Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Neurooncol ; 168(1): 125-138, 2024 May.
Article in English | MEDLINE | ID: mdl-38563850

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) often metastasizes to the central nervous system (CNS) and has the highest propensity among breast cancer subtypes to develop leptomeningeal disease (LMD). LMD is a spread of cancer into leptomeningeal space that speeds up the disease progression and severely aggravates the prognosis. LMD has limited treatment options. We sought to test whether the common anti-helminthic drug mebendazole (MBZ) may be effective against murine TNBC LMD. METHODS: A small-molecule screen involving TNBC cell lines identified benzimidazoles as potential therapeutic agents for further study. In vitro migration assays were used to evaluate cell migration capacity and the effect of MBZ. For in vivo testing, CNS metastasis was introduced into BALB/c athymic nude mice through internal carotid artery injections of brain-tropic MDA-MB-231-BR or MCF7-BR cells. Tumor growth and spread was monitored by bioluminescence imaging and immunohistochemistry. MBZ was given orally at 50 and 100 mg/kg doses. MBZ bioavailability was assayed by mass spectrometry. RESULTS: Bioinformatic analysis and migration assays revealed higher migratory capacity of TNBC compared to other breast cancer subtypes. MBZ effectively slowed down migration of TNBC cell line MDA-MB-231 and its brain tropic derivative MDA-MB-231-BR. In animal studies, MBZ reduced leptomeningeal spread, and extended survival in brain metastasis model produced by MDA-MB-231-BR cells. MBZ did not have an effect in the non-migratory MCF7-BR model. CONCLUSIONS: We demonstrated that MBZ is a safe and effective oral agent in an animal model of TNBC CNS metastasis. Our findings are concordant with previous efforts involving MBZ and CNS pathology and support the drug's potential utility to slow down leptomeningeal spread.


Subject(s)
Cell Movement , Drug Repositioning , Mebendazole , Mice, Inbred BALB C , Mice, Nude , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Animals , Humans , Female , Mebendazole/pharmacology , Mebendazole/therapeutic use , Mice , Cell Movement/drug effects , Xenograft Model Antitumor Assays , Cell Line, Tumor , Central Nervous System Neoplasms/secondary , Central Nervous System Neoplasms/drug therapy , Cell Proliferation/drug effects
2.
Res Sq ; 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38405839

ABSTRACT

Purpose: Triple-negative breast cancer (TNBC) is an aggressive subtype that often metastasizes to the brain. Leptomeningeal disease (LMD), a devastating brain metastasis common in TNBC, has limited treatment options. We sought to test whether the common anti-helminthic drug mebendazole (MBZ) may be effective against murine TNBC LMD. Methods: A small-molecule screen involving TNBC cell lines identified benzimidazoles as potential therapeutic agents for further study. In vitro migration assays were used to evaluate cell migration capacity and the effect of MBZ. For in vivo testing, LMD was introduced into BALB/c athymic nude mice through internal carotid artery injections of brain-tropic MDA-MB-231-BR or MCF7-BR cells. Tumor growth and spread was monitored by bioluminescence imaging. MBZ was given orally at 50 and 100 mg/kg doses. MBZ bioavailability was assayed by mass spectrometry. Results: Bioinformatic analysis and migration assays revealed higher migratory capacity of TNBC compared to other breast cancer subtypes. MBZ effectively slowed down migration of TNBC cell line MDA-MB-231 and its brain tropic derivative MDA-MB-231-BR. In animal studies, MBZ reduced tumor growth and extended survival in the LMD model produced by MDA-MB-231-BR cells. MBZ did not have an effect in the non-migratory MCF7-BR model. Conclusions: We demonstrated that MBZ is a safe and effective oral agent in an animal model of TNBC LMD. Our findings are concordant with previous efforts involving MBZ and central nervous system pathology and further support the drug's potential utility as an alternative therapeutic for TNBC LMD.

3.
Steroids ; 201: 109336, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37944652

ABSTRACT

Our research used glucocorticoids as a medically relevant molecular probe to identify a previously unrecognized ADAMTS1-PTN-Wnt pathway. We elucidated the role of this pathway in regulating adipose precursor cell (APC) behavior to either proliferate or differentiate in response to systemic cues, such as elevated caloric intake. Further, our studies identified the non-muscle myosin protein MYH9 as a key target of this pathway to modulate adipogenesis in vivo. These findings enable strategies toward developing novel therapeutics for obesity and related metabolic disorders.


Subject(s)
Adipogenesis , Glucocorticoids , Humans , Glucocorticoids/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Adipocytes/metabolism
4.
STAR Protoc ; 4(4): 102607, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37742183

ABSTRACT

Pro-preadipocytes are adipocyte progenitor cells within subcutaneous adipose tissue that are conserved in human adipose tissue with distinct cellular energetics. Here, we detail a protocol to quantify cellular oxygen consumption rates of primary human cells harvested from adipose tissue. We describe steps for primary cell expansion, cell seeding, transfection, differentiation, and respirometry followed by Agilent Seahorse Analytics. The measurement of bioenergetic profiles and resulting data further expand our knowledge of the functional properties of primary cells isolated from adipose tissue. For complete details on the use and execution of this protocol, please refer to Chen et al. (2023).1.


Subject(s)
Adipocytes , Adipose Tissue , Humans , Gene Knockdown Techniques , Cell Differentiation/genetics , Stem Cells
5.
Cell Rep ; 42(5): 112440, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37119138

ABSTRACT

Elucidating the transitional stages that define the pathway stem cells progress through during differentiation advances our understanding of biology and fosters the identification of therapeutic opportunities. However, distinguishing progenitor cells from other cell types and placing them in an epistatic pathway is challenging. This is exemplified in the adipocyte lineage, where the stromal vascular fraction (SVF) from adipose tissue is enriched for progenitor cells but also contains heterogeneous populations of cells. Single-cell RNA sequencing (scRNA-seq) has begun to facilitate the deconvolution of cell types in the SVF, and a hierarchical structure is emerging. Here, we use scRNA-seq to discover a population of CD31- CD45- cells in the SVF that are distinguished by a specific expression profile. Further, we place this population on an epistatic pathway upstream of the previously defined preadipocyte population. Finally, we discover functional properties of this population with broad implications, including revealing physiological mechanisms that regulate adipogenesis.


Subject(s)
Adipose Tissue , Stromal Cells , Stromal Cells/metabolism , Adipose Tissue/metabolism , Adipocytes , Cell Differentiation , Stem Cells
6.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33986190

ABSTRACT

White adipose tissue not only serves as a reservoir for energy storage but also secretes a variety of hormonal signals and modulates systemic metabolism. A substantial amount of adipose tissue develops in early postnatal life, providing exceptional access to the formation of this important tissue. Although a number of factors have been identified that can modulate the differentiation of progenitor cells into mature adipocytes in cell-autonomous assays, it remains unclear which are connected to physiological extracellular inputs and are most relevant to tissue formation in vivo. Here, we elucidate that mature adipocytes themselves signal to adipose depot-resident progenitor cells to direct depot formation in early postnatal life and gate adipogenesis when the tissue matures. Our studies revealed that as the adipose depot matures, a signal generated in mature adipocytes is produced, converges on progenitor cells to regulate the cytoskeletal protein MYH9, and attenuates the rate of adipogenesis in vivo.


Subject(s)
ADAMTS1 Protein/genetics , Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue, White/metabolism , Homeostasis/genetics , Myosin Heavy Chains/genetics , Stem Cells/metabolism , ADAMTS1 Protein/metabolism , Adipose Tissue/metabolism , Animals , Male , Mice , Mice, Transgenic , Myosin Heavy Chains/metabolism
7.
ACS Chem Biol ; 15(6): 1381-1391, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32255605

ABSTRACT

The complexity of glucocorticoid receptor (GR) signaling cannot be measured with direct tissue analysis in living subjects, which has stifled our understanding of GR's role in human physiology or disease and impeded the development of selective GR modulators. Herein, we report 18F-5-(4-fluorobenzyl)-10-methoxy-2,2,4-trimethyl-2,5-dihydro-1H-chromeno[3,4-f]quinoline (18F-YJH08), a radioligand that enables noninvasive measurements of tissue autonomous GR expression levels in vivo with positron emission tomography (PET). YJH08 potently binds GR (Ki ∼ 0.4 nM) with ∼100-fold selectivity compared to nuclear hormone receptors in the same subfamily. 18F-YJH08 was prepared via Cu(OTf)2(py)4-mediated radiofluorination of an arylboronic acid pinacol ester with ∼12% decay corrected radiochemical yield from the starting 18F-fluoride ion. We applied treatment with the tissue-wide GR agonist dexamethasone and adrenalectomy and generated an adipocyte specific GR knockout mouse to show that 18F-YJH08 specifically binds GR in normal mouse tissues, including those for which aberrant GR expression is thought to drive severe diseases (e.g., brain, adipose tissue, kidneys). Remarkably, 18F-YJH08 PET also revealed that JG231, a potent and bioavailable HSP70 inhibitor, selectively degrades GR only in the adipose tissue of mice, a finding that foreshadows how GR targeted PET might be integrated into drug discovery to screen for selective GR modulation at the tissue level, beyond the historical screening that was performed at the transcriptional level. In summary, 18F-YJH08 enables a quantitative assessment of GR expression levels in real time among multiple tissues simultaneously, and this technology is a first step toward unraveling the daunting complexity of GR signaling and rationally engineering tissue specific therapeutic modulators in vivo.


Subject(s)
Fluorine Radioisotopes/chemistry , Positron-Emission Tomography/methods , Quinolines/chemistry , Receptors, Glucocorticoid/analysis , Animals , Dexamethasone/pharmacology , Gene Expression/drug effects , Glucocorticoids/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/genetics
8.
PLoS One ; 14(2): e0211596, 2019.
Article in English | MEDLINE | ID: mdl-30730939

ABSTRACT

The objective of this study was to test if a novel platform could be used for isotype-specific autoantibody testing in humans. Further, we evaluated if testing with this novel platform enables earlier detection of insulin autoantibodies in individuals that have first-degree relatives with type-1 diabetes than currently used approaches. Longitudinal serum samples from participants were collected before and after they converted to become positive for insulin autoantibodies by the current standardly used assays. Using a novel plasmonic gold chip platform, we tested these samples for IgM isotype-specific autoantibodies. Serial serum samples from individuals without diabetes were also tested as a comparison control cohort. Our results demonstrate proof-of-concept that a plasmonic gold chip can specifically detect the IgM insulin autoantibody. Five out of the six individuals that converted to being positive for insulin autoantibodies by standard testing had significant IgM autoantibodies on the plasmonic chip platform. The plasmonic chip platform detected IgM autoantibodies earlier than standard testing by up to 4 years. Our results indicate that the plasmonic gold platform can specifically detect the IgM isotype autoantibodies and suggest that combining isotype-specific testing with currently used approaches enables earlier detection of insulin autoantibodies in individuals that have first-degree relatives with type 1 diabetes.


Subject(s)
Autoantibodies/immunology , Immunoglobulin Isotypes/immunology , Adolescent , Child , Child, Preschool , Cohort Studies , Diabetes Mellitus, Type 1/immunology , Female , Gold/immunology , Humans , Immunoglobulin M/immunology , Insulin/immunology , Insulin Antibodies/immunology , Male
9.
Adipocyte ; 7(4): 273-276, 2018.
Article in English | MEDLINE | ID: mdl-30153756

ABSTRACT

The circadian clock is an intricate molecular network that paces a variety of physiological process to ~ 24 hour day/night cycles. Whereas the central circadian clock in the brain is primarily entrained by light signals, peripheral circadian clocks, which are in most cells in the body, receive cues not only from the central pacemaker but also endocrine and other systemic and tissue-specific signals. Prior studies have connected peripheral circadian clocks to metabolism, primarily with studies focused on the robust clock in the liver that responds to feeding/fasting cycles. Adipose tissue is also critical for metabolism and adipocytes have circadian clocks. Yet, the role of the circadian clock in adipocytes is poorly understood. Here we describe our studies that revealed components of the circadian clock in primary adipocyte precursor cells (APCs) in mice. We made the surprising discovery of a particularly prominent role for the circadian gene Period 3 (Per3) in the APC clock. Furthermore, we elucidated that Per3 directly regulates an output pathway of the APC clock to modulate the expression of the Kruppel-like factor 15 (Klf15) gene. Finally, we discovered that this clock-Klf15 pathway regulates adipogenesis in APCs. These finding have important implications for our understanding of adipose tissue biology and metabolism and, we speculate, will generate opportunities to develop novel therapeutic strategies based on the context-specific features of the circadian clock in APCs.


Subject(s)
Adipocytes/cytology , Adipogenesis , Circadian Clocks , Period Circadian Proteins/metabolism , Adipocytes/metabolism , Animals , Cells, Cultured , Circadian Rhythm , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors , Mice , Period Circadian Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Steroid Biochem Mol Biol ; 177: 155-158, 2018 03.
Article in English | MEDLINE | ID: mdl-28710021

ABSTRACT

Several epidemiological studies have found that low vitamin D levels are associated with worse prognosis and poorer outcomes in patients with breast cancer (BCa), although some studies have failed to find this association. In addition, prior research has found that BCa patients with vitamin D deficiency have a more aggressive molecular phenotype and worse prognostic biomarkers. As vitamin D deficiency is common in patients diagnosed with BCa, elucidating the cause of the association between poor outcomes and vitamin D deficiency promises to have a significant impact on improving care for patients with BCa including enabling the development of novel therapeutic approaches. Here we review our recent findings in this area, including our data revealing that reduction of the expression of the vitamin D receptor (Vdr) within BCa cells accelerates primary tumor growth and enables the development of metastases, demonstrating a tumor autonomous effect of vitamin D signaling to suppress BCa metastases. We believe that these findings are likely relevant to humans as we discovered evidence that a mechanism of VDR regulation identified in our mouse models is conserved in human BCa. In particular, we identified a negative correlation between serum 25(OH)D concentration and the level of expression of the tumor progression factor ID1 in primary tumors from patients with breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Vitamin D/metabolism , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Female , Humans
11.
Cell Rep ; 21(9): 2367-2375, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29186676

ABSTRACT

The generation of new adipocytes from precursor cells (adipogenesis) has implications for systemic metabolism and is a commonly used model for studying the process of cell differentiation in vitro. Previous studies from us and others suggested that the peripheral circadian clock can influence adipogenesis in vitro, but the mechanisms driving this activity and the relevance for adipogenesis in vivo are unknown. Here we reveal that mouse adipocyte precursor cells (APCs) contain a circadian clock that oscillates in vivo. We expose context-specific features of the clock in APCs: expression of the canonical core clock component Per1 does not significantly oscillate, whereas the lesser-understood paralog Per3 has a prominent rhythm. We discovered that deletion of Per3 promotes adipogenesis in vivo by a clock output pathway in which PER3 and BMAL1 directly regulate Klf15 expression. These findings demonstrate that Per3 has a major role in the APC clock and regulates adipogenesis in vivo.


Subject(s)
Adipogenesis/physiology , Circadian Clocks/physiology , DNA-Binding Proteins/metabolism , Period Circadian Proteins/metabolism , Transcription Factors/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Circadian Clocks/genetics , Circadian Rhythm/genetics , Circadian Rhythm/physiology , DNA-Binding Proteins/genetics , Kruppel-Like Transcription Factors , Mice , Period Circadian Proteins/genetics , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/genetics
12.
Nat Commun ; 8(1): 669, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28939843

ABSTRACT

Coordinated activation of muscle stem cells (known as satellite cells) is critical for postnatal muscle growth and regeneration. The muscle stem cell niche is central for regulating the activation state of satellite cells, but the specific extracellular signals that coordinate this regulation are poorly understood. Here we show that macrophages at sites of muscle injury induce activation of satellite cells via expression of Adamts1. Overexpression of Adamts1 in macrophages in vivo is sufficient to increase satellite cell activation and improve muscle regeneration in young mice. We demonstrate that NOTCH1 is a target of ADAMTS1 metalloproteinase activity, which reduces Notch signaling, leading to increased satellite cell activation. These results identify Adamts1 as a potent extracellular regulator of satellite cell activation and have significant implications for understanding the regulation of satellite cell activity and regeneration after muscle injury.Satellite cells are crucial for growth and regeneration of skeletal muscle. Here the authors show that in response to muscle injury, macrophages secrete Adamts1, which induces satellite cell activation by modulating Notch1 signaling.


Subject(s)
ADAMTS1 Protein/genetics , Macrophages/metabolism , Receptor, Notch1/genetics , Satellite Cells, Skeletal Muscle/metabolism , ADAMTS1 Protein/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice, Transgenic , Muscle Development/genetics , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Mutation , Receptor, Notch1/metabolism , Regeneration/genetics , Signal Transduction/genetics
13.
Adipocyte ; 6(4): 293-297, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28700319

ABSTRACT

Intuitively, excess caloric intake causes adipose tissue expansion. However, the signals and mechanisms by which this systemic trigger directs a local response in the adipose tissue are incompletely understood. Both hypertrophy of existing adipocytes and the generation of new adipocytes through differentiation of adipocyte precursor cells (APCs), contribute to adipose tissue expansion in response to changes in the diet. Ex vivo studies of this process elucidated an elegant network of mostly transcription factors that drive APCs through the differentiation (adipogenesis) process. Here we discuss our study that identified an Adamts1 signal as a glucocorticoid and diet responsive regulator of an extracellular relay system that modulates the initiation of this intracellular adipogenesis program in APCs. Furthermore, we describe how we applied sensitive tools that enable monitoring of endogenous APC activity to study the early response to high-fat diet in vivo.


Subject(s)
ADAMTS1 Protein/genetics , Adipocytes/metabolism , Adipogenesis , Cues , ADAMTS1 Protein/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Differentiation/genetics , Diet, High-Fat , Male , Mice , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Stem Cells/cytology , Stem Cells/metabolism
14.
JCI Insight ; 2(2): e90427, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28138562

ABSTRACT

Amphetamine (AMPH) or methamphetamine (METH) abuse can cause oxidative damage and is a risk factor for diseases including pulmonary arterial hypertension (PAH). Pulmonary artery endothelial cells (PAECs) from AMPH-associated-PAH patients show DNA damage as judged by γH2AX foci and DNA comet tails. We therefore hypothesized that AMPH induces DNA damage and vascular pathology by interfering with normal adaptation to an environmental perturbation causing oxidative stress. Consistent with this, we found that AMPH alone does not cause DNA damage in normoxic PAECs, but greatly amplifies DNA damage in hypoxic PAECs. The mechanism involves AMPH activation of protein phosphatase 2A, which potentiates inhibition of Akt. This increases sirtuin 1, causing deacetylation and degradation of HIF1α, thereby impairing its transcriptional activity, resulting in a reduction in pyruvate dehydrogenase kinase 1 and impaired cytochrome c oxidase 4 isoform switch. Mitochondrial oxidative phosphorylation is inappropriately enhanced and, as a result of impaired electron transport and mitochondrial ROS increase, caspase-3 is activated and DNA damage is induced. In mice given binge doses of METH followed by hypoxia, HIF1α is suppressed and pulmonary artery DNA damage foci are associated with worse pulmonary vascular remodeling. Thus, chronic AMPH/METH can induce DNA damage associated with vascular disease by subverting the adaptive responses to oxidative stress.


Subject(s)
Amphetamine-Related Disorders/genetics , Amphetamines/pharmacology , DNA Damage/drug effects , Endothelial Cells/drug effects , Hypertension, Pulmonary/genetics , Hypoxia/genetics , Methamphetamine/pharmacology , Mitochondria/drug effects , Adult , Amphetamine-Related Disorders/metabolism , Animals , Caspase 3/drug effects , Caspase 3/metabolism , Electron Transport/drug effects , Endothelial Cells/metabolism , Female , Humans , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Male , Mice , Middle Aged , Mitochondria/metabolism , Oxidative Phosphorylation , Protein Phosphatase 2/drug effects , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Reactive Oxygen Species/metabolism , Sirtuin 1/drug effects , Sirtuin 1/metabolism , Vascular Remodeling/drug effects , Vascular Remodeling/genetics
15.
Sci Signal ; 9(451): ra103, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27811141

ABSTRACT

Obesity is driven by excess caloric intake, which leads to the expansion of adipose tissue by hypertrophy and hyperplasia. Adipose tissue hyperplasia results from the differentiation of adipocyte precursor cells (APCs) that reside in adipose depots. Investigation into this process has elucidated a network of mostly transcription factors that drive APCs through the differentiation process. Using in vitro and in vivo approaches, our study revealed a signaling pathway that inhibited the initiation of the adipocyte differentiation program. Mouse adipocytes secreted the extracellular protease ADAMTS1, which triggered the production of the cytokine pleiotrophin (PTN) through the Wnt/ß-catenin pathway, and promoted proliferation rather than differentiation of APCs. Glucocorticoid exposure in vitro or in vivo reduced ADAMTS1 abundance in adipocytes. In addition, mice fed a high-fat diet showed decreased Adamts1 expression in the visceral perigonadal adipose depot, which expanded by adipogenesis in response to the diet, and increased Adamts1 expression in the subcutaneous inguinal adipose depot, which did not induce adipogenesis. Similar to what occurred in mouse subcutaneous adipose tissue, diet-induced weight gain increased the expression of ADAMTS1, PTN, and certain Wnt target genes in the subcutaneous adipose depot of human volunteers, suggesting the relevance of this pathway to physiological adipose tissue homeostasis and the pathogenesis of obesity. Thus, this pathway functions as a toggle on APCs, regulating a decision between differentiation and proliferation and coordinating the response of adipose tissue to systemic cues.


Subject(s)
Adipocytes/metabolism , Diet/adverse effects , Glucocorticoids/pharmacology , Obesity/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/drug effects , ADAMTS1 Protein/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Mice , Obesity/chemically induced , Obesity/pathology , Stem Cells/pathology
16.
Endocrinology ; 157(4): 1341-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26934299

ABSTRACT

Patients with breast cancer (BCa) frequently have preexisting vitamin D deficiency (low serum 25-hydroxyvitamin D) when their cancer develops. A number of epidemiological studies show an inverse association between BCa risk and vitamin D status in humans, although some studies have failed to find an association. In addition, several studies have reported that BCa patients with vitamin D deficiency have a more aggressive molecular phenotype and worse prognostic indicators. However, it is unknown whether this association is mechanistically causative and, if so, whether it results from systemic or tumor autonomous effects of vitamin D signaling. We found that ablation of vitamin D receptor expression within BCa cells accelerates primary tumor growth and enables the development of metastases, demonstrating a tumor autonomous effect of vitamin D signaling to suppress BCa metastases. We show that vitamin D signaling inhibits the expression of the tumor progression gene Id1, and this pathway is abrogated in vitamin D deficiency in vivo in 2 murine models of BCa. These findings are relevant to humans, because we discovered that the mechanism of VDR regulation of Inhibitor of differentiation 1 (ID1) is conserved in human BCa cells, and there is a negative correlation between serum 25-hydroxyvitamin D levels and the level of ID1 in primary tumors from patients with BCa.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction , Vitamin D Deficiency/metabolism , Animals , Blotting, Western , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/genetics , Mice, Inbred BALB C , Neoplasm Metastasis , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D/metabolism , Vitamin D Deficiency/complications , Vitamin D Deficiency/genetics
17.
Endocr Relat Cancer ; 23(4): 251-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26817629

ABSTRACT

Obesity is an established risk factor for postmenopausal breast cancer (BCa), insulin resistance, and vitamin D deficiency, and all contribute to increased synthesis of mammary estrogens, the drivers of estrogen receptor-positive (ER+) BCa growth. As both dietary vitamin D and calcitriol treatments inhibit breast estrogen synthesis and signaling, we hypothesized that vitamin D would be especially beneficial in mitigating the adverse effects of obesity on ER+BCa. To assess whether obesity exerted adverse effects on BCa growth and whether vitamin D compounds could reduce these unfavorable effects, we employed a diet-induced obesity (DIO) model in ovariectomized C57BL/6 mice. Breast tumor cells originally from syngeneic Mmtv-Wnt1 transgenic mice were then implanted into the mammary fat pads of lean and obese mice. DIO accelerated the initiation and progression of the mammary tumors. Treatments with either calcitriol or dietary vitamin D reduced the adverse effects of obesity causing a delay in tumor appearance and inhibiting continued tumor growth. Beneficial actions of treatments with vitamin D or calcitriol on BCa and surrounding adipose tissue included repressed Esr1, aromatase, and Cox2 expression; decreased tumor-derived estrogen and PGE2; reduced expression of leptin receptors; and increased adiponectin receptors. We demonstrate that vitamin D treatments decreased insulin resistance, reduced leptin, and increased adiponectin signaling and also regulated the LKB1/AMPK pathway contributing to an overall decrease in local estrogen synthesis in the obese mice. We conclude that calcitriol and dietary vitamin D, acting by multiple interrelated pathways, mitigate obesity-enhanced BCa growth in a postmenopausal setting.


Subject(s)
Dietary Supplements , Mammary Neoplasms, Experimental/metabolism , Obesity/metabolism , Vitamin D/pharmacology , AMP-Activated Protein Kinases/metabolism , Adiponectin/blood , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Aromatase/genetics , Calcium/blood , Cyclooxygenase 2/genetics , Diet, High-Fat , Dinoprostone/metabolism , Estradiol/metabolism , Estrogens/metabolism , Estrone/metabolism , Female , Humans , Leptin/blood , MCF-7 Cells , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/complications , Obesity/pathology , Ovariectomy , RNA, Messenger/metabolism , Tumor Burden , Vitamin D/blood
18.
Endocrinology ; 157(1): 91-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26488808

ABSTRACT

Fatty acids (FAs) are a major energy source in the body. White adipose tissue (WAT) is a primary site where FAs are stored as triacylglycerols. Brown adipose tissue also stores and recruits FAs as a carbon source for uncoupled ß-oxidation during thermogenesis. The deletion of the vitamin D nuclear hormone receptor (VDR) gene in mice (VDRKO) results in a lean WAT phenotype with increased levels of expression of the brown adipose tissue marker Ucp1 in the WAT. However, the impact of vitamin D/VDR on FA composition in WAT has not been explored in detail. To address this question, we examined the FA composition of sc and visceral white adipose depots of VDRKO mice. We found that the levels of a subset of saturated and monounsaturated FAs of C18-C24 are specifically increased in the sc adipose depot in VDRKO mice. We revealed that a specific elongase enzyme (Elovl3), which has an important role in brown fat biology, is directly regulated by VDR and likely contributes to the altered FA composition in VDRKO mice. We also demonstrate that Elovl3 is regulated by vitamin D in vivo and tissue specifically in the sc WAT depot. We discovered that regulation of Elovl3 expression is mediated by ligand-dependent VDR occupancy of a negative-response element in the promoter proximal region of the Elovl3 gene. These data suggest that vitamin D/VDR tissue specifically modulates FA composition in sc WAT through direct regulation of Elovl3 expression.


Subject(s)
Acetyltransferases/metabolism , Calcitriol/metabolism , Fatty Acids/metabolism , Receptors, Calcitriol/agonists , Signal Transduction , Subcutaneous Fat/metabolism , Acetyltransferases/genetics , Animals , Calcitriol/administration & dosage , Cells, Cultured , Chromatin Immunoprecipitation , Fatty Acid Elongases , Gene Expression Regulation, Enzymologic , Genes, Reporter , Injections, Intraperitoneal , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/enzymology , Intra-Abdominal Fat/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Ligands , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Subcutaneous Fat/cytology , Subcutaneous Fat/enzymology , Vitamin D Response Element
19.
Pediatrics ; 136(5): e1377-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26459648

ABSTRACT

Cushing's syndrome (CS) in the pediatric population is challenging to diagnose and treat. Although next-generation medical therapies are emerging for adults with CS, none are currently approved or used in children. Here we describe the first use of mifepristone, a glucocorticoid receptor antagonist, to treat CS in a pediatric subject. The patient, a 14-year-old girl with an 18-month history of metastatic neuroendocrine carcinoma, suffered from fatigue, profound myopathy, irritability, and depression. She was found to have hypertension, hypokalemia, and worsening control of her preexisting type 1 diabetes. In this report, we detail our clinical evaluation that confirmed CS caused by an ectopic adrenocorticotropic hormone secreting tumor. Surgical and radiation therapies were not pursued because of her poor functional status and limited life expectancy, and medical treatment of CS was indicated for symptom relief. Mifepristone treatment provided rapid improvement in glycemic control, insulin resistance, and hypertension as well as significant diminishment of her myopathy and fatigue. Hypokalemia was managed with an oral potassium replacement and dose escalation of spironolactone; no other significant adverse effects were observed. Despite successful palliation of Cushing's signs and symptoms, the patient died of progression of her cancer. This case demonstrates the safety and efficacy of mifepristone treatment in a pediatric patient with symptomatic, ectopic CS. We conclude that, in appropriate pediatric patients with CS, glucocorticoid receptor antagonism with mifepristone should be considered to control the effects of hypercortisolism and to improve quality of life.


Subject(s)
Cushing Syndrome/drug therapy , Mifepristone/therapeutic use , Adolescent , Female , Humans
20.
Mol Cancer Ther ; 14(8): 1951-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25934710

ABSTRACT

The anticancer actions of vitamin D and its hormonally active form, calcitriol, have been extensively documented in clinical and preclinical studies. However, the mechanisms underlying these actions have not been completely elucidated. Here, we examined the effect of dietary vitamin D and calcitriol on mouse breast tumor-initiating cells (TICs, also known as cancer stem cells). We focused on MMTV-Wnt1 mammary tumors, for which markers for isolating TICs have previously been validated. We confirmed that these tumors expressed functional vitamin D receptors and estrogen receptors (ER) and exhibited calcitriol-induced molecular responses including ER downregulation. Following orthotopic implantation of MMTV-Wnt1 mammary tumor cells into mice, calcitriol injections or a vitamin D-supplemented diet caused a striking delay in tumor appearance and growth, whereas a vitamin D-deficient diet accelerated tumor appearance and growth. Calcitriol inhibited TIC tumor spheroid formation in a dose-dependent manner in primary cultures and inhibited TIC self-renewal in secondary passages. A combination of calcitriol and ionizing radiation inhibited spheroid formation more than either treatment alone. Further, calcitriol significantly decreased TIC frequency as evaluated by in vivo limiting dilution analyses. Calcitriol inhibition of TIC spheroid formation could be overcome by the overexpression of ß-catenin, suggesting that the inhibition of Wnt/ß-catenin pathway is an important mechanism mediating the TIC inhibitory activity of calcitriol in this tumor model. Our findings indicate that vitamin D compounds target breast TICs reducing tumor-initiating activity. Our data also suggest that combining vitamin D compounds with standard therapies may enhance anticancer activity and improve therapeutic outcomes.


Subject(s)
Calcitriol/pharmacology , Neoplastic Stem Cells/drug effects , Vitamin D/pharmacology , Animals , Body Weight , Calcium/blood , Cell Line, Tumor , Estrogens/metabolism , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/mortality , Mammary Neoplasms, Experimental/pathology , Mice , Neoplastic Stem Cells/metabolism , Receptors, Calcitriol/metabolism , Receptors, Estrogen/metabolism , Tumor Burden , Vitamin D/metabolism , Wnt Signaling Pathway/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...