and cell type(s) of origin for cancer has been poorly understood. Here we show that a known regulator of prostate epithelial differentiation, the homeobox gene Nkx3-1, marks a stem cell population that functions during prostate regeneration. Genetic lineage-marking demonstrates that rare luminal cells that express Nkx3-1 in the absence of testicular androgens (castration-resistant Nkx3-1-expressing cells, CARNs) are bipotential and can self-renew in vivo, and single-cell transplantation assays show that CARNs can reconstitute prostate ducts in renal grafts. Functional assays of Nkx3-1 mutant mice in serial prostate regeneration suggest that Nkx3-1 is required for stem cell maintenance. Furthermore, targeted deletion of the Pten tumour suppressor gene in CARNs results in rapid carcinoma formation after androgen-mediated regeneration. These observations indicate that CARNs represent a new luminal stem cell population that is an efficient target for oncogenic transformation in prostate cancer.


Subject(s)
Cell Lineage , Epithelial Cells/pathology , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Androgens/deficiency , Androgens/metabolism , Animals , Castration , Cell Differentiation , Cell Division , Cell Transformation, Neoplastic , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kidney , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Regeneration , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Genes Dev ; 20(7): 784-94, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16600910

ABSTRACT

The mechanisms by which homeoproteins bind selectively to target genes in vivo have long remained unresolved. Here we report that PIAS1 confers DNA-binding specificity on the Msx1 homeoprotein by regulating its subnuclear localization and proximity to target genes. We demonstrate that the interaction of Msx1 with PIAS1, but not its sumoylation, is required for Msx1 to function as an inhibitor of myoblast differentiation through repression of myogenic regulatory genes, such as MyoD. We find that PIAS1 enables Msx1 to bind selectively to a key regulatory element in MyoD, the CER, in myoblast cells and to distinguish the CER from other nonregulatory TAAT-containing sequences. We show that PIAS1 is required for the appropriate localization and retention of Msx1 at the nuclear periphery in myoblast cells. Furthermore, we demonstrate that myogenic regulatory genes that are repressed by Msx1, namely MyoD and Myf5, are located at the nuclear periphery in myoblast cells. We propose that a key regulatory event for DNA-binding specificity by homeoproteins in vivo is their appropriate targeting to subnuclear compartments where their target genes are located, which can be achieved by cofactors such as PIAS1.


Subject(s)
DNA/metabolism , MSX1 Transcription Factor/metabolism , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Binding Sites/genetics , Cell Line , Cell Nucleus/metabolism , DNA/genetics , Humans , MSX1 Transcription Factor/genetics , Mice , Models, Biological , Muscle Development , Myoblasts/metabolism , Protein Inhibitors of Activated STAT/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/metabolism
17.
Cancer Res ; 63(14): 3886-90, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12873978

ABSTRACT

Recent studies have shown that several loss-of-function mouse models of prostate carcinogenesis can develop a spectrum of precancerous lesions that resemble human prostatic intraepithelial neoplasia (PIN). Here, we have investigated the malignant potential of the high-grade PIN lesions that form in Nkx3.1(+/-); Pten(+/-) compound mutant mice and demonstrate their neoplastic progression in a serial transplantation/tissue recombination assay. Furthermore, we find that a majority of Nkx3.1(+/-); Pten(+/-) mice greater than 1 year of age develop invasive adenocarcinoma, which is frequently accompanied by metastases to lymph nodes. Finally, we observe androgen independence of high-grade PIN lesions after androgen ablation of Nkx3.1(+/-); Pten(+/-) mice. We conclude that Nkx3.1(+/-); Pten(+/-) mice recapitulate key features of advanced prostate cancer and represent a useful model for investigating associated molecular mechanisms and for evaluating therapeutic approaches.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Homeodomain Proteins/genetics , Mutation , Phosphoric Monoester Hydrolases/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adenocarcinoma/surgery , Animals , Disease Models, Animal , Humans , Lymphatic Metastasis , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Orchiectomy , PTEN Phosphohydrolase , Prostatic Neoplasms/surgery , Rats , Testosterone/blood
18.
Dev Biol ; 256(2): 317-30, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12679105

ABSTRACT

To address the expression and function of Hoxb13, the 5' most Hox gene in the HoxB cluster, we have generated mice with loss-of-function and beta-galactosidase reporter insertion alleles of this gene. Mice homozygous for Hoxb13 loss-of-function mutations show overgrowth in all major structures derived from the tail bud, including the developing secondary neural tube (SNT), the caudal spinal ganglia, and the caudal vertebrae. Using the beta-galactosidase reporter allele of Hoxb13, also a loss-of-function allele, we found that the expression patterns of Hoxb13 in the developing spinal cord and caudal mesoderm are closely associated with overgrowth phenotypes in the tails of homozygous mutant animals. These phenotypes can be explained by the observed increased cell proliferation and decreased levels of apoptosis within the tail of homozygous mutant mice. This analysis of Hoxb13 function suggests that this 5' Hox gene may act as an inhibitor of neuronal cell proliferation, an activator of apoptotic pathways in the SNT, and as a general repressor of growth in the caudal vertebrae.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mutation , Spine/embryology , Tail/embryology , Animals , Axons/pathology , Ganglia, Spinal/abnormalities , Mice , Myoblasts/physiology , Spinal Cord/abnormalities , Spinal Cord/embryology , Spine/abnormalities , Tail/abnormalities
19.
Development ; 130(10): 2061-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12668621

ABSTRACT

The murine prostate is a structure that is made up of four distinct lobes; the dorsal and lateral prostates (often grouped together as the dorsolateral prostate), the anterior (coagulating gland) and the ventral prostate. Previous work has implicated Hox genes in the development of these structures, but how each lobe acquires unique identities for specific functions has not been addressed. In this study, the ventral prostate-specific function of Hoxb13 is described. Mice lacking Hoxb13 function show normal numbers of duct tips, but mice mutant for both Hoxb13 and Hoxd13 exhibit severe hypoplasia of the duct tips, revealing a role for Hoxb13 in ventral prostate morphogenesis. Additionally, a ventral lobe-specific defect was identified in Hoxb13 mutants wherein the epithelium is composed of simple cuboidal cells rather than of tall columnar cells. Ventral prostate ducts appear devoid of contents and do not express the ventral prostate-specific secretory proteins p12, a kazal-type protease inhibitor and p25, a spermine binding protein. These defects are not due to reduction of Nkx3.1 expression or to a global effect on androgen receptor signaling. These results suggest a specific role for Hoxb13 in a differentiation pathway that gives the ventral prostate epithelium a unique identity, as well as a more general role in ventral prostate morphogenesis that is redundant with other Hox13 paralogs.


Subject(s)
Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Prostate/growth & development , Prostate/metabolism , Prostatic Secretory Proteins/metabolism , Animals , Biomarkers , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Homeodomain Proteins/genetics , Humans , Male , Mice , Mutation , Phenotype , Prostate/anatomy & histology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...