Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
bioRxiv ; 2023 Oct 29.
Article in English | MEDLINE | ID: mdl-37961493

ABSTRACT

Haematopoietic stem cells (HSCs) reside in specialized microenvironments, also referred to as niches, and it has been widely believed that HSC numbers are determined by the niche size alone 1-5 . However, the vast excess of the number of niche cells over that of HSCs raises questions about this model. We initially established a mathematical model of niche availability and occupancy, which predicted that HSC numbers are restricted at both systemic and local levels. To address this question experimentally, we developed a femoral bone transplantation system, enabling us to increase the number of available HSC niches. We found that the addition of niches does not alter total HSC numbers in the body, regardless of whether the endogenous (host) niche is intact or defective, suggesting that HSC numbers are limited at the systemic level. Additionally, HSC numbers in transplanted wild-type femurs did not increase beyond physiological levels when HSCs were mobilized from defective endogenous niches to the periphery, indicating that HSC numbers are also constrained at the local level. Our study demonstrates that HSC numbers are not solely determined by niche availability, thereby rewriting the long-standing model for the regulation of HSC numbers.

2.
bioRxiv ; 2023 Sep 13.
Article in English | MEDLINE | ID: mdl-37745570

ABSTRACT

Vitamin D deficiency is a common deficiency worldwide, particularly among women of reproductive age. During pregnancy, it increases the risk of immune-related diseases in offspring later in life. However, exactly how the body remembers exposure to an adverse environment during development is poorly understood. Herein, we explore the effects of prenatal vitamin D deficiency on immune cell proportions in offspring using vitamin D deficient mice established by dietary manipulation. We show that prenatal vitamin D deficiency alters immune cell proportions in offspring by changing the transcriptional properties of genes downstream of vitamin D receptor signaling in hematopoietic stem and progenitor cells of both the fetus and adults. Further investigations of the associations between maternal vitamin D levels and cord blood immune cell profiles from 75 healthy pregnant women and their term babies also confirm that maternal vitamin D levels significantly affect immune cell proportions in the babies. Thus, lack of prenatal vitamin D, particularly at the time of hematopoietic stem cell migration from the liver to the bone marrow, has long-lasting effects on immune cell proportions. This highlights the importance of providing vitamin D supplementation at specific stages of pregnancy.

3.
Cancer Res ; 82(15): 2678-2691, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35919990

ABSTRACT

Radionuclide irradiators (137Cs and 60Co) are commonly used in preclinical studies ranging from cancer therapy to stem cell biology. Amidst concerns of radiological terrorism, there are institutional initiatives to replace radionuclide sources with lower energy X-ray sources. As researchers transition, questions remain regarding whether the biological effects of γ-rays may be recapitulated with orthovoltage X-rays because different energies may induce divergent biological effects. We therefore sought to compare the effects of orthovoltage X-rays with 1-mm Cu or Thoraeus filtration and 137Cs γ-rays using mouse models of acute radiation syndrome. Following whole-body irradiation, 30-day overall survival was assessed, and the lethal dose to provoke 50% mortality within 30-days (LD50) was calculated by logistic regression. LD50 doses were 6.7 Gy, 7.4 Gy, and 8.1 Gy with 1-mm Cu-filtered X-rays, Thoraeus-filtered X-rays, and 137Cs γ-rays, respectively. Comparison of bone marrow, spleen, and intestinal tissue from mice irradiated with equivalent doses indicated that injury was most severe with 1-mm Cu-filtered X-rays, which resulted in the greatest reduction in bone marrow cellularity, hematopoietic stem and progenitor populations, intestinal crypts, and OLFM4+ intestinal stem cells. Thoraeus-filtered X-rays provoked an intermediate phenotype, with 137Cs showing the least damage. This study reveals a dichotomy between physical dose and biological effect as researchers transition to orthovoltage X-rays. With decreasing energy, there is increasing hematopoietic and intestinal injury, necessitating dose reduction to achieve comparable biological effects. SIGNIFICANCE: Understanding the significance of physical dose delivered using energetically different methods of radiation treatment will aid the transition from radionuclide γ-irradiators to orthovoltage X-irradiators.


Subject(s)
Cesium Radioisotopes , Whole-Body Irradiation , Animals , Gamma Rays , Mice , X-Rays
4.
Sci Transl Med ; 14(637): eabc1600, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35320003

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease. Tumors are poorly immunogenic and immunosuppressive, preventing T cell activation in the tumor microenvironment. Here, we present a microbial-based immunotherapeutic treatment for selective delivery of an immunogenic tetanus toxoid protein (TT856-1313) into PDAC tumor cells by attenuated Listeria monocytogenes. This treatment reactivated preexisting TT-specific memory T cells to kill infected tumor cells in mice. Treatment of KrasG12D,p53R172H, Pdx1-Cre (KPC) mice with Listeria-TT resulted in TT accumulation inside tumor cells, attraction of TT-specific memory CD4 T cells to the tumor microenvironment, and production of perforin and granzyme B in tumors. Low doses of gemcitabine (GEM) increased immune effects of Listeria-TT, turning immunologically cold into hot tumors in mice. In vivo depletion of T cells from Listeria-TT + GEM-treated mice demonstrated a CD4 T cell-mediated reduction in tumor burden. CD4 T cells from TT-vaccinated mice were able to kill TT-expressing Panc-02 tumor cells in vitro. In addition, peritumoral lymph node-like structures were observed in close contact with pancreatic tumors in KPC mice treated with Listeria-TT or Listeria-TT + GEM. These structures displayed CD4 and CD8 T cells producing perforin and granzyme B. Whereas CD4 T cells efficiently infiltrated the KPC tumors, CD8 T cells did not. Listeria-TT + GEM treatment of KPC mice with advanced PDAC reduced tumor burden by 80% and metastases by 87% after treatment and increased survival by 40% compared to nontreated mice. These results suggest that Listeria-delivered recall antigens could be an alternative to neoantigen-mediated cancer immunotherapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Listeria , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Death , Disease Models, Animal , Mice , Pancreatic Neoplasms/drug therapy , Tetanus Toxoid/therapeutic use , Tumor Microenvironment
5.
Cancers (Basel) ; 11(11)2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31731687

ABSTRACT

Radiation therapy (RT) has traditionally not been widely used in the management of hepatic malignancies for fear of toxicity in the form of radiation-induced liver disease (RILD). Pre-clinical hepatic irradiation models can provide clinicians with better understanding of the radiation tolerance of the liver, which in turn may lead to the development of more effective cancer treatments. Previous models of hepatic irradiation are limited by either invasive laparotomy procedures, or the need to irradiate the whole or large parts of the liver using external skin markers. In the setting of modern-day radiation oncology, a truly translational animal model would require the ability to deliver RT to specific parts of the liver, through non-invasive image guidance methods. To this end, we developed a targeted hepatic irradiation model on the Small Animal Radiation Research Platform (SARRP) using contrast-enhanced cone-beam computed tomography image guidance. Using this model, we showed evidence of the early development of region-specific RILD through functional single photon emission computed tomography (SPECT) imaging.

6.
Int J Mol Sci ; 20(22)2019 Nov 13.
Article in English | MEDLINE | ID: mdl-31766149

ABSTRACT

Patients with KRAS mutated colorectal cancer (CRC) represent a cohort with unmet medical needs, with limited options of FDA-approved therapies. Representing 40-45% of all CRC patients, they are considered ineligible to receive anti-EGFR monoclonal antibodies that have added a significant therapeutic benefit for KRAS wild type CRC patients. Although several mouse models of CRC have been developed during the past decade, one genetically resembling the KRAS mutated CRC is yet to be established. In this study C57 BL/6 mice with truncated adenomatous polyposis coli (APC) floxed allele was crossed with heterozygous KRAS floxed outbred mice to generate an APCf/f KRAS+/f mouse colony. In another set of breeding, APC floxed mice were crossed with CDX2-Cre-ERT2 mice and selected for APCf/f CDX2-Cre-ERT2 after the second round of inbreeding. The final model of the disease was generated by the cross of the two parental colonies and viable APC f/f KRAS +/f CDX2-Cre-ERT2 (KPC: APC) were genotyped and characterized. The model animals were tamoxifen (TAM) induced to generate tumors. Micro-positron emission tomography (PET) scan was used to detect and measure tumor volume and standard uptake value (SUV). Hematoxylin and eosin (H&E) staining was performed to establish neoplasm and immunohistochemistry (IHC) was performed to determine histological similarities with human FFPE biopsies. The MSI/microsatellite stable (MSS) status was determined. Finally, the tumors were extensively characterized at the molecular level to establish similarities with human CRC tumors. The model KPC: APC animals are conditional mutants that developed colonic tumors upon induction with tamoxifen in a dose-dependent manner. The tumors were confirmed to be malignant within four weeks of induction by H&E staining and higher radioactive [18F] fluoro-2-deoxyglucose (FDG) uptake (SUV) in micro-PET scan. Furthermore, the tumors histologically and molecularly resembled human colorectal carcinoma. Post tumor generation, the KPC: APC animals died of cachexia and rectal bleeding. Implications: This model is an excellent preclinical platform to molecularly characterize the KRAS mutated colorectal tumors and discern appropriate therapeutic strategies to improve disease management and overall survival.


Subject(s)
Colorectal Neoplasms/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , CDX2 Transcription Factor/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL
7.
Oncotarget ; 8(13): 20729-20740, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28186976

ABSTRACT

Our laboratory has developed a novel delivery platform using an attenuated non-toxic and non-pathogenic bacterium Listeria monocytogenes that infects tumor cells and selectively survives and multiplies in metastases and primary tumors with help of myeloid-derived suppressor cells (MDSC) and immune suppression in the tumor microenvironment (TME). 32P was efficiently incorporated into the Listeria bacteria by starvation of the bacteria in saline, and then cultured in phosphorus-free medium complemented with 32P as a nutrient. Listeria-32P kills tumor cells through both 32P-induced ionizing radiation and Listeria-induced reactive oxygen species (ROS). The levels of 32P and Listeria were studied in various normal and tumor tissues, at sequential time points after injection of mice with pancreatic cancer (syngeneic model Panc-02). We found that 32P and Listeria predominantly accumulated in tumors and metastases, with their highest accumulation 4 hrs (32P) and 3 days (Listeria) after injection. Listeria also penetrated the transgenic KPC (conditionally express endogenous Kras-G12D and p53-R172H mutant alleles) pancreatic tumors and metastases. This is remarkable since KPC tumors, like human tumors, exhibit a stromal barrier, which prevents most drugs from penetrating the pancreatic tumors. Therapeutic treatment with Listeria -32P resulted in a strong reduction of the growth of pancreatic cancer at early and late stages in Panc-02 and KPC mice. These results highlight the power of Listeria as new delivery platform of anticancer agents to the TME. Not only were therapeutic levels of radioactive Listeria reached in tumors and metastases but the selective delivery also led to minimal side effects.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Listeria monocytogenes , Pancreatic Neoplasms/pathology , Phosphorus Radioisotopes/administration & dosage , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Microscopy, Confocal
8.
PLoS Negl Trop Dis ; 8(10): e3118, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25275627

ABSTRACT

BACKGROUND: Trypanosoma cruzi, the causative agent of Chagas disease, has high affinity for lipoproteins and adipose tissue. Infection results in myocarditis, fat loss and alterations in lipid homeostasis. This study was aimed at analyzing the effect of high fat diet (HFD) on regulating acute T. cruzi infection-induced myocarditis and to evaluate the effect of HFD on lipid metabolism in adipose tissue and heart during acute T. cruzi infection. METHODOLOGY/PRINCIPAL FINDINGS: CD1 mice were infected with T. cruzi (Brazil strain) and fed either a regular control diet (RD) or HFD for 35 days following infection. Serum lipid profile, tissue cholesterol levels, blood parasitemia, and tissue parasite load were analyzed to evaluate the effect of diet on infection. MicroPET and MRI analysis were performed to examine the morphological and functional status of the heart during acute infection. qPCR and immunoblot analysis were carried out to analyze the effect of diet on the genes involved in the host lipid metabolism during infection. Oil red O staining of the adipose tissue demonstrated reduced lipolysis in HFD compared to RD fed mice. HFD reduced mortality, parasitemia and cardiac parasite load, but increased parasite load in adipocytes. HFD decreased lipolysis during acute infection. Both qPCR and protein analysis demonstrated alterations in lipid metabolic pathways in adipose tissue and heart in RD fed mice, which were further modulated by HFD. Both microPET and MRI analyses demonstrated changes in infected RD murine hearts which were ameliorated by HFD. CONCLUSION/SIGNIFICANCE: These studies indicate that Chagasic cardiomyopathy is associated with a cardiac lipidpathy and that both cardiac lipotoxicity and adipose tissue play a role in the pathogenesis of Chagas disease. HFD protected mice from T. cruzi infection-induced myocardial damage most likely due to the effects of HFD on both adipogenesis and T. cruzi infection-induced cardiac lipidopathy.


Subject(s)
Chagas Cardiomyopathy/metabolism , Myocarditis/metabolism , Adipogenesis , Adipose Tissue, White/metabolism , Animals , Brazil , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/pathology , Cholesterol, LDL/blood , Diet, High-Fat , Lipid Metabolism , Male , Mice , Mice, Inbred C3H , Myocarditis/parasitology , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology
9.
Expert Rev Anticancer Ther ; 14(10): 1243-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25156106

ABSTRACT

BACKGROUND: Novel approaches to treatment of pancreatic cancer (PCa) are urgently needed. A chimeric monoclonal antibody (mAb) chTNT3 binds to single-strand DNA (ssDNA) and RNA released from the non-viable cells in fast growing tumors. Here the authors investigated whether radioimmunotherapy (RIT) using chTNT3 mAb radiolabeled with 213-Bismuth ((213)Bi) could be effective in treatment of experimental PCa. METHODS: Two human PCa cell lines, Panc1 and MiaPaCa-2, were used for in vitro experiments. The xenografts in mice were established using MiaPaCa-2 cells. Therapy compared (213)Bi-chTNT3 (700 µCi) to gemcitabine or cisplatin, untreated controls and 'cold' chTNT3. RESULTS: RIT abrogated the tumors growth while tumors in control groups grew aggressively. Chemotherapy was less effective than RIT and toxic to mice while RIT did not have any side effects. CONCLUSIONS: RIT with (213)Bi-chTNT3 was safe and effective in the treatment of experimental PCa in comparison with chemotherapy. This makes α-RIT targeting ssDNA a promising modality for further development.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Bismuth/administration & dosage , Pancreatic Neoplasms/therapy , Radioimmunotherapy/methods , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/adverse effects , Cisplatin/therapeutic use , DNA/metabolism , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/pathology , Radioimmunotherapy/adverse effects , Radioisotopes/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine
10.
Semin Nucl Med ; 44(1): 57-65, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24314046

ABSTRACT

The scientific study of living animals may be dated to Aristotle's original dissections, but modern animal studies are perhaps a century in the making, and advanced animal imaging has emerged only during the past few decades. In vivo imaging now occupies a growing role in the scientific research paradigm. Imaging of small animals has been particularly useful to help understand human molecular biology and pathophysiology using rodents, especially using genetically engineered mice (GEM) with spontaneous diseases that closely mimic human diseases. Specific examples of GEM models of veterinary diseases exist, but in general, GEM for veterinary research has lagged behind human research applications. However, the development of spontaneous disease models from GEM may also hold potential for veterinary research. The imaging techniques most widely used in small-animal research are CT, PET, single-photon emission CT, MRI, and optical fluorescent and luminescent imaging.


Subject(s)
Body Size , Diagnostic Imaging/veterinary , Research/instrumentation , Animal Husbandry , Animals , Aquatic Organisms , Diagnostic Imaging/instrumentation , Disease Models, Animal , Humans , Radioactive Tracers
11.
Glycobiology ; 23(12): 1477-90, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24037315

ABSTRACT

Bisected, complex N-glycans on glycoproteins are generated by the glycosyltransferase MGAT3 and cause reduced cell surface binding of galectins. Previously, we showed that MGAT3 reduces growth factor signaling and retards mammary tumor progression driven by the Polyoma middle T antigen (PyMT) expressed in mammary epithelium under the mouse mammary tumor virus (MMTV) promoter. However, the penetrance of the tumor phenotype became variable in mixed FVB/N and C57BL/6 female mice and we therefore investigated a congenic C57BL/6 Mgat3(-/-)/MMTV-PyMT model. In the absence of MGAT3, C57BL/6 Mgat3(-/-)/MMTV-PyMT females exhibited accelerated tumor appearance and increased tumor burden, glucose uptake in tumors and lung metastasis. Nevertheless, activation of extracellular signal-regulated kinase (ERK)1/2 or protein kinase B (AKT) was reduced in ∼20-week C57BL/6 MMTV-PyMT tumors lacking MGAT3. Activation of focal adhesion kinase (FAK), protein tyrosine kinase Src, and p38 mitogen-activated protein kinase were similar to that of controls. All the eight mouse galectin genes were expressed in mammary tumors and tumor epithelial cells (TECs), but galectin-2 and -12 were not detected by western analysis in tumors, and galectin-7 was not detected in 60% of the TEC lines. From microarray data reported for human breast cancers, at least 10 galectin and 7 N-glycan N-acetylglucosaminyl (GlcNAc)-transferase (MGAT) genes are expressed in tumor tissue, and expression often varies significantly between different breast cancer subtypes. Thus, in summary, while MGAT3 and bisected complex N-glycans retard mouse mammary tumor progression, genetic background may modify this effect; identification of key galectins that promote mammary tumor progression in mice is not straightforward because all the eight galectin genes are expressed; and high levels of MGAT3, galectin-4, -8, -10, -13 and -14 transcripts correlate with better relapse-free survival in human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Disease Progression , Galectins/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Polysaccharides/metabolism , Animals , Antigens, Polyomavirus Transforming/metabolism , Breast Neoplasms/genetics , Disease Models, Animal , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/metabolism , Mice , Mice, Inbred Strains , N-Acetylglucosaminyltransferases/deficiency , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism
12.
Am J Pathol ; 182(2): 319-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23219729

ABSTRACT

For the greater part of the last century, basic science research has been limited to in vitro studies of cellular processes and ex vivo tissue examination from suitable animal models of disease. In the last three decades, however, new techniques have been developed that permit the imaging of live animals using X-rays, radiotracer emissions, magnetic resonance signals, sound waves and optical fluorescence, and bioluminescence. The objective of this review is to provide a broad overview of common animal imaging modalities, with a focus on positron emission tomography (PET), single photon emission computed tomography (SPECT), and computed tomography (CT). Important examples, benefits, and limits of microPET/SPECT/CT technologies in current use, and their central role in improving our understanding of biological behavior and in facilitating the development of treatments from bench to bedside are included.


Subject(s)
Disease Models, Animal , Positron-Emission Tomography/methods , Tomography, Emission-Computed, Single-Photon/methods , Tomography, X-Ray Computed/methods , Animals , Humans
13.
PLoS Negl Trop Dis ; 6(12): e1971, 2012.
Article in English | MEDLINE | ID: mdl-23272265

ABSTRACT

BACKGROUND: Chagas disease, resulting from infection with the parasite Trypanosoma cruzi (T. cruzi), is a major cause of cardiomyopathy in Latin America. Drug therapy for acute and chronic disease is limited. Stem cell therapy with bone marrow mesenchymal cells (MSCs) has emerged as a novel therapeutic option for cell death-related heart diseases, but efficacy of MSC has not been tested in Chagas disease. METHODS AND RESULTS: We now report the use of cell-tracking strategies with nanoparticle labeled MSC to investigate migration of transplanted MSC in a murine model of Chagas disease, and correlate MSC biodistribution with glucose metabolism and morphology of heart in chagasic mice by small animal positron emission tomography (microPET). Mice were infected intraperitoneally with trypomastigotes of the Brazil strain of T. cruzi and treated by tail vein injection with MSC one month after infection. MSCs were labeled with near infrared fluorescent nanoparticles and tracked by an in vivo imaging system (IVIS). Our IVIS results two days after transplant revealed that a small, but significant, number of cells migrated to chagasic hearts when compared with control animals, whereas the vast majority of labeled MSC migrated to liver, lungs and spleen. Additionally, the microPET technique demonstrated that therapy with MSC reduced right ventricular dilation, a phenotype of the chagasic mouse model. CONCLUSIONS: We conclude that the beneficial effects of MSC therapy in chagasic mice arise from an indirect action of the cells in the heart rather than a direct action due to incorporation of large numbers of transplanted MSC into working myocardium.


Subject(s)
Bone Marrow Transplantation/methods , Chagas Cardiomyopathy/therapy , Animals , Biological Availability , Bone Marrow Cells/cytology , Chagas Cardiomyopathy/pathology , Disease Models, Animal , Liver/pathology , Lung/pathology , Male , Mice , Myocardium/pathology , Spleen/pathology , Staining and Labeling
14.
Surgery ; 152(6): 1068-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23102680

ABSTRACT

BACKGROUND: Pasireotide (SOM230), a long-acting somatostatin analogue (LAR), has improved agonist activity at somatostatin receptors. We tested the effect of SOM230 on insulin secretion, serum glucose concentrations, tumor growth, and survival using an MEN1 transgenic mouse model. METHODS: Eight 12-month-old conditional Men1 knockout mice with insulinoma were assessed. The treatment (n = 4) and control groups (n = 4) received monthly subcutaneous injections of SOM230 or PBS. Serum insulin and glucose levels were determined by enzyme-linked immunosorbent assay and enzymatic colorimetric assay, respectively. Tumor activity, growth, and apoptosis were determined by microPET/CT scan and histologic analysis. RESULTS: On day 7, there was a decrease in serum insulin levels from 1.06 ± 0.28 µg/L to 0.37 ± 0.17 µg/L (P = .0128) and a significant increase in serum glucose from 4.2 ± 0.45 mmol/L to 7.12 ± 1.06 mmol/L (P = .0075) in the treatment group but no change in the control group. Tumor size was less in the treatment group (2,098 ± 388 µm(2)) compared with the control group (7,067 ± 955 µm(2); P = .0024). Furthermore, apoptosis was increased in the treatment group (6.9 ± 1.23%) compared with the control group (0.29 ± 0.103%; P = .002). CONCLUSION: SOM230 demonstrates antisecretory, antiproliferative, and proapoptotic activity in our MEN1 model of insulinoma. Further studies of the effects of SOM230 in PNET patients with MEN1 mutations are warranted.


Subject(s)
Multiple Endocrine Neoplasia Type 1/drug therapy , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Somatostatin/analogs & derivatives , Animals , Apoptosis , Blood Glucose/analysis , Insulin/blood , Mice , Mice, Knockout , Multimodal Imaging , Multiple Endocrine Neoplasia Type 1/genetics , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Positron-Emission Tomography , Somatostatin/therapeutic use , Tomography, X-Ray Computed
15.
FASEB J ; 26(8): 3252-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22549510

ABSTRACT

The KCNQ1 α subunit and the KCNE2 ß subunit form a potassium channel in thyroid epithelial cells. Genetic disruption of KCNQ1-KCNE2 causes hypothyroidism in mice, resulting in cardiac hypertrophy, dwarfism, alopecia, and prenatal mortality. Here, we investigated the mechanistic requirement for KCNQ1-KCNE2 in thyroid hormone biosynthesis, utilizing whole-animal dynamic positron emission tomography. The KCNQ1-specific antagonist (-)-[3R,4S]-chromanol 293B (C293B) significantly impaired thyroid cell I(-) uptake, which is mediated by the Na(+)/I(-) symporter (NIS), in vivo (dSUV/dt: vehicle, 0.028 ± 0.004 min(-1); 10 mg/kg C293B, 0.009 ± 0.006 min(-1)) and in vitro (EC(50): 99 ± 10 µM C293B). Na(+)-dependent nicotinate uptake by SMCT, however, was unaffected. Kcne2 deletion did not alter the balance of free vs. thyroglobulin-bound I(-) in the thyroid (distinguished using ClO(4)(-), a competitive inhibitor of NIS), indicating that KCNQ1-KCNE2 is not required for Duox/TPO-mediated I(-) organification. However, Kcne2 deletion doubled the rate of free I(-) efflux from the thyroid following ClO(4)(-) injection, a NIS-independent process. Thus, KCNQ1-KCNE2 is necessary for adequate thyroid cell I(-) uptake, the most likely explanation being that it is prerequisite for adequate NIS activity.


Subject(s)
Iodides/metabolism , KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Thyroid Gland/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Hypothyroidism/genetics , KCNQ1 Potassium Channel/antagonists & inhibitors , KCNQ1 Potassium Channel/genetics , Mice , Positron-Emission Tomography , Potassium Channels, Voltage-Gated/genetics , Symporters/antagonists & inhibitors , Symporters/metabolism , Thyroid Gland/drug effects
16.
Semin Nucl Med ; 41(3): 151-65, 2011 May.
Article in English | MEDLINE | ID: mdl-21440693

ABSTRACT

Imaging devices for small animals have emerged in the past 10 years as extraordinarily useful tools in translational research and drug development. The Food and Drug Administration requires animal testing after in vitro drug discovery but before human application. Many small animal instruments have been developed in analogy to human scale devices, including positron emission tomography, single-photon emission computed tomography, computed tomography, magnetic resonance imaging, and ultrasound. Conversely, optical imaging with fluorescent and bioluminescent tracer technology, originating in single-cell in vitro studies, has been scaled up to whole-body animal imaging. Imaging that uses multiple devices permits a comparison of different aspects of function, anatomy, gene expression, and phenotype by the use of software algorithms or more recently with hybrid instruments. Animal imaging facilitates "bench-to-bedside" drug development in 2 ways. Longitudinal imaging improves the science of animal research through the benefit of paired statistics with the use of animals as their own controls while it simultaneously reduces animal sacrifice. In addition, imaging makes explicit the development of diagnostic and therapeutic agents on nearly identical molecular synthesis platforms, therefore linking drug discovery to the development of imaging tracers. This powerful paradigm, now known as diagnostic/therapeutic pairing or theranostics, is already familiar from the use of (123)I used for thyroid diagnosis and (131)I for therapy of benign and malignant thyroid conditions. Many newer examples exist, such as "cold" or "hot" octreotide and meta-iodobenzylguanidine in neuroendocrine tumors; and rituximab in pharmaceutical doses, or with beta emitter tags, for therapy of indolent non-Hodgkin's lymphoma. Theranostic agents are also rapidly emerging that use nanoparticles, aptamers, peptides, and antibodies for magnetic resonance imaging/positron emission tomography/single-photo emission computed tomography/computed tomography imaging devices in animals with subsequent therapeutic drug development for translation to human use.


Subject(s)
Diagnostic Imaging/instrumentation , Models, Animal , Animal Welfare , Animals , Body Size , Humans , Optical Phenomena , Radioactive Tracers , Systems Integration , Therapeutics
17.
PLoS One ; 6(2): e16959, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21347238

ABSTRACT

Chagas disease, caused by infection with Trypanosoma cruzi, is an important cause of cardiovascular disease. It is increasingly clear that parasite-derived prostaglandins potently modulate host response and disease progression. Here, we report that treatment of experimental T. cruzi infection (Brazil strain) beginning 5 days post infection (dpi) with aspirin (ASA) increased mortality (2-fold) and parasitemia (12-fold). However, there were no differences regarding histopathology or cardiac structure or function. Delayed treatment with ASA (20 mg/kg) beginning 60 dpi did not increase parasitemia or mortality but improved ejection fraction. ASA treatment diminished the profile of parasite- and host-derived circulating prostaglandins in infected mice. To distinguish the effects of ASA on the parasite and host bio-synthetic pathways we infected cyclooxygenase-1 (COX-1) null mice with the Brazil-strain of T. cruzi. Infected COX-1 null mice displayed a reduction in circulating levels of thromboxane (TX)A(2) and prostaglandin (PG)F(2α). Parasitemia was increased in COX-1 null mice compared with parasitemia and mortality in ASA-treated infected mice indicating the effects of ASA on mortality potentially had little to do with inhibition of prostaglandin metabolism. Expression of SOCS-2 was enhanced, and TRAF6 and TNFα reduced, in the spleens of infected ASA-treated mice. Ablation of the initial innate response to infection may cause the increased mortality in ASA-treated mice as the host likely succumbs more quickly without the initiation of the "cytokine storm" during acute infection. We conclude that ASA, through both COX inhibition and other "off-target" effects, modulates the progression of acute and chronic Chagas disease. Thus, eicosanoids present during acute infection may act as immunomodulators aiding the transition to and maintenance of the chronic phase of the disease. A deeper understanding of the mechanism of ASA action may provide clues to the differences between host response in the acute and chronic T. cruzi infection.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Chagas Disease/drug therapy , Trypanosoma cruzi/pathogenicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/therapeutic use , Chagas Disease/metabolism , Chagas Disease/parasitology , Chagas Disease/physiopathology , Chronic Disease , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Cytokines/metabolism , Eicosanoids/biosynthesis , Gene Deletion , Male , Mice , Parasitemia/drug therapy , Parasitemia/metabolism , Parasitemia/parasitology , Parasitemia/physiopathology , Stroke Volume/drug effects , Thromboxane-A Synthase/deficiency , Thromboxane-A Synthase/genetics , Time Factors , Trypanosoma cruzi/drug effects
18.
Proc Natl Acad Sci U S A ; 108(1): 319-24, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21173237

ABSTRACT

Rhabdoid tumors (RTs) are rare, highly aggressive pediatric malignancies with poor prognosis and with no standard or effective treatment strategies. RTs are characterized by biallelic inactivation of the INI1 tumor suppressor gene. INI1 directly represses CCND1 and activates cyclin-dependent kinase (cdk) inhibitors p16(Ink4a) and p21(CIP). RTs are exquisitely dependent on cyclin D1 for genesis and survival. To facilitate translation of unique therapeutic strategies, we have used genetically engineered, Ini1(+/-) mice for therapeutic testing. We found that PET can be used to noninvasively and accurately detect primary tumors in Ini1(+/-) mice. In a PET-guided longitudinal study, we found that treating Ini1(+/-) mice bearing primary tumors with the pan-cdk inhibitor flavopiridol resulted in complete and stable regression of some tumors. Other tumors showed resistance to flavopiridol, and one of the resistant tumors overexpressed cyclin D1, more than flavopiridol-sensitive cells. The concentration of flavopiridol used was not sufficient to down-modulate the high level of cyclin D1 and failed to induce cell death in the resistant cells. Furthermore, FISH and PCR analyses indicated that there is aneuploidy and increased CCND1 copy number in resistant cells. These studies indicate that resistance to flavopiridol may be correlated to elevated cyclin D1 levels. Our studies also indicate that Ini1(+/-) mice are valuable tools for testing unique therapeutic strategies and for understanding mechanisms of drug resistance in tumors that arise owing to loss of Ini1, which is essential for developing effective treatment strategies against these aggressive tumors.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Cyclin D1/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Rhabdoid Tumor/drug therapy , Animals , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Primers/genetics , Flavonoids/therapeutic use , Gene Silencing , Histological Techniques , Immunoblotting , Immunohistochemistry , In Situ Hybridization, Fluorescence , Longitudinal Studies , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Piperidines/therapeutic use , Polymerase Chain Reaction , Positron-Emission Tomography , Rhabdoid Tumor/genetics , Rhabdoid Tumor/ultrastructure , SMARCB1 Protein
19.
PLoS Negl Trop Dis ; 4(10): e853, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20976055

ABSTRACT

Scedosporium apiospermum is part of the Pseudallescheria-Scedosporium complex. Peptidorhamnomannans (PRMs) are cell wall glycopeptides present in some fungi, and their structures have been characterized in S. apiospermum, S. prolificans and Sporothrix schenckii. Prior work shows that PRMs can interact with host cells and that the glycopeptides are antigenic. In the present study, three monoclonal antibodies (mAbs, IgG1) to S. apiospermum derived PRM were generated and their effects on S. apiospermum were examined in vitro and in vivo. The mAbs recognized a carbohydrate epitope on PRM. In culture, addition of the PRM mAbs increased S. apiospermum conidia germination and reduced conidial phagocytosis by J774.16 macrophages. In a murine infection model, mice treated with antibodies to PRM died prior to control animals. Thus, PRM is involved in morphogenesis and the binding of this glycopeptide by mAbs enhanced the virulence of the fungus. Further insights into the effects of these glycopeptides on the pathobiology of S. apiospermum may lead to new avenues for preventing and treating scedosporiosis.


Subject(s)
Antibodies, Fungal/immunology , Antibodies, Monoclonal/immunology , Antibody-Dependent Enhancement , Glycoproteins/immunology , Mycoses/microbiology , Mycoses/mortality , Scedosporium/pathogenicity , Animals , Antibodies, Fungal/isolation & purification , Antibodies, Monoclonal/isolation & purification , Cell Line , Disease Models, Animal , Epitopes/immunology , Female , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mycoses/immunology , Phagocytosis , Scedosporium/growth & development , Scedosporium/immunology , Survival Analysis
20.
Am J Trop Med Hyg ; 81(5): 900-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19861629

ABSTRACT

Noninvasive assessment of cardiac structure and function is essential to understand the natural course of murine infection with Trypanosoma cruzi. Magnetic resonance imaging (MRI) and echocardiography have been used to monitor anatomy and function; positron emission tomography (PET) is ideal for monitoring metabolic events in the myocardium. Mice infected with T. cruzi (Brazil strain) were imaged 15-100 days post infection (dpi). Quantitative (18)F-FDG microPET imaging, MRI and echocardiography were performed and compared. Tracer ((18)F-FDG) uptake was significantly higher in infected mice at all days of infection, from 15 to 100 dpi. Dilatation of the right ventricular chamber was observed by MRI from 30 to 100 dpi in infected mice. Echocardiography revealed significantly reduced ejection fraction by 60 dpi. Combination of these three complementary imaging modalities makes it possible to noninvasively quantify cardiovascular function, morphology, and metabolism from the earliest days of infection through the chronic phase.


Subject(s)
Chagas Cardiomyopathy/pathology , Positron-Emission Tomography/methods , Animals , Echocardiography , Humans , Male , Mice , Myocardium/metabolism , Myocardium/pathology , Staining and Labeling , Trypanosoma cruzi
SELECTION OF CITATIONS
SEARCH DETAIL
...