Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Pharmaceutics ; 14(7)2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35890398

ABSTRACT

Opening the blood brain barrier (BBB) under imaging guidance may be useful for the treatment of many brain disorders. Rapidly applied magnetic fields have the potential to generate electric fields in brain tissue that, if properly timed, may enable safe and effective BBB opening. By tuning magnetic pulses generated by a novel electropermanent magnet (EPM) array, we demonstrate the opening of tight junctions in a BBB model culture in vitro, and show that induced monophasic electrical pulses are more effective than biphasic ones. We confirmed, with in vivo contrast-enhanced MRI, that the BBB can be opened with monophasic pulses. As electropermanent magnets have demonstrated efficacy at tuning B0 fields for magnetic resonance imaging studies, our results suggest the possibility of implementing an EPM-based hybrid theragnostic device that could both image the brain and enhance drug transport across the BBB in a single sitting.

2.
IEEE Open J Eng Med Biol ; 1: 265-267, 2020.
Article in English | MEDLINE | ID: mdl-33748768

ABSTRACT

GOAL: To develop a micron-scale device that can operate as an MRI-based reporter for the presence of SARS-CoV-2 virus. METHODS: Iron rod microdevices were constructed via template-guided synthesis and suspended in phosphate buffered saline (PBS). Heat-inactivated SARS-CoV-2 viruses were added to the samples and imaged with low-field MRI. RESULTS: MRI of microdevices and viruses showed decreased signal intensity at low concentrations of viruses that recovered at higher concentrations. Electron micrographs suggest that reduced MRI intensity may be due to concentration-dependent shielding of water protons from local magnetic inhomogeneities caused by the iron microdevices. CONCLUSIONS: The preliminary results presented in this letter provide justification for further studies exploring the potential diagnostic role of magnetic microdevices in assessing the presence and concentration of SARS-CoV-2 viruses.

3.
Mol Cancer Res ; 17(9): 1815-1827, 2019 09.
Article in English | MEDLINE | ID: mdl-31164413

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with limited and, very often, ineffective medical and surgical therapeutic options. The treatment of patients with advanced unresectable PDAC is restricted to systemic chemotherapy, a therapeutic intervention to which most eventually develop resistance. Recently, nab-paclitaxel (n-PTX) has been added to the arsenal of first-line therapies, and the combination of gemcitabine and n-PTX has modestly prolonged median overall survival. However, patients almost invariably succumb to the disease, and little is known about the mechanisms underlying n-PTX resistance. Using the conditionally reprogrammed (CR) cell approach, we established and verified continuously growing cell cultures from treatment-naïve patients with PDAC. To study the mechanisms of primary drug resistance, nab-paclitaxel-resistant (n-PTX-R) cells were generated from primary cultures and drug resistance was verified in vivo, both in zebrafish and in athymic nude mouse xenograft models. Molecular analyses identified the sustained induction of c-MYC in the n-PTX-R cells. Depletion of c-MYC restored n-PTX sensitivity, as did treatment with either the MEK inhibitor, trametinib, or a small-molecule activator of protein phosphatase 2a. IMPLICATIONS: The strategies we have devised, including the patient-derived primary cells and the unique, drug-resistant isogenic cells, are rapid and easily applied in vitro and in vivo platforms to better understand the mechanisms of drug resistance and for defining effective therapeutic options on a patient by patient basis.


Subject(s)
Albumins/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Drug Resistance, Neoplasm , Paclitaxel/pharmacology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Up-Regulation , Aged , Aged, 80 and over , Albumins/therapeutic use , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Transplantation , Paclitaxel/therapeutic use , Pancreatic Neoplasms/drug therapy , Primary Cell Culture , Tumor Cells, Cultured , Zebrafish , Pancreatic Neoplasms
4.
J Neurol Neurosurg Psychiatry ; 89(10): 1071-1081, 2018 10.
Article in English | MEDLINE | ID: mdl-29735511

ABSTRACT

BACKGROUND AND OBJECTIVE: Dysferlinopathies are a group of muscle disorders caused by mutations in the DYSF gene. Previous muscle imaging studies describe a selective pattern of muscle involvement in smaller patient cohorts, but a large imaging study across the entire spectrum of the dysferlinopathies had not been performed and previous imaging findings were not correlated with functional tests. METHODS: We present cross-sectional T1-weighted muscle MRI data from 182 patients with genetically confirmed dysferlinopathies. We have analysed the pattern of muscles involved in the disease using hierarchical analysis and presented it as heatmaps. Results of the MRI scans have been correlated with relevant functional tests for each region of the body analysed. RESULTS: In 181 of the 182 patients scanned, we observed muscle pathology on T1-weighted images, with the gastrocnemius medialis and the soleus being the most commonly affected muscles. A similar pattern of involvement was identified in most patients regardless of their clinical presentation. Increased muscle pathology on MRI correlated positively with disease duration and functional impairment. CONCLUSIONS: The information generated by this study is of high diagnostic value and important for clinical trial development. We have been able to describe a pattern that can be considered as characteristic of dysferlinopathy. We have defined the natural history of the disease from a radiological point of view. These results enabled the identification of the most relevant regions of interest for quantitative MRI in longitudinal studies, such as clinical trials. CLINICAL TRIAL REGISTRATION: NCT01676077.


Subject(s)
Muscle, Skeletal/diagnostic imaging , Muscular Dystrophies, Limb-Girdle/diagnostic imaging , Adult , Cross-Sectional Studies , Female , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Male , Middle Aged
5.
J Am Coll Radiol ; 15(2): 313-318, 2018 02.
Article in English | MEDLINE | ID: mdl-29128502

ABSTRACT

PURPOSE: To evaluate defect detection in radiation protective apparel, typically called lead aprons, using infrared (IR) thermal imaging. The use of IR lighting eliminates the need for access to x-ray-emitting equipment and radiation dose to the inspector. MATERIALS AND METHODS: The performance of radiation workers was prospectively assessed using both a tactile inspection and the IR inspection with a lead apron phantom over a 2-month period. The phantom was a modified lead apron with a series of nine holes of increasing diameter ranging from 2 to 35 mm in accordance with typical rejection criteria. Using the tactile method, a radiation worker would feel for the defects in the lead apron. For the IR inspection, a 250-W IR light source was used to illuminate the lead apron phantom; an IR camera detected the transmitted radiation. The radiation workers evaluated two stills from the IR camera. RESULTS: From the 31 participants inspecting the lead apron phantom with the tactile method, only 2 participants (6%) correctly discovered all 9 holes and 1 participant reported a defect that was not there; 10 of the 20 participants (50%) correctly identified all 9 holes using the IR method. Using a weighted average, 5.4 defects were detected with the tactile method and 7.5 defects were detected with the IR method. CONCLUSION: IR light can penetrate an apron's protective outer fabric and illuminate defects below the current standard rejection size criteria. The IR method improves defect detectability as compared with the tactile method.


Subject(s)
Equipment Failure Analysis/methods , Occupational Exposure/prevention & control , Protective Clothing/standards , Radiation Protection/instrumentation , Humans , Infrared Rays , Lead , Phantoms, Imaging , Prospective Studies
6.
Neuroradiol J ; 30(5): 418-424, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28195509

ABSTRACT

Background and purpose Enlargement of the subarachnoid spaces in infancy (ESSI) is a common cause of macrocephaly without proven explanation. We have observed subarachnoid diffusion to be decreased in these patients. We aim to quantify the diffusivity of ventricular and subarachnoid cerebrospinal fluid in ESSI patients, to determine if diffusion characteristics deviate from normocephalic infants, and to propose a unique mechanism for ESSI. Materials and methods 227 consecutive brain magnetic resonance exams from different macrocephalic children were retrospectively reviewed after institutional review board waiver. Patients with noncommunicating hydrocephalus, substantial ventriculomegaly, atrophy, structural bone and/parenchymal abnormalities, abnormal brain signal, hemorrhages, meningitis, and normal imaging were excluded. A total of 53 exams from macrocephalic patients and 21 normocephalic subjects were analyzed. Mean quantitative apparent diffusion coefficient (ADC) values were obtained from the ventricular frontal horn and frontal subarachnoid spaces. The subarachnoid:ventricular ADC ratios were compared using a Mann-Whitney U-test. Results The mean age was 13 +/-8 months (macrocephalic cohort) and 13 +/- 6 months (normocephalic cohort). The subarachnoid fluid mean ADC was 2.50+/-0.26 × 10-3 mm2/s in the macrocephalic group and 2.84+/-0.29 × 10-3 mm2/s in the normocephalic group. The ventricular fluid mean ADC was 2.97+/-0.37 × 10-3 mm2/s and 2.74 +/-0.32 × 10-6 mm2/s, respectively. The mean quantitative ADC ratios in the macrocephalic group were 0.85, significantly smaller than the normocephalic group (1) ( z = -6.3; p = 0). Conclusion Subarachnoid space fluid diffusivity is reduced in patients with enlarged subarachnoid spaces of infancy. We propose insufficient frontotemporal capillary protein resorption to be the initiating factor in ESSI, leading to unbalanced osmotic/hydrostatic pressures, and secondary congestion.


Subject(s)
Cerebrospinal Fluid/diagnostic imaging , Diffusion Magnetic Resonance Imaging/methods , Megalencephaly/diagnostic imaging , Subarachnoid Space/diagnostic imaging , Female , Humans , Image Interpretation, Computer-Assisted , Infant , Male , Retrospective Studies
7.
J Neurotrauma ; 33(7): 662-71, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26414591

ABSTRACT

Traumatic brain injury (TBI) caused by explosive munitions, known as blast TBI, is the signature injury in recent military conflicts in Iraq and Afghanistan. Diagnostic evaluation of TBI, including blast TBI, is based on clinical history, symptoms, and neuropsychological testing, all of which can result in misdiagnosis or underdiagnosis of this condition, particularly in the case of TBI of mild-to-moderate severity. Prognosis is currently determined by TBI severity, recurrence, and type of pathology, and also may be influenced by promptness of clinical intervention when more effective treatments become available. An important task is prevention of repetitive TBI, particularly when the patient is still symptomatic. For these reasons, the establishment of quantitative biological markers can serve to improve diagnosis and preventative or therapeutic management. In this study, we used a shock-tube model of blast TBI to determine whether manganese-enhanced magnetic resonance imaging (MEMRI) can serve as a tool to accurately and quantitatively diagnose mild-to-moderate blast TBI. Mice were subjected to a 30 psig blast and administered a single dose of MnCl2 intraperitoneally. Longitudinal T1-magnetic resonance imaging (MRI) performed at 6, 24, 48, and 72 h and at 14 and 28 days revealed a marked signal enhancement in the brain of mice exposed to blast, compared with sham controls, at nearly all time-points. Interestingly, when mice were protected with a polycarbonate body shield during blast exposure, the marked increase in contrast was prevented. We conclude that manganese uptake can serve as a quantitative biomarker for TBI and that MEMRI is a minimally-invasive quantitative approach that can aid in the accurate diagnosis and management of blast TBI. In addition, the prevention of the increased uptake of manganese by body protection strongly suggests that the exposure of an individual to blast risk could benefit from the design of improved body armor.


Subject(s)
Blast Injuries/diagnostic imaging , Brain Injuries, Traumatic/diagnostic imaging , Contrast Media , Magnetic Resonance Imaging/methods , Manganese , Animals , Disease Models, Animal , Image Processing, Computer-Assisted , Male , Mice , Mice, Inbred C57BL
8.
Clin Perinatol ; 42(2): 337-61, ix, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26042908

ABSTRACT

Up to 14% of patients with congenital metabolic disease may show structural brain abnormalities from perturbation of cell proliferation, migration, and/or organization. Most inborn errors of metabolism have a postnatal onset. Abnormalities from genetic disease processes have a prenatal onset. Energy impairment, substrate insufficiency, cell membrane receptor and cell signaling abnormalities, and toxic byproduct accumulation are associations between genetic disorders and structural brain anomalies. Collective imaging patterns of brain abnormalities can provide clues to the underlying etiology. We review selected metabolic diseases associated with brain malformations and highlight characteristic clinical and imaging manifestations that help narrow the differential diagnosis.


Subject(s)
Brain Diseases , Diagnostic Imaging , Genetic Testing/methods , Brain Diseases/congenital , Brain Diseases/diagnosis , Brain Diseases/genetics , Humans , Infant, Newborn
9.
Am J Pathol ; 182(2): 312-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23219428

ABSTRACT

Biologically accurate mouse models of human cancer have become important tools for the study of human disease. The anatomical location of various target organs, such as brain, pancreas, and prostate, makes determination of disease status difficult. Imaging modalities, such as magnetic resonance imaging, can greatly enhance diagnosis, and longitudinal imaging of tumor progression is an important source of experimental data. Even in models where the tumors arise in areas that permit visual determination of tumorigenesis, longitudinal anatomical and functional imaging can enhance the scope of studies by facilitating the assessment of biological alterations, (such as changes in angiogenesis, metabolism, cellular invasion) as well as tissue perfusion and diffusion. One of the challenges in preclinical imaging is the development of infrastructural platforms required for integrating in vivo imaging and therapeutic response data with ex vivo pathological and molecular data using a more systems-based multiscale modeling approach. Further challenges exist in integrating these data for computational modeling to better understand the pathobiology of cancer and to better affect its cure. We review the current applications of preclinical imaging and discuss the implications of applying functional imaging to visualize cancer progression and treatment. Finally, we provide new data from an ongoing preclinical drug study demonstrating how multiscale modeling can lead to a more comprehensive understanding of cancer biology and therapy.


Subject(s)
Magnetic Resonance Imaging/methods , Neoplasms/diagnosis , Research , Systems Biology/methods , Animals , Antineoplastic Agents/therapeutic use , Computational Biology , Humans , Neoplasms/drug therapy
10.
Cell Cycle ; 11(20): 3801-9, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22983062

ABSTRACT

The development of new small molecule-based therapeutic drugs requires accurate quantification of drug bioavailability, biological activity and treatment efficacy. Rapidly measuring these endpoints is often hampered by the lack of efficient assay platforms with high sensitivity and specificity. Using an in vivo model system, we report a simple and sensitive liquid chromatography-tandem mass spectrometry assay to quantify the bioavailability of a recently developed novel cyclin-dependent kinase inhibitor VMY-1-103, a purvalanol B-based analog whose biological activity is enhanced via dansylation. We developed a rapid organic phase extraction technique and validated wide and functional VMY-1-103 distribution in various mouse tissues, consistent with its enhanced potency previously observed in a variety of human cancer cell lines. More importantly, in vivo MRI and single voxel proton MR-Spectroscopy further established that VMY-1-103 inhibited disease progression and affected key metabolites in a mouse model of hedgehog-driven medulloblastoma.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Cerebellar Neoplasms/drug therapy , Cyclin-Dependent Kinases/antagonists & inhibitors , Dansyl Compounds/pharmacology , Medulloblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Adenine/chemistry , Adenine/pharmacokinetics , Adenine/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Cell Cycle/drug effects , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Chromatography, Liquid , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Dansyl Compounds/pharmacokinetics , Humans , Magnetic Resonance Imaging , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Protein Kinase Inhibitors/pharmacokinetics , Tandem Mass Spectrometry
11.
Med Phys ; 39(5): 2578-83, 2012 May.
Article in English | MEDLINE | ID: mdl-22559628

ABSTRACT

PURPOSE: A time-varying magnetic field can cause unpleasant peripheral nerve stimulation (PNS) when the maximum excursion of the magnetic field (ΔB) is above a frequency-dependent threshold level [P. Mansfield and P. R. Harvey, Magn. Reson. Med. 29, 746-758 (1993)]. Clinical and research magnetic resonance imaging (MRI) gradient systems have been designed to avoid such bioeffects by adhering to regulations and guidelines established on the basis of clinical trials. Those trials, generally employing sinusoidal waveforms, tested human responses to magnetic fields at frequencies between 0.5 and 10 kHz [W. Irnich and F. Schmitt, Magn. Reson. Med. 33, 619-623 (1995), T. F. Budinger et al., J. Comput. Assist. Tomogr. 15, 909-914 (1991), and D. J. Schaefer et al., J. Magn. Reson. Imaging 12, 20-29 (2000)]. PNS thresholds for frequencies higher than 10 kHz had been extrapolated, using physiological models [J. P. Reilly et al., IEEE Trans. Biomed. Eng. BME-32(12), 1001-1011 (1985)]. The present study provides experimental data on human PNS thresholds to oscillating magnetic field stimulation from 2 to 183 kHz. Sinusoidal waveforms were employed for several reasons: (1) to facilitate comparison with earlier reports that used sine waves, (2) because prior designers of fast gradient hardware for generalized waveforms (e.g., including trapezoidal pulses) have employed quarter-sine-wave resonant circuits to reduce the rise- and fall-times of pulse waveforms, and (3) because sinusoids are often used in fast pulse sequences (e.g., spiral scans) [S. Nowak, U.S. patent 5,245,287 (14 September 1993) and K. F. King and D. J. Schaefer, J. Magn. Reson. Imaging 12, 164-170 (2000)]. METHODS: An IRB-approved prospective clinical trial was performed, involving 26 adults, in which one wrist was exposed to decaying sinusoidal magnetic field pulses at frequencies from 2 to 183 kHz and amplitudes up to 0.4 T. Sham exposures (i.e., with no magnetic fields) were applied to all subjects. RESULTS: For 0.4 T pulses at 2, 25, 59, 101, and 183 kHz, stimulation was reported by 22 (84.6%), 24 (92.3%), 15 (57.7%), 2 (7.7%), and 1 (3.8%) subjects, respectively. CONCLUSIONS: The probability of PNS due to brief biphasic time-varying sinusoidal magnetic fields with magnetic excursions up to 0.4 T is shown to decrease significantly at and above 101 kHz. This phenomenon may have particular uses in dynamic scenarios (e.g., cardiac imaging) and in studying processes with short decay times (e.g., electron paramagnetic resonance imaging, bone and solids imaging). The study suggests the possibility of new designs for human and preclinical MRI systems that may be useful in clinical practice and scientific research.


Subject(s)
Magnetic Fields/adverse effects , Peripheral Nerves/physiology , Adult , Humans , Probability , Sensation/physiology , Sensory Thresholds
12.
J Magn Reson Imaging ; 32(4): 836-46, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20882614

ABSTRACT

PURPOSE: To investigate the utility of noninvasive magnetic resonance imaging (MRI) protocols to demonstrate pathological differences between rats and mice after spinal cord injury (SCI). Rats and mice are commonly used to model SCI; however, histology and immunohistochemistry have shown differences in neuropathology between the two species, including cavity formation and scar/inflammatory responses. MATERIALS AND METHODS: Moderate contusion SCI was performed on adult male rats and mice. At 28 days postinjury, animals underwent T1-weighted (T1W), with or without gadolinium contrast, or T2-weighted (T2W) magnetic resonance imaging (MRI), to be compared with histology at the same timepoint. RESULTS: In both species, all MRI methods demonstrated changes in spinal cord anatomy. Immunohistochemistry indicated that T2W accurately reflected areas of inflammation and glial scar formation in rats and mice. Quantitation of lesion volume by histology and functional performance correlated best with T2W measurements in both species. Gadolinium contrast accurately reflected the blood-spinal cord-barrier permeability in both species, which appeared greater in rats than in mice. CONCLUSION: These data demonstrate that MRI, with either a T1W or T2W protocol, can effectively distinguish pathological differences between rats and mice.


Subject(s)
Gadolinium/pharmacology , Magnetic Resonance Imaging/methods , Spinal Cord Injuries/physiopathology , Animals , Contrast Media/pharmacology , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Permeability , Rats , Rats, Sprague-Dawley , Species Specificity , Spinal Cord/pathology , Spinal Cord Injuries/diagnosis , Time Factors
13.
Cell Cycle ; 9(9): 1824-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20404514

ABSTRACT

Diet and obesity, and their associated metabolic alterations, are some of the fastest-growing causes of disease and death in America. Findings from epidemiological studies correlating obesity, the sources of dietary fat and prostate cancer (PCa) are conflicting. We have previously shown that 15% of PB-ErbB-2 x pten(+/-) mice developed PCa and exhibited increased phosphorylated 4E-BP1, but not the key PI3-kinase intermediary phospho-protein, mTOR, when maintained on unrefined mouse chow. We report herein that 100% of animals fed refined, westernized AIN-93-based diets containing corn oil developed PCa by 12 months of age. Increases in visceral fat and mTO R activation in the tumors were also observed. Furthermore, nuclear cyclin E levels were significantly induced by the AIN-93-corn oil-based diets versus chow. Replacing 50% of the corn oil with menhaden oil, with 21% of its triglycerides being n-3 PUFA's, had no effect on tumorigenesis, fat deposition, cyclin E or mTOR. Phosphorylated BAD levels were similar in the tumors of mice in all three diets. Our data demonstrated that in the context of our preclinical model, components of crude chow, but not dietary n-3 PUFAs, protect against PCa progression. In addition, these data establish phosphorylated mTOR, nuclear cyclin E and visceral fat deposits as possible biomarkers of increased dietary risk for PCa.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/prevention & control , Receptor, ErbB-2/genetics , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cyclin E/metabolism , Disease Models, Animal , Disease Progression , Eukaryotic Initiation Factors , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , PTEN Phosphohydrolase/metabolism , Phosphoproteins/metabolism , Phosphorylation , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/metabolism , TOR Serine-Threonine Kinases , bcl-Associated Death Protein/metabolism
14.
Mol Genet Metab ; 94(1): 52-60, 2008 May.
Article in English | MEDLINE | ID: mdl-18262815

ABSTRACT

We used (1)H MRS to evaluate brain metabolic differences in sisters with partial ornithine transcarbamylase deficiency (OTCD) who had discordant clinical symptoms and urea synthetic capabilities to assess whether a brain biomarker of partial OTCD correlated with urea synthetic ability and clinical severity. We performed single voxel 3.0T (1)H MRS in two adult sisters with partial OTCD, one symptomatic and one asymptomatic, in a stable medical state and compared it to one age matched adult control, as well as data collected on an additional 13 subjects with partial OTCD and 12 controls. Data from voxels placed in frontal and parietal white matter (FWM, PWM), posterior cingulate gray matter (PCGM), and thalamus (tha), were corrected for partial volume and analyzed using "LCModel". All three subjects as well as the symptomatic mother of the two sisters, had neurocognitive testing, plasma ammonia levels, plasma amino acid, and urine organic acid analysis. Previous urea synthetic capabilities had been measured by stable isotope analysis. We found IQ scores to be inversely related to symptoms. Decreased myoinositol (mI) identified OTCD subjects, even the sister who is asymptomatic, in the posterior parietal white matter and frontal white matter. Brain metabolism is impaired in partial OTCD. Abnormal metabolism in apparently asymptomatic OTCD females may provide an explanation for neurocognitive impairments previously reported. The concentration of mI seen on (1)H MRS in PWM and FWM in this family could be used to deduce clinical symptomatology and may serve as a non-invasive marker of brain liability in OTCD.


Subject(s)
Brain Chemistry , Cognition Disorders/diagnosis , Inositol/analysis , Magnetic Resonance Spectroscopy , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Adult , Age of Onset , Brain/pathology , Cell Differentiation , Cognition Disorders/pathology , Female , Humans , Hydrogen , Magnetic Resonance Imaging , Ornithine Carbamoyltransferase Deficiency Disease/pathology , Pedigree , Phenotype , Siblings
15.
Exp Neurol ; 209(1): 224-33, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18028910

ABSTRACT

Animal models treated with agricultural chemicals, such as rotenone, reproduce several degenerative features of human central nervous system (CNS) diseases. Glutamate is the most abundant excitatory amino acid transmitter in the mammalian central nervous system and its transmission is implicated in a variety of brain functions including mental behavior and memory. Dysfunction of glutamate neurotransmission in the CNS has been associated with a number of human neurodegenerative diseases, either as a primary or as a secondary factor in the excitotoxic events leading to neuronal death. Since many human CNS disorders do not arise spontaneously in animals, characteristic functional changes have to be mimicked by toxic agents. Candidate environmental toxins bearing any direct or indirect effects on the pathogenesis of human disease are particularly useful. The present longitudinal Magnetic Resonance Imaging (MRI) studies show, for the first time, significant variations in the properties of brain ventricles in a rotenone-treated (2 mg/kg) mouse model over a period of 4 weeks following 3 days of rotenone treatment. Histopathological analysis reveals death of stria terminalis neurons following this short period of rotenone treatment. Furthermore, in vivo voxel localized (1)H MR spectroscopy also shows for the first time significant bio-energetic and metabolic changes as well as temporal alterations in the levels of glutamate in the degenerating striatal region. These studies provide novel insights on the effects of environmental toxins on glutamate and other amino acid neurotransmitters in human neurodegenerative diseases.


Subject(s)
Glutamic Acid/physiology , Neostriatum/physiopathology , Neurotoxicity Syndromes/physiopathology , Rotenone/toxicity , Synaptic Transmission/physiology , Uncoupling Agents/toxicity , Animals , Brain/pathology , Cell Death/drug effects , Citric Acid Cycle/drug effects , Energy Metabolism/drug effects , Feedback/physiology , Immunohistochemistry , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Neostriatum/metabolism , Oxidative Phosphorylation/drug effects , Synaptic Transmission/drug effects , Tyrosine 3-Monooxygenase/metabolism
16.
Cell Cycle ; 6(15): 1946-50, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671425

ABSTRACT

Pheochromocytoma (PCC) is a rare catecholamine-producing tumor that arises from the adrenal medulla and is often familial. The genetic basis for familial PCC involves mutations of RET, VHL, SHDx or NF-1 in more than 20% of cases. Additional genes may be important in pathogenesis of both familial and sporadic PCC. ErbB-2/Her2/Neu is a growth factor receptor tyrosine kinase that is frequently overexpressed in tumors and there is clinical evidence suggesting that enhanced ErbB-2 growth factor receptor signaling may play a role in PCC. In the present study, ectopic expression of an activated ErbB-2 transgene resulted in bilateral adrenal PCC. Analyses of tumor samples and normal adrenal tissue revealed that levels of the Pten tumor suppressor protein were greatly reduced in PCCs, while levels of the cell cycle regulatory protein cyclin D1 were usually increased. In addition, levels of phospo-AKT were increased in PCCs versus normal adrenal tissue. Biochemical analyses established that PCC's were functionally active, producing abundant levels of the catecholamines, epinephrine and norepinephrine. These data establish that increased ErbB-2 growth factor receptor signaling in the adrenal medulla can lead to PCC through combined influences on Pten, AKT andcyclin D1.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Receptor, ErbB-2/metabolism , Adrenal Gland Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Immunohistochemistry , Mice , PTEN Phosphohydrolase/genetics , Pheochromocytoma/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
17.
Nat Med ; 13(7): 803-11, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17603492

ABSTRACT

The relationship between stress and obesity remains elusive. In response to stress, some people lose weight, whereas others gain. Here we report that stress exaggerates diet-induced obesity through a peripheral mechanism in the abdominal white adipose tissue that is mediated by neuropeptide Y (NPY). Stressors such as exposure to cold or aggression lead to the release of NPY from sympathetic nerves, which in turn upregulates NPY and its Y2 receptors (NPY2R) in a glucocorticoid-dependent manner in the abdominal fat. This positive feedback response by NPY leads to the growth of abdominal fat. Release of NPY and activation of NPY2R stimulates fat angiogenesis, macrophage infiltration, and the proliferation and differentiation of new adipocytes, resulting in abdominal obesity and a metabolic syndrome-like condition. NPY, like stress, stimulates mouse and human fat growth, whereas pharmacological inhibition or fat-targeted knockdown of NPY2R is anti-angiogenic and anti-adipogenic, while reducing abdominal obesity and metabolic abnormalities. Thus, manipulations of NPY2R activity within fat tissue offer new ways to remodel fat and treat obesity and metabolic syndrome.


Subject(s)
Diet , Neuropeptide Y/genetics , Obesity/metabolism , Stress, Physiological/metabolism , 3T3-L1 Cells , Adipose Tissue, White/metabolism , Animals , Cold Temperature , Dietary Fats , Gene Deletion , Gene Expression Regulation , Male , Metabolic Syndrome , Mice , Mice, Inbred C57BL , Mice, Nude , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/genetics , Up-Regulation
18.
Mol Cell Biol ; 26(14): 5449-69, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809779

ABSTRACT

The cyclin D1 gene encodes a regulatory subunit of the holoenzyme that phosphorylates and inactivates the pRb tumor suppressor to promote nuclear DNA synthesis. cyclin D1 is overexpressed in human breast cancers and is sufficient for the development of murine mammary tumors. Herein, cyclin D1 is shown to perform a novel function, inhibiting mitochondrial function and size. Mitochondrial activity was enhanced by genetic deletion or antisense or small interfering RNA to cyclin D1. Global gene expression profiling and functional analysis of mammary epithelial cell-targeted cyclin D1 antisense transgenics demonstrated that cyclin D1 inhibits mitochondrial activity and aerobic glycolysis in vivo. Reciprocal regulation of these genes was observed in cyclin D1-induced mammary tumors. Cyclin D1 thus integrates nuclear DNA synthesis and mitochondrial function.


Subject(s)
Cyclin D1/metabolism , Mitochondria/metabolism , Animals , Base Sequence , Cyclin D1/deficiency , Cyclin D1/genetics , DNA/genetics , Female , Gene Expression Profiling , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Humans , Lipogenesis/genetics , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Models, Biological , Multigene Family , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics
19.
Prostate ; 66(7): 708-17, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16425198

ABSTRACT

BACKGROUND: Mouse prostate cancer modeling presents unique obstacles to the study of spontaneous tumor initiation and progression due to the anatomical location of the tissue. RESULTS: High resolution (130 microm(x) x 130 microm(y) x 300 microm(z)), three-dimensional MRI allowed for the visualization, segmentation, and volumetric measurement of the prostate from normal and genetically engineered animals, in vivo. Additionally, MRS performed on the prostate epithelia of probasin-ErbB-2Delta x Pten(+/-) mice identified changes in the relative concentrations of the metabolites choline and citrate, which was not observed in TRAMP mice. METHODS: T1-weighted MRI was performed on normal, TRAMP, probasin-ErbB-2/Her2/Neu (probasin-ErbB-2Delta), and probasin-ErbB-2Delta in the context of decreased Pten activity (probasin-ErbB-2Delta x Pten(+/-)) mice. Volume-localized single-voxel proton magnetic resonance spectroscopy (SVS (1)H MRS) was also performed. CONCLUSIONS: The data presented supports the use of combined MRI and MRS for the measurement of biochemical and morphometric alterations in mouse models of prostate cancer.


Subject(s)
Disease Models, Animal , Prostatic Neoplasms/pathology , Prostatic Neoplasms/veterinary , Animals , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice
20.
Neurobiol Dis ; 17(1): 29-43, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15350963

ABSTRACT

Experimental models of traumatic brain injury have been developed to replicate selected aspects of human head injury, such as contusion, concussion, and/or diffuse axonal injury. Although diffuse axonal injury is a major feature of clinical head injury, relatively few experimental models of diffuse traumatic brain injury (TBI) have been developed, particularly in smaller animals such as rodents. Here, we describe the pathophysiological consequences of moderate diffuse TBI in rats generated by a newly developed, highly controlled, and reproducible model. This model of TBI caused brain edema beginning 20 min after injury and peaking at 24 h post-trauma, as shown by wet weight/dry weight ratios and diffusion-weighted magnetic resonance imaging. Increased permeability of the blood-brain barrier was present up to 4 h post-injury as evaluated using Evans blue dye. Phosphorus magnetic resonance spectroscopy showed significant declines in brain-free magnesium concentration and reduced cytosolic phosphorylation potential at 4 h post-injury. Diffuse axonal damage was demonstrated using manganese-enhanced magnetic resonance imaging, and intracerebral injection of a fluorescent vital dye (Fluoro-Ruby) at 24-h and 7-day post-injury. Morphological evidence of apoptosis and caspase-3 activation were also found in the cerebral hemisphere and brainstem at 24 h after trauma. These results show that this model is capable of reproducing major biochemical and neurological changes of diffuse clinical TBI.


Subject(s)
Brain Injuries/pathology , Brain/pathology , Disease Models, Animal , Animals , Apoptosis/physiology , Brain/metabolism , Brain Injuries/metabolism , Caspase 3 , Caspases/metabolism , Male , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...