Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Br J Cancer ; 103(8): 1297-304, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20842126

ABSTRACT

BACKGROUND: Human medulloblastomas exhibit diverse molecular pathology. Aberrant hedgehog signalling is found in 20-30% of human medulloblastomas with largely unknown metabolic consequences. METHODS: Transgenic mice over-expressing smoothened (SMO) receptor in granule cell precursors with high incidence of exophytic medulloblastomas were sequentially followed up by magnetic resonance imaging (MRI) and characterised for metabolite phenotypes by ¹H MR spectroscopy (MRS) in vivo and ex vivo using high-resolution magic angle spinning (HR-MAS) ¹H MRS. RESULTS: Medulloblastomas in the SMO mice presented as T2 hyperintense tumours in MRI. These tumours showed low concentrations of N-acetyl aspartate and high concentrations of choline-containing metabolites (CCMs), glycine, and taurine relative to the cerebellar parenchyma in the wild-type (WT) C57BL/6 mice. In contrast, ¹H MRS metabolite concentrations in normal appearing cerebellum of the SMO mice were not different from those in the WT mice. Macromolecule and lipid ¹H MRS signals in SMO medulloblastomas were not different from those detected in the cerebellum of WT mice. The HR-MAS analysis of SMO medulloblastomas confirmed the in vivo ¹H MRS metabolite profiles, and additionally revealed that phosphocholine was strongly elevated in medulloblastomas accounting for the high in vivo CCM. CONCLUSIONS: These metabolite profiles closely mirror those reported from human medulloblastomas confirming that SMO mice provide a realistic model for investigating metabolic aspects of this disease. Taurine, glycine, and CCM are potential metabolite biomarkers for the SMO medulloblastomas. The MRS data from the medulloblastomas with defined molecular pathology is discussed in the light of metabolite profiles reported from human tumours.


Subject(s)
Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Metabolome , Nuclear Magnetic Resonance, Biomolecular , Receptors, G-Protein-Coupled/genetics , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellum/chemistry , Cerebellum/metabolism , Cerebellum/pathology , Choline/analysis , Choline/metabolism , Hydrogen/chemistry , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, G-Protein-Coupled/physiology , Smoothened Receptor , Taurine/analysis , Taurine/metabolism , Tumor Burden/physiology
2.
Eur J Radiol ; 56(2): 160-4, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16233889

ABSTRACT

In proton magnetic resonance spectroscopic imaging (1H MRSI), the recorded spectra are often linear combinations of spectra from different cell and tissue types within the voxel. This produces problems for data analysis and interpretation. A sophisticated approach is proposed here to handle the complexity of tissue heterogeneity in MRSI data. The independent component analysis (ICA) method was applied without prior knowledge to decompose the proton spectral components that relate to the heterogeneous cell populations with different proliferation and metabolism that are present in gliomas. The ability to classify brain tumours based on IC decomposite spectra was studied by grouping the components with histopathology. To this end, 10 controls and 34 patients with primary brain tumours were studied. The results indicate that ICA may reveal useful information from metabolic profiling for clinical purposes using long echo time MRSI of gliomas.


Subject(s)
Brain Neoplasms/pathology , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Algorithms , Aspartic Acid/analogs & derivatives , Aspartic Acid/analysis , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Cell Proliferation , Choline/analysis , Creatine/analysis , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/metabolism , Glioma/pathology , Humans , Hydrogen , Image Interpretation, Computer-Assisted , Lactic Acid/analysis , Lipids/analysis , Oligodendroglioma/metabolism , Oligodendroglioma/pathology , Phosphocreatine/analysis
3.
Magn Reson Med ; 49(1): 47-60, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509819

ABSTRACT

Gradient-echo (GRE) blood oxygen level-dependent (BOLD) effects have both intra- and extravascular contributions. To better understand the intravascular contribution in quantitative terms, the spin-echo (SE) and GRE transverse relaxation rates, R(2) and R(2)(*), of isolated blood were measured as a function of oxygenation in a perfusion system. Over the normal oxygenation saturation range of blood between veins, capillaries, and arteries, the difference between these rates, R'(2) = R(2)(*) - R(2), ranged from 1.5 to 2.1 Hz at 1.5 T and from 26 to 36 Hz at 4.7 T. The blood data were used to calculate the expected intravascular BOLD effects for physiological oxygenation changes that are typical during visual activation. This modeling showed that intravascular DeltaR(2)(*) is caused mainly by R(2) relaxation changes, namely 85% and 78% at 1.5T and 4.7T, respectively. The simulations also show that at longer TEs (>70 ms), the intravascular contribution to the percentual BOLD change is smaller at high field than at low field, especially for GRE experiments. At shorter TE values, the opposite is the case. For pure parenchyma, the intravascular BOLD signal changes originate predominantly from venules for all TEs at low field and for short TEs at high field. At longer TEs at high field, the capillary contribution dominates. The possible influence of partial volume contributions with large vessels was also simulated, showing large (two- to threefold) increases in the total intravascular BOLD effect for both GRE and SE.


Subject(s)
Cerebrovascular Circulation , Magnetic Resonance Imaging , Oxygen/blood , Animals , Cattle , Cerebrovascular Circulation/physiology , Hematocrit , Humans , Magnetic Resonance Spectroscopy , Microcirculation , Models, Cardiovascular , Phantoms, Imaging
4.
Biochem Biophys Res Commun ; 289(4): 813-8, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11735118

ABSTRACT

T1 relaxation in the rotating frame (T1rho) is a sensitive magnetic resonance imaging (MRI) contrast for acute brain insults. Biophysical mechanisms affecting T1rho relaxation rate (R1rho) and R1rho dispersion (dependency of R1rho on the spin-lock field) were studied in protein solutions by varying their chemical environment and pH in native, heat-denatured, and glutaraldehyde (GA) cross-linked samples. Low pH strongly reduced R1rho in heat-denatured phantoms displaying proton resonances from a number of side-chain chemical groups in high-resolution 1H NMR spectra. At pH of 5.5, R1rho dispersion was completely absent. In contrast, in the GA-treated phantoms with very few NMR visible side chain groups, acidic pH showed virtually no effect on R1rho. The present data point to a crucial role of proton exchange on R1rho and R1rho dispersion in immobilized protein solution mimicking tissue relaxation properties.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Proteins/chemistry , Animals , Biophysical Phenomena , Biophysics , Brain Ischemia/metabolism , Cattle , Cross-Linking Reagents , Glutaral , Hydrogen-Ion Concentration , In Vitro Techniques , Magnetic Resonance Imaging , Phantoms, Imaging , Protein Denaturation , Protons , Serum Albumin, Bovine/chemistry , Solutions
5.
NMR Biomed ; 14(7-8): 432-40, 2001.
Article in English | MEDLINE | ID: mdl-11746935

ABSTRACT

The introduction of new neuroprotective treatment strategies for acute stroke patients has provided a requirement for neuroimaging methods capable of identifying salvageable tissue in acute stroke patients. Substantial positron emission tomography evidence points to the fact that a peri-infarct zone with blood flow of 20-45% of normal, metabolic rate of oxygen of >35% of normal and oxygen extraction ratio (OER) of >0.7 are indices of tissue at risk of infarction, yet with potential for recovery. The sensitivity of T(2) to blood oxygen level dependent (BOLD) effects allows the mismatch between oxygen delivery and consumption in the brain to be imaged. Previous evidence from animal models of cerebral hypoperfusion and ischemic stroke strongly suggest that T(2) BOLD MRI highlights viable and salvageable brain regions. The Hahn-echo T(2) and diffusion show distinct flow thresholds in the rat brain so that the former parameter probes areas with high OER and the latter genuine ischemia. In the flow-compromised tissue showing negative T(2) BOLD, substantial residual perfusion is evident as revealed by bolus-tracking perfusion MRI, in agreement with the idea that tissue metabolic viability must be preserved for expression of BOLD. It is concluded that BOLD MRI may have potential for the assessment of tissue viability in acute ischemic stroke.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Magnetic Resonance Imaging , Oxygen/blood , Stroke/metabolism , Animals , Cerebrovascular Circulation , Humans , Oxygen Consumption , Rats
6.
Magn Reson Med ; 46(5): 946-54, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11675647

ABSTRACT

The present study was designed to investigate whether T(2)-weighted signal changes obtained by microimaging of paraformaldehyde-fixed brain correlate with the histologically quantified damage in a model of status epilepticus (SE) induced by kainic acid in the rat. Animals were killed at several time points up to 8 weeks after a single intraperitoneal kainate (KA) injection (9 mg/kg). Perfusion-fixed brains were embedded in gelatin for MR microimaging at 9.4T. After the MRI analysis, the gelatin was removed and the brains were cryoprotected and processed for quantitative histology. Severity of neuronal damage and gliosis were assessed from thionin-stained serial sections. Correlative analysis of microimaging and histology data was done in the hippocampus, amygdala, parietal rhinal cortex (PaRH), piriform cortex (Pir), and entorhinal cortex. The relative signal intensities in T(2)-weighted images correlate with the severity of neuronal damage in the matched histological sections (correlation coefficients of 0.752-0.826). Our data show that MR microimaging ex vivo detects the degree of neuronal damage and its anatomical distribution after KA-induced SE, thus providing a useful tool for detecting the dynamics of progressive neuronal damage after prolonged seizures.


Subject(s)
Brain/pathology , Magnetic Resonance Imaging/methods , Status Epilepticus/pathology , Animals , Kainic Acid , Male , Neurons/pathology , Rats , Rats, Wistar , Status Epilepticus/chemically induced
7.
Magn Reson Med ; 46(3): 565-72, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11550250

ABSTRACT

Time-dependent changes of T1 in the rotating frame (T1rho), diffusion, T2, and magnetization transfer contrast on cardiac arrest-induced global ischemia in rat were investigated. T1rho, as acquired with spin lock amplitudes >0.6 G, started to increase 10-20 sec after cardiac arrest followed by an increase within 3-4 min to a level that was 6-8% greater than in normal brain. The ischemic T1rho response coincided with the drop of water diffusion coefficient in normoglycemic animals. However, unlike the rate of diffusion, the kinetics of T1rho were not affected by either preischemic hypoglycemia or hyperglycemia. Similar to diffusion, the kinetics of anoxic depolarization were dependent on preischemic blood glucose levels. Ischemia caused a reduction in the Hahn spin echo T2 as a result of blood oxygenation level-dependent (BOLD) effect; maximal negative BOLD seen by 40 sec. In the animals injected with an ironoxide particle contrast agent, AMI-227, prior to the insult, both T1rho and T2 immediately increased in concert on induction of ischemia. In contrast to the T1rho and diffusion changes, a much slower change in magnetization transfer contrast was evident over the first 20 min of ischemia. These data demonstrate that T1rho immediately increases following ischemia and that the pathophysiological mechanisms affecting this relaxation time may not directly involve magnetization transfer. The mechanisms prolonging T1rho differ from those affecting water diffusion with respect to their sensitivities to glucose and are apparently independent of membrane depolarization.


Subject(s)
Blood Glucose/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Image Enhancement , Magnetic Resonance Imaging , Animals , Blood Volume/physiology , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/physiopathology , Brain Mapping , Diffusion , Heart Arrest, Induced , Male , Rats , Rats, Wistar , Somatosensory Cortex/blood supply , Somatosensory Cortex/physiopathology
8.
Cancer Gene Ther ; 8(7): 529-36, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11498775

ABSTRACT

We have treated Caki-2 human renal cell carcinoma in vivo using herpes simplex virus thymidine kinase (HSV-tk) gene therapy. Both stably transduced Caki-2 tumors, generated using retrovirus-mediated ex vivo HSV-tk gene transfer and direct intratumoral adenovirus-mediated HSV-tk gene transfer of wild type tumors, were tested. Similar treatments with LacZ containing retro- and adenoviruses were used as controls. The outcome was evaluated by imaging the tumors before and after the treatment with magnetic resonance imaging, and using histology, immunocytochemistry, and survival analysis. When implanted orthotopically into nude mouse kidneys, Caki-2 cells formed reproducible cystic papillary kidney carcinomas. In vivo magnetic resonance imaging provided an important tool for the evaluation of tumor growth. Transduction efficiency of wild-type tumors in vivo with adeno-LacZ was 22+/-14%. Significant tumor regression was achieved with direct intratumoral adeno-HSV-tk transduction followed by intraperitoneal ganciclovir (GCV) (P<.001). Also, the treatment of stably transduced Caki-2 tumors with intraperitoneal GCV resulted in a significant treatment response in the HSV-tk group as compared to the LacZ group (P<.009). Increased apoptosis and macrophage infiltrations, reduced proliferation, and degenerative changes were observed in the tumors treated with HSV-tk and GCV. Also, significant prolongation in survival was achieved with adeno-HSV-tk- and GCV-treated mice as compared to the controls. It is concluded that adeno-HSV-tk gene therapy may be useful for the treatment of renal cell carcinoma in vivo.


Subject(s)
Carcinoma, Renal Cell/therapy , Genetic Therapy/methods , Kidney Neoplasms/therapy , Simplexvirus/genetics , Thymidine Kinase/genetics , Adenoviridae/genetics , Animals , Antiviral Agents/pharmacology , Apoptosis , Cell Division , Ganciclovir/pharmacology , Gene Transfer Techniques , Humans , Immunohistochemistry , Kidney Neoplasms/pathology , Lac Operon , Macrophages/metabolism , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Genetic , Neoplasm Transplantation , Retroviridae/genetics , Time Factors , Transduction, Genetic , Tumor Cells, Cultured
9.
Magn Reson Med ; 46(2): 282-91, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11477631

ABSTRACT

It has recently been shown that parenchymal oxygen extraction ratios (OERs) can be quantified using the absolute T(2) of venous blood draining from this tissue (Oja et al., J Cereb Blood Flow Metab 1999;19:1289-1295). Here, a modified Carr-Purcell-Meiboom-Gill (CPMG) multiecho experiment was used to increase the efficiency and precision of this approach and to test the applicability of the two-compartment exchange model for spin-echo BOLD effects in pure venous blood. Relaxation measurements on bovine blood as a function of CPMG interecho spacing, oxygen saturation, and hematocrit provided the baseline relaxation and susceptibility shift parameters necessary to directly relate OER to T(2) of venous blood in vivo. Using an interecho spacing of 25 ms, the results on visual activation studies in eight volunteers showed T(2)(CPMG) values increasing from 128 +/- 9 ms to 174 +/- 18 ms upon activation, corresponding to local OER values of 0.38 +/- 0.04 and 0.18 +/- 0.05 during baseline activity and visual stimulation, respectively. These OER values are in good agreement with literature data on venous oxygenation and numbers determined previously using a single-echo approach, while the measured T(2)s are about 20-40 ms longer.


Subject(s)
Blood/metabolism , Brain/blood supply , Echo-Planar Imaging/methods , Oxygen/metabolism , Adult , Animals , Blood Gas Analysis , Brain/metabolism , Brain Mapping/methods , Cattle , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Oxygen Consumption , Phantoms, Imaging , Photic Stimulation
10.
MAGMA ; 12(1): 32-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11255090

ABSTRACT

Inadequate blood supply relative to metabolic demand, a haemodynamic condition termed as misery perfusion, often occurs in conjunction with acute ischaemic stroke. Misery perfusion results in adaptive changes in cerebral physiology including increased cerebral blood volume (CBV) and oxygen extraction ratio (OER) to secure substrate supply for the brain. It has been suggested that the presence of misery perfusion may be an indication of reversible ischaemia, thus detection of this condition may have clinical impact in acute stroke imaging. The ability of single spin echo T(2) to detect misery perfusion in the rat brain at 1.5 T owing to its sensitivity to blood oxygenation level dependent (BOLD) contrast was studied both theoretically and experimentally. Based on the known physiology of misery perfusion, tissue morphometry and blood relaxation data, T(2) behaviour in misery perfusion was simulated. The interpretation of these computations was experimentally assessed by quantifying T(2) in a rat model for cerebral misery perfusion. CBF was quantified with the H(2) clearance method. A drop of CBF from 58+/-8 to 17+/-3 ml/100 g/min in the parieto-frontal cortex caused shortening of T(2) from 66.9+/-0.4 to 64.6+/-0.5 ms. Under these conditions, no change in diffusion MRI was detected. In contrast, the cortex with CBF of 42+/-7 ml/100 g/min showed no change in T(2). Computer simulations accurately predicted these T(2) responses. The present study shows that the acute drop of CBF by 70% causes a negative BOLD that is readily detectable by T(2) MRI at 1.5 T. Thus BOLD may serve as an index of misery perfusion thus revealing viable tissue with increased OER.


Subject(s)
Brain Ischemia/physiopathology , Brain/blood supply , Brain/metabolism , Cerebrovascular Circulation , Hemodynamics , Magnetic Resonance Imaging/methods , Animals , Brain Ischemia/diagnosis , Hemoglobins/metabolism , Humans , Models, Biological , Oxygen/blood , Oxygen Consumption , Parietal Lobe/blood supply , Rats , Rats, Wistar , Sensitivity and Specificity
11.
Magn Reson Med ; 44(6): 833-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108619

ABSTRACT

Interrelation of T(1) and diffusion of water was studied in rat models of acute global and focal cerebral ischemia. Cortical T(1), as quantified with an inversion recovery method, increased by 4-7% within a few minutes of global ischemia at 4.7 and 9.4 T, but a significantly smaller change was detected at 1.5 T. The initial T(1) change occurred within seconds of cardiac arrest, much earlier than the extensive diffusion drop after 1-2 min. Thus, the initial increase in T(1) upon acute cerebral ischemia is directly caused by cessation of blood flow. In transient middle cerebral artery occlusion (MCAO), prolonged T(1) relaxation was detected within 10 min, with a subsequent increase during the course of ischemia. Spin density did not change during the first hour, showing that T(1) increase was not caused by net accumulation of water. Interestingly, partial recovery of T(1) upon release of MCAO, occurring independent of long-term tissue outcome, was observed only in concert with diffusion recovery.


Subject(s)
Body Water/metabolism , Brain Ischemia/diagnosis , Brain/metabolism , Magnetic Resonance Imaging/methods , Acute Disease , Analysis of Variance , Animals , Brain/pathology , Brain Ischemia/metabolism , Diffusion , Disease Models, Animal , Infarction, Middle Cerebral Artery/diagnosis , Infarction, Middle Cerebral Artery/metabolism , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/statistics & numerical data , Male , Rats , Rats, Wistar , Reperfusion Injury/diagnosis , Reperfusion Injury/metabolism , Time Factors
12.
J Cereb Blood Flow Metab ; 20(10): 1457-66, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11043908

ABSTRACT

The impact of brain imaging on the assessment of tissue status is likely to increase with the advent of treatment methods for acute cerebral ischemia. Multimodal magnetic resonance imaging (MRI) demonstrates potential for selecting stroke therapy patients by identifying the presence of acute ischemia, delineating the perfusion defect, and excluding hemorrhage. Yet, the identification of tissue subject to reversible or irreversible ischemia has proven to be difficult. Here, the authors show that T1 relaxation time in the rotating frame, so-called T1rho, serves as a sensitive MRI indicator of cerebral ischemia in the rat. The T1rho prolongs within minutes after a drop in the CBF of less than 22 mL 100 g(-1) min(-1). Dependence of T1rho on spin-lock amplitude, termed as T1rho dispersion, increases by approximately 20% on middle cerebral artery (MCA) occlusion, comparable with the magnitude of diffusion reduction. The T1rho dispersion change dynamically increases to be 38% +/- 10% by the first 60 minutes of ischemia in the brain region destined to develop infarction. Following reperfusion after 45 minutes of MCA occlusion, the tissue with elevated T1rho dispersion (yet normal diffusion) develops severe histologically verified neuronal damage; thus, the former parameter unveils an irreversible condition earlier than currently available MRI methods. The T1rho dispersion as a novel MRI index of cerebral ischemia may be useful in determination of the therapeutic window for acute ischemic stroke.


Subject(s)
Brain Ischemia/diagnosis , Magnetic Resonance Imaging/methods , Animals , Arterial Occlusive Diseases/diagnosis , Arterial Occlusive Diseases/pathology , Arterial Occlusive Diseases/physiopathology , Body Temperature , Brain/physiopathology , Brain Ischemia/physiopathology , Cerebral Arteries , Cerebrovascular Circulation , Male , Nerve Tissue Proteins/metabolism , Phantoms, Imaging , Rats , Rats, Wistar , Time Factors
13.
Trends Biochem Sci ; 25(8): 357-62, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10916153

ABSTRACT

A functionally and metabolically interesting class of cell lipid can be observed by 1H nuclear magnetic resonance (NMR) spectroscopy in situ. These prominent resonances are not only associated with malignancy and cell death, but also act as heralds of benign processes, such as cell activation and proliferation. Originally, these NMR observations were explained with a membrane lipid microdomain model. However, recent studies have identified intracellular droplets, so called lipid bodies, as important contributors to these resonances. This finding bears novel implications for our understanding and assessment of lipid biochemistry in the life and death of cells.


Subject(s)
Lipids/physiology , Magnetic Resonance Spectroscopy , Triglycerides/metabolism , Animals , Apoptosis , Brain Neoplasms/pathology , Cell Division , Cell Transformation, Neoplastic , Humans , Lipid Bilayers/metabolism , Necrosis
14.
Mol Cell Neurosci ; 16(2): 87-96, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10924253

ABSTRACT

It has been suggested that the increased production of endogenous BDNF after brain insults supports the survival of injured neurons and limits the spread of the damage. In order to test this hypothesis experimentally, we have produced transgenic mouse lines that overexpress the dominant-negative truncated splice variant of BDNF receptor trkB (trkB.T1) in postnatal cortical and hippocampal neurons. When these mice were exposed to transient focal cerebral ischemia by occluding the middle cerebral artery for 45 min and the damage was assessed 24 h later, transgenic mice had a significantly larger damage than wild-type littermates in the cerebral cortex (204 +/- 32% of wild-type, P = 0.02), but not in striatum, where the transgene is not expressed. Our results support the notion that endogenously expressed BDNF is neuroprotective and that BDNF signaling may have an important role in preventing brain damage after transient ischemia.


Subject(s)
Ischemic Attack, Transient/genetics , Neurons/physiology , Receptor, trkB/genetics , Alternative Splicing/physiology , Animals , Brain Chemistry/genetics , Brain-Derived Neurotrophic Factor/genetics , Gene Expression/physiology , Genetic Predisposition to Disease , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Transgenic , Mutagenesis/physiology , Neurons/chemistry , RNA, Messenger/analysis
15.
In Vivo ; 14(3): 393-400, 2000.
Article in English | MEDLINE | ID: mdl-10904872

ABSTRACT

BACKGROUND: Human renal cell carcinoma (RCC) is the most common kidney malignancy with significant mortality. Human tumor xenograft models are important tools for cancer research. MATERIALS AND METHODS: We have established and characterized a new animal model for human RCC using Caki-2 cells implanted into the renal subcapsule (RSC) of nude mice. Histology, immunocytochemistry, in situ hybridization and magnetic resonance imaging (MRI) were used to analyze the tumors. RESULTS: The implantations generated reproducible carcinomas which closely resemble human RCC. The tumors showed cystic-papillary structures, rich capillary network and fibro-septa formations. Proliferation varied from 0-5% and from 1-60% in cystic and solid areas, respectively. Apoptosis was less than 1%. Macrophages and other inflammatory cell infiltrations were detected in the tumors. VEGF-A and angiopoietin I were expressed in a small number of cells in large tumors. Tumors did not metastasize outside peritoneal cavity. Survival of the tumor bearing animals was 23 +/- 3 weeks. CONCLUSIONS: It is concluded that Caki-2 carcinomas implanted into renal subcapsule of nude mice resemble human RCC in several aspects and represent a good animal model for studies regarding the pathogenesis and treatment of human RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Disease Models, Animal , Kidney Neoplasms/pathology , Animals , Apoptosis , Carcinoma, Renal Cell/classification , Carcinoma, Renal Cell/metabolism , Humans , Ki-67 Antigen/analysis , Kidney Neoplasms/classification , Kidney Neoplasms/metabolism , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Tumor Cells, Cultured
16.
J Cereb Blood Flow Metab ; 20(2): 316-26, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10698069

ABSTRACT

The ability of transverse nuclear magnetic resonance relaxation time, T2, to reveal acutely reduced CBF was assessed using magnetic resonance imaging (MRI). Graded reduction of CBF was produced in rats using a modification of Pulsinelli's four-vessel occlusion model. The CBF in cerebral cortex was quantified using the hydrogen clearance method, and both T2 and the trace of the diffusion tensor (Dav = 1/3TraceD) in the adjacent cortical tissue were determined as a function of reduced CBF at 4.7 T. A previously published theory, interrelating cerebral hemodynamic parameters, hemoglobin, and oxygen metabolism with T2, was used to estimate the effects of reduced CBF on cerebral T2. The MRI data show that T2 reduces in a U-shape manner as a function of CBF, reaching a level that is 2.5 to 2.8 milliseconds (5% to 6%) below the control value at CBF, between 15% and 60% of normal. This reduction could be estimated by the theory using the literature values of cerebral blood volume, oxygen extraction ratio, and precapillary oxygen extraction during compromised CBF. Dav dropped with two apparent flow thresholds, so that a small 11% to 17% reduction occurred between CBF values of 16% to 45% of normal, followed by a precipitous collapse by more than 20% at CBF below 15% of normal. The current data show that T2 can be used as an indicator of acute hypoperfusion because of its ability to indicate blood oxygenation level-dependent phenomena on reduced CBF.


Subject(s)
Brain Ischemia/physiopathology , Cerebrovascular Circulation/physiology , Computer Simulation , Magnetic Resonance Spectroscopy/methods , Models, Cardiovascular , Animals , Brain/blood supply , Brain/metabolism , Male , Oxygen/analysis , Oxygen/metabolism , Rats , Rats, Wistar , Reaction Time/physiology
17.
J Cereb Blood Flow Metab ; 20(2): 405-11, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10698079

ABSTRACT

Proton (1H) nuclear magnetic resonance (NMR) diffusion spectroscopy was used to assess apparent diffusion coefficients (ADCs) in rat brain slices. Aglycemic hypoxia caused reductions in the ADC of N-acetylaspartate (NAA) (0.15 to 0.09 x 10(-3) mm2/s) and "slow" diffusion coefficient (D2) of tissue water (0.51 to 0.37 x 10(-3) mm2/s), together with a 32+/-11% increase in tissue water volume, attributable to tissue swelling. The ADC and D2 reductions were diminished, however, by removing external Ca2+, and under 10 mmol/L Mg2+, normoxic diffusion coefficients persisted until 40 minutes of hypoxia. The data suggest that the shift of water into the intracellular space alone cannot satisfactorily explain the reduced cerebral diffusion upon energy failure and that external Mg2+ and Ca2+ play crucial modulatory roles.


Subject(s)
Cerebral Cortex/metabolism , Energy Metabolism/physiology , Water/metabolism , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Calcium/pharmacokinetics , Cations/pharmacokinetics , Diffusion , Hypoxia, Brain/metabolism , In Vitro Techniques , Magnesium/pharmacokinetics , Magnetic Resonance Spectroscopy , Male , Membrane Potentials/drug effects , Potassium Chloride/pharmacology , Protons , Rats , Rats, Wistar , Sodium Chloride/pharmacology
18.
J Cereb Blood Flow Metab ; 19(12): 1289-95, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10598932

ABSTRACT

The oxygen extraction ratio (OER) of a tissue describes the interplay between oxygen delivery and consumption and, as such, directly reflects the viability and activity of any organ. It is shown that OER can be quantified using a single magnetic resonance imaging observable, namely the relaxation time T2 of venous blood draining from the tissue. This principle is applied to study local OER changes in the brain on visual stimulation in humans, unambiguously demonstrating a mismatch between changes in blood flow and oxygen metabolism on activation.


Subject(s)
Brain/physiology , Cerebrovascular Circulation , Magnetic Resonance Imaging/methods , Oxygen Consumption , Oxygen/blood , Brain/blood supply , Hematocrit , Humans , Photic Stimulation
20.
Magn Reson Med ; 42(4): 617-26, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10502748

ABSTRACT

The spin-echo response to visual activation was studied as a function of spatial resolution at a field of 1.5 T. The results showed that the increase in absolute T(2) upon activation was as large as 22.8 +/- 3.1% (P < 0.05) at the highest resolution (5.3 mm(3)), while it was as small as 3.5 +/- 0.2% (P < 0.05) at the lowest resolution (42.2 mm(3)). In addition, upon increasing resolution, the spin-echo signal decay as a function of echo time changed from monoexponential to nonexponential. These data indicate that, when using the standard resolution for fMRI studies at 1.5 T, the effects of spin-echo changes in the draining veins are of major contribution to the total blood oxygenation level-dependent (BOLD) signal changes measured in voxels encompassing the activated brain areas. The data can be quantitatively accounted for using a model based on the intravascular origin of the spin-echo effect including both macrovascular and microvascular effects. Existing theories for the spin-echo BOLD effect based on diffusion through field gradients predict negligible spin-echo effects inside the large vessels and are therefore incompatible with the data. Magn Reson Med 42:617-626, 1999.


Subject(s)
Evoked Potentials, Visual/physiology , Magnetic Resonance Imaging/methods , Visual Cortex/blood supply , Brain/physiology , Cerebral Veins/anatomy & histology , Cerebral Veins/physiology , Humans , Microcirculation/physiology , Oxygen/blood , Visual Cortex/physiology
SELECTION OF CITATIONS
SEARCH DETAIL