Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Comp Med ; 71(3): 256-266, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34082858

ABSTRACT

The gray mouse lemur (Microcebus murinus, GML) is a nocturnal, arboreal, prosimian primate that is native to Madagascar. Captive breeding colonies of GMLs have been established primarily for noninvasive studies on questions related to circadian rhythms and metabolism. GMLs are increasingly considered to be a strong translational model for neurocognitive aging due to overlapping histopathologic features shared with aged humans. However, little information is available describing the clinical presentations, naturally occurring diseases, and histopathology of aged GMLs. In our colony, a 9 y-old, male, GML was euthanized after sudden onset of weakness, lethargy, and tibial fracture. Evaluation of this animal revealed widespread fibrous osteodystrophy (FOD) of the mandible, maxilla, cranium, appendicular, and vertebral bones. FOD and systemic metastatic mineralization were attributed to underlying chronic renal disease. Findings in this GML prompted periodic colony-wide serum biochemical screenings for azotemia and electrolyte abnormalities. Subsequently, 3 additional GMLs (2 females and 1 male) were euthanized due to varying clinical and serum biochemical presentations. Common to all 4 animals were FOD, chronic renal disease, uterine adenocarcinoma (females only), cataracts, and osteoarthritis. This case study highlights the concurrent clinical and histopathologic abnormalities that are relevant to use of GMLs in the expanding field of aging research.


Subject(s)
Adenocarcinoma , Cheirogaleidae , Renal Insufficiency, Chronic , Adenocarcinoma/veterinary , Aging , Animals , Circadian Rhythm , Female , Male , Mice
2.
Comp Med ; 71(2): 166-176, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33536115

ABSTRACT

The use of percutaneous cranial implants in rhesus macaques (Macaca mulatta) has long been a valuable tool for neuroscience research. However, when treating and assessing these animals, veterinarians are required to make assumptions about diagnostic results due to a lack of research into how these implants affect physiology. Microbial cultures of cranial implant sites show an abundance of colonizing bacteria, but whether these microbes affect animal health and wellbeing is poorly understood. In addition, microbial antibiotic resistance can present significant health concerns for both the animals and the researchers. To help elucidate the relationship between percutaneous cranial implants and blood parameters, complete blood cell counts and serum chemistry results were assessed on 57 nonhuman primates at our institution from September 2001 to March 2017. Generalized estimating equations were used to compare the results before and after an animal's first implant surgery. This modelling showed that cranial implants were a significant predictor of alterations in the number of neutrophils, lymphocytes, and red blood cells, and in the concentration of hemoglobin, alkaline phosphatase, creatinine, calcium, phos- phorus, total protein, albumin, and globulin. Anaerobic and aerobic bacterial cultures were performed to identify bacteria associated with cranial implants. Staphylococcus spp., Streptococcus spp., and Corynebacterium spp. comprised the majority of the aerobic bacterial isolates, while Fusobacterium spp., Peptostreptococcus spp. and Bacterioides fragilis comprised the majority of anaerobic bacterial isolates. Using a Pearson r correlation for statistical analysis, we assessed whether any of these bacterial isolates developed antibiotic resistances over time. Cefazolin, the most frequently used antibiotic in monkeys in this study, was the only antimicrobial out of 41 agents tested to which bacteria developed resistance over time. These results indicate that percutaneous implants are associated with a generalized inflammatory state, multiple bacterial species are present at the implant site, and these bacteria may contribute to the inflammatory response.


Subject(s)
Hematology , Prostheses and Implants , Animals , Macaca mulatta , Staphylococcus , Streptococcus
3.
JCI Insight ; 2(17)2017 09 07.
Article in English | MEDLINE | ID: mdl-28878134

ABSTRACT

Advanced breast cancer is frequently associated with skeletal metastases and accelerated bone loss. Recombinant parathyroid hormone [teriparatide, PTH(1-34)] is the first anabolic agent approved in the US for treatment of osteoporosis. While signaling through the PTH receptor in the osteoblast lineage regulates bone marrow hematopoietic niches, the effects of anabolic PTH on the skeletal metastatic niche are unknown. Here, we demonstrate, using orthotopic and intratibial models of 4T1 murine and MDA-MB-231 human breast cancer tumors, that anabolic PTH decreases both tumor engraftment and the incidence of spontaneous skeletal metastasis in mice. Microcomputed tomography and histomorphometric analyses revealed that PTH increases bone volume and reduces tumor engraftment and volume. Transwell migration assays with murine and human breast cancer cells revealed that PTH alters the gene expression profile of the metastatic niche, in particular VCAM-1, to inhibit recruitment of cancer cells. While PTH did not affect growth or migration of the primary tumor, it elicited several changes in the tumor gene expression profile resulting in a less metastatic phenotype. In conclusion, PTH treatment in mice alters the bone microenvironment, resulting in decreased cancer cell engraftment, reduced incidence of metastases, preservation of bone microarchitecture and prolonged survival.


Subject(s)
Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Parathyroid Hormone/therapeutic use , Animals , Bone Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Cell Line, Tumor , Cellular Microenvironment , Female , Gene Expression Profiling , Heterografts , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mice , Splenic Neoplasms/secondary , Survival Analysis , X-Ray Microtomography
4.
Genetics ; 206(2): 651-664, 2017 06.
Article in English | MEDLINE | ID: mdl-28592502

ABSTRACT

Systematic genetic studies of a handful of diverse organisms over the past 50 years have transformed our understanding of biology. However, many aspects of primate biology, behavior, and disease are absent or poorly modeled in any of the current genetic model organisms including mice. We surveyed the animal kingdom to find other animals with advantages similar to mice that might better exemplify primate biology, and identified mouse lemurs (Microcebus spp.) as the outstanding candidate. Mouse lemurs are prosimian primates, roughly half the genetic distance between mice and humans. They are the smallest, fastest developing, and among the most prolific and abundant primates in the world, distributed throughout the island of Madagascar, many in separate breeding populations due to habitat destruction. Their physiology, behavior, and phylogeny have been studied for decades in laboratory colonies in Europe and in field studies in Malagasy rainforests, and a high quality reference genome sequence has recently been completed. To initiate a classical genetic approach, we developed a deep phenotyping protocol and have screened hundreds of laboratory and wild mouse lemurs for interesting phenotypes and begun mapping the underlying mutations, in collaboration with leading mouse lemur biologists. We also seek to establish a mouse lemur gene "knockout" library by sequencing the genomes of thousands of mouse lemurs to identify null alleles in most genes from the large pool of natural genetic variants. As part of this effort, we have begun a citizen science project in which students across Madagascar explore the remarkable biology around their schools, including longitudinal studies of the local mouse lemurs. We hope this work spawns a new model organism and cultivates a deep genetic understanding of primate biology and health. We also hope it establishes a new and ethical method of genetics that bridges biological, behavioral, medical, and conservation disciplines, while providing an example of how hands-on science education can help transform developing countries.


Subject(s)
Behavior, Animal/physiology , Cheirogaleidae/genetics , Genome , Primates/genetics , Animals , Cheirogaleidae/physiology , Genetic Variation , Humans , Models, Genetic , Phylogeny , Primates/physiology
5.
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
6.
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
7.
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
8.
J Am Assoc Lab Anim Sci ; 54(1): 25-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25651087

ABSTRACT

Cardiomyopathy is a leading cause of mortality in aging squirrel monkeys (Saimiri spp.). However, data regarding echocardiographic measures obtained from clinically healthy nonsedated squirrel monkeys have not been published, and few electrocardiographic data are available. Here we obtained echocardiographs without sedation and electrocardiographs with minimal sedation from 63 clinically healthy squirrel monkeys that ranged from 3 to 20 y in age. 2D and M-mode echocardiography were performed on nonsedated monkeys to determine the left ventricular internal diameters at systole and diastole and the ejection fraction. Electrocardiography was performed under sedation with ketamine (15 mg/kg). Parameters evaluated included heart rate; P-wave duration; lengths of the PR, QRS, and QT intervals; R-wave amplitude, and P-wave amplitude. Initial physical examination, electrocardiography, and echocardiography indicated normal cardiac function for all monkeys. The objectives of this study were to provide reference values for nonsedated echocardiography and ketamine-sedated electrocardiography of clinically normal squirrel monkeys and to determine correlates of age and sex in these values.


Subject(s)
Echocardiography/veterinary , Electrocardiography/veterinary , Saimiri/physiology , Animals , Cardiomyopathies/veterinary , Female , Heart Rate , Male , Reference Values , Systole
9.
Comp Med ; 64(5): 404-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25402181

ABSTRACT

An 10-y-old, intact male rhesus macaque (Macaca mulatta) presented for bilateral scrotal swelling and a distended abdomen. A soft mass in the left upper quadrant of the abdomen was palpated. A barium study did not reveal any gastrointestinal abnormalities. Exploratory laparotomy revealed a large (1.25 kg, 15.0 × 13.0 × 9.5 cm), red and tan, soft, circumscribed, spherical mass within the greater omentum and 10 to 20 smaller (diameter, 1 to 4 cm), soft to firm masses in the mesentery and greater omentum. The resected mass was a self-strangulating abdominal lipoma, a pedunculated neoplasm composed of white adipocytes arising from peritoneal adipose tissue undergoing secondary coagulation necrosis after strangulation of the blood supply due to twisting of the mass around the peduncle. The smaller masses were histologically consistent with simple or self-strangulating pedunculated abdominal lipomas. The macaque presented again 9 mo later with a firm, 5.0-cm mass in the midabdomen, with intestinal displacement visible on radiographs. Given this animal's medical history and questionable prognosis, euthanasia was elected. Necropsy revealed numerous, multifocal to coalescing, 1.0- to 15.0-cm, pale tan to yellow, circumscribed, soft to firm, spherical to ellipsoid, pedunculated masses that were scattered throughout the mesentery, greater omentum, lesser omentum, and serosal surfaces of the gastrointestinal tract. All of the masses were pedunculated abdominal lipomas, and most demonstrated coagulation necrosis due to self-strangulation of the blood supply. To our knowledge, this report is the first to describe abdominal lipomatosis with secondary self-strangulation of masses in a rhesus macaque.


Subject(s)
Animals, Laboratory , Lipomatosis/veterinary , Macaca mulatta , Monkey Diseases/pathology , Neoplasms/veterinary , Peritoneal Neoplasms/veterinary , Animals , Histological Techniques/veterinary , Lipomatosis/pathology , Male , Necrosis/pathology , Necrosis/veterinary , Neoplasms/blood supply , Neoplasms/pathology , Omentum/pathology , Peritoneal Neoplasms/pathology
10.
Am J Gastroenterol ; 108(8): 1344-51, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732466

ABSTRACT

OBJECTIVES: Histological examination of duodenal biopsies is the gold standard for assessing intestinal damage in celiac disease (CD). A noninvasive marker of disease status is necessary, because obtaining duodenal biopsies is invasive and not suitable for routine monitoring of CD patients. As the small intestine is a major site of cytochrome P450 3A4 (CYP3A4) activity and also the location of the celiac lesion, we investigated whether patients with active CD display abnormal pharmacokinetics of an orally administered CYP3A4 substrate, simvastatin (SV), which could potentially be used for noninvasive assessment of their small intestinal health. METHODS: Preclinical experiments were performed in CYP3A4-humanized mice to examine the feasibility of the test. Subsequently, a clinical trial was undertaken with 11 healthy volunteers, 18 newly diagnosed patients with CD, and 25 celiac patients who had followed a gluten-free diet (GFD) for more than 1 year. The maximum concentration (Cmax) of orally administered SV plus its major non-CYP3A4-derived metabolite SV acid (SV equivalent (SVeq)) was measured, and compared with clinical, histological, and serological parameters. RESULTS: In CYP3A4-humanized mice, a marked decrease in SV metabolism was observed in response to enteropathy. In the clinical setting, untreated celiac patients displayed a significantly higher SVeq Cmax (46±24 nM) compared with treated patients (21±16 nM, P<0.001) or healthy subjects (19±11 nM, P<0.005). SVeq Cmax correctly predicted the diagnosis in 16/18 untreated celiac patients, and also the recovery status of all follow-up patients that exhibited normal or near-normal biopsies (Marsh 0-2). All patients with abnormal SVeq Cmax showed a reduction in the value after 1 year of following a GFD. CONCLUSIONS: SVeq Cmax is a promising noninvasive marker for assessment of small intestinal health. Further studies are warranted to establish its clinical utility for assessing gut status of patients with CD.


Subject(s)
Celiac Disease/drug therapy , Celiac Disease/metabolism , Cytochrome P-450 CYP3A/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Intestine, Small/drug effects , Intestine, Small/metabolism , Simvastatin/pharmacokinetics , Adolescent , Adult , Animals , Biomarkers/metabolism , Diet, Gluten-Free , Feasibility Studies , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Middle Aged , ROC Curve
11.
Endocrinology ; 153(6): 2576-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22454149

ABSTRACT

1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3) or calcitriol], the hormonally active vitamin D metabolite, exhibits anticancer actions in models of breast cancer and prostate cancer. Because CYP27B1 (1α-hydroxylase), the enzyme catalyzing 1,25(OH)(2)D(3) formation in the kidney, is also expressed in extrarenal tissues, we hypothesize that dietary vitamin D(3) will be converted to 25(OH)D(3) in the body and then to 1,25(OH)(2)D(3) locally in the cancer microenvironment in which it will exert autocrine/paracrine anticancer actions. Immunocompromised mice bearing MCF-7 breast cancer xenografts showed significant tumor shrinkage (>50%) after ingestion of a vitamin D(3)-supplemented diet (5000 IU/kg) compared with a control diet (1000 IU/kg). Dietary vitamin D(3) inhibition of tumor growth was equivalent to administered calcitriol (0.025, 0.05, or 0.1 µg/mouse, three times a week). Both treatments equivalently inhibited PC-3 prostate cancer xenograft growth but to a lesser extent than the MCF-7 tumors. Calcitriol at 0.05 µg and 0.1 µg caused modest but statistically significant increases in serum calcium levels indicating that the dietary vitamin D(3) comparison was to a maximally safe calcitriol dose. Dietary vitamin D(3) did not increase serum calcium, demonstrating its safety at the concentration tested. The vitamin D(3) diet raised circulating 1,25 dihydroxyvitamin D levels and did not alter CYP27B1 mRNA in the kidney but increased it in the tumors, suggesting that extrarenal sources including the tumors contributed to the elevated circulating 1,25 dihydroxyvitamin D(3). Both calcitriol and dietary vitamin D(3) were equipotent in suppressing estrogen synthesis and signaling and other proinflammatory and growth signaling pathways. These preclinical data demonstrate the potential utility of dietary vitamin D(3) supplementation in cancer prevention and therapy.


Subject(s)
Breast Neoplasms/drug therapy , Calcitriol/pharmacology , Cholecalciferol/pharmacology , Prostatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Animals , Body Weight/drug effects , Breast Neoplasms/pathology , Calcitriol/administration & dosage , Calcium/blood , Cell Line, Tumor , Cholecalciferol/administration & dosage , Dietary Supplements , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Ovariectomy , Prostatic Neoplasms/pathology , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroid Hydroxylases/genetics , Tumor Burden/drug effects , Vitamin D3 24-Hydroxylase , Vitamins/pharmacology
12.
Horm Cancer ; 2(3): 190-202, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21686077

ABSTRACT

Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active metabolite of vitamin D, exerts many anticancer effects in breast cancer (BCa) cells. We have previously shown using cell culture models that calcitriol acts as a selective aromatase modulator (SAM) and inhibits estrogen synthesis and signaling in BCa cells. We have now examined calcitriol effects in vivo on aromatase expression, estrogen signaling, and tumor growth when used alone and in combination with aromatase inhibitors (AIs). In immunocompromised mice bearing MCF-7 xenografts, increasing doses of calcitriol exhibited significant tumor inhibitory effects (~50% to 70% decrease in tumor volume). At the suboptimal doses tested, anastrozole and letrozole also caused significant tumor shrinkage when used individually. Although the combinations of calcitriol and the AIs caused a statistically significant increase in tumor inhibition in comparison to the single agents, the cooperative interaction between these agents appeared to be minimal at the doses tested. Calcitriol decreased aromatase expression in the xenograft tumors. Importantly, calcitriol also acted as a SAM in the mouse, decreasing aromatase expression in the mammary adipose tissue, while increasing it in bone marrow cells and not altering it in the ovaries and uteri. As a result, calcitriol significantly reduced estrogen levels in the xenograft tumors and surrounding breast adipose tissue. In addition, calcitriol inhibited estrogen signaling by decreasing tumor ERα levels. Changes in tumor gene expression revealed the suppressive effects of calcitriol on inflammatory and growth signaling pathways and demonstrated cooperative interactions between calcitriol and AIs to modulate gene expression. We hypothesize that cumulatively these calcitriol actions would contribute to a beneficial effect when calcitriol is combined with an AI in the treatment of BCa.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/biosynthesis , Calcitriol/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Vitamins/pharmacology , Anastrozole , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogens/metabolism , Female , Humans , Immunohistochemistry , Letrozole , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Nude , Nitriles/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Triazoles/pharmacology , Xenograft Model Antitumor Assays
13.
Mol Cancer Res ; 6(11): 1691-701, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19010817

ABSTRACT

Gain-of-function mutations in the androgen receptor (AR) are found in prostate cancer and are implicated in the failure of hormone therapy. Most studies have emphasized the ligand-binding domain (LBD) where mutations can create promiscuous receptors, but mutations in the NH(2)-terminal transactivation domain have also been found. To assess AR alteration as a mechanism of treatment resistance, a mouse model (h/mAR-TRAMP) was used in which the murine AR coding region is replaced by human sequence and prostate cancer initiated by a transgenic oncogene. Mice received either no treatment, androgen depletion by castration, or treatment with antiandrogens, and 20 AR transcripts were sequenced per end-stage tumor. All tumors expressed several mutant alleles, although most mutations were low frequency. Some mutations that occurred multiple times within the population were differentially located dependent on treatment. Mutations in castrated or antiandrogen-treated mice were widely dispersed but with a prominent cluster in the LBD (amino acids 736-771), whereas changes in intact mice centered near the NH(2)-terminal polymorphic glutamine tract. Functional characterization of selected LBD mutant alleles showed diverse effects on AR activity, with about half of the mutations reducing transactivation in vitro. One receptor, AR-R753Q, behaved in a cell- and promoter-dependent manner, although as a germ-line mutation it causes androgen insensitivity syndrome. This suggests that alleles that are loss of function during development may still activate a subset of AR targets to become gain of function in tumorigenesis. Mutant ARs may thus use multiple mechanisms to evade cancer treatment.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Mutation , Orchiectomy , Prostatic Neoplasms/therapy , Receptors, Androgen/genetics , Animals , DNA Mutational Analysis , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Gene Knock-In Techniques/methods , Humans , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Interaction Domains and Motifs/genetics , Receptors, Androgen/physiology
14.
J Steroid Biochem Mol Biol ; 108(3-5): 230-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17936615

ABSTRACT

Androgen, acting via the androgen receptor (AR), is central to male development, differentiation and hormone-dependent diseases such as prostate cancer. AR is actively involved in the initiation of prostate cancer, the transition to androgen independence, and many mechanisms of resistance to therapy. To examine genetic variation of AR in cancer, we created mice by germ-line gene targeting in which human AR sequence replaces that of the mouse. Since shorter length of a polymorphic N-terminal glutamine (Q) tract has been linked to prostate cancer risk, we introduced alleles with 12, 21 or 48 Qs to test this association. The three "humanized" AR mouse strains (h/mAR) are normal physiologically, as well as by cellular and molecular criteria, although slight differences are detected in AR target gene expression, correlating inversely with Q tract length. However, distinct allele-dependent differences in tumorigenesis are evident when these mice are crossed to a transgenic prostate cancer model. Remarkably, Q tract variation also differentially impacts disease progression following androgen depletion. This finding emphasizes the importance of AR function in androgen-independent as well as androgen-dependent disease. These mice provide a novel genetic paradigm in which to dissect opposing functions of AR in tumor suppression versus oncogenesis.


Subject(s)
Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Amino Acid Sequence , Animals , Glutamine/genetics , Humans , Male , Mice , Mice, Transgenic , Peptides/chemistry , Peptides/genetics , Prostatic Neoplasms/metabolism
15.
Hum Mol Genet ; 17(1): 98-110, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17906287

ABSTRACT

The androgen receptor (AR) is involved in the initiation and progression of prostate cancer and its transition to androgen independence. Genetic variation in AR may contribute to disease risk and has been studied for a polymorphic N-terminal glutamine (Q) tract that shows population heterogeneity. While the length of this tract is known to affect AR in vitro, association with disease is complicated by genetic and environmental factors that have led to discordant epidemiological findings. To clarify the effect of Q tract polymorphism on prostate cancer, we created mice bearing humanized AR genes (h/mAr) varying in Q tract length. ARs with short Q tracts (12Q), which are transcriptionally more active, induce earlier disease in the transgene-induced TRAMP prostate cancer model than alleles with median (21Q) or long (48Q) tracts. Disease length varies within each genotype, with greater differentiation and AR expression in slower growing tumors. Remarkably, following androgen ablation, Q tract length has effects that are also allele-dependent and in directions opposite to those in hormone intact mice. Differences in AR activity conferred by Q tract length thus appear to direct distinct pathways of androgen-independent as well as androgen-dependent progression, and highlight substantial risk that may be associated with alterations in the androgen axis. This AR allelic series in humanized mice provides an experimental paradigm to dissect the role of AR in prostate cancer initiation and progression, to model response to treatment and to test therapies targeted specifically to the human AR.


Subject(s)
Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Peptides/chemistry , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Animals , Base Sequence , DNA Primers/genetics , Genotype , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Hormone-Dependent/etiology , Orchiectomy , Prostatic Intraepithelial Neoplasia/etiology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/etiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Androgen/metabolism
16.
Mol Endocrinol ; 20(6): 1248-60, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16601069

ABSTRACT

Polymorphism in the length of the N-terminal glutamine (Q) tract in the human androgen receptor (AR) has been implicated in affecting aspects of male health ranging from fertility to cancer. Extreme expansion of the tract underlies Kennedy disease, and in vitro the AR Q tract length correlates inversely with transactivation capacity. However, whether normal variation influences physiology or the etiology of disease has been controversial. To assess directly the functional significance of Q tract variation, we converted the mouse AR to the human sequence by germline gene targeting, introducing alleles with 12, 21, or 48 glutamines. These three "humanized" AR (h/mAR) mouse lines were grossly normal in growth, behavior, fertility, and reproductive tract morphology. Phenotypic analysis revealed traits that varied subtly with Q tract length, including body fat amount and, more notably, seminal vesicle weight. Upon molecular analysis, tissue-specific differences in AR levels and target gene expression were detected between mouse lines. In the prostate, probasin, Nkx3.1, and clusterin mRNAs trended in directions predicted for inverse correlation of Q tract length with AR activation. Remarkably, when crossed with transgenic adenocarcinoma of mouse prostate (TRAMP) mice, striking genotype-dependent differences in prostate cancer initiation and progression were revealed. This link between Q tract length and prostate cancer, likely due to differential activation of AR targets, corroborates human epidemiological studies. This h/mAR allelic series in a homogeneous mouse genetic background allows examination of numerous physiological traits for Q tract influences and provides an animal model to test novel drugs targeted specifically to human AR.


Subject(s)
Receptors, Androgen/genetics , Receptors, Androgen/physiology , Alleles , Animals , Base Sequence , DNA, Recombinant/genetics , Female , Gene Expression , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/etiology , Peptides/chemistry , Peptides/genetics , Phenotype , Prostate/anatomy & histology , Prostate/physiology , Prostatic Neoplasms/etiology , Receptors, Androgen/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Testis/anatomy & histology , Testis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...