Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Neuropharmacology ; 165: 107924, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31881169

ABSTRACT

Dysregulation in glucocorticoid stress and accumbal dopamine reward systems can alter reward salience to increase motivational drive in control conditions while contributing to relapse during drug withdrawal. Amphetamine withdrawal is associated with dysphoria and stress hypersensitivity that may be mediated, in part, by enhanced stress-induced corticosterone observed in the ventral hippocampus. Electrical stimulation of the ventral hippocampus enhances accumbal shell dopamine release, establishing a functional connection between these two regions. However, the effects of ventral hippocampal corticosterone on this system are unknown. To address this, a stress-relevant concentration of corticosterone (0.24ng/0.5 µL) or vehicle were infused into the ventral hippocampus of urethane-anesthetized adult male rats in control and amphetamine withdrawn conditions. Accumbal dopamine output was assessed with in vivo chronoamperometry. Corticosterone infused into the ventral hippocampus rapidly enhanced accumbal dopamine output in control conditions, but produced a biphasic reduction of accumbal dopamine output in amphetamine withdrawal. Selectively blocking glucocorticoid-, mineralocorticoid-, or cytosolic receptors prevented the effects of corticosterone. Overall, these results suggest that the ability of corticosterone to alter accumbal dopamine output requires cooperative activation of mineralocorticoid and glucocorticoid receptors in the cytosol, which is dysregulated during amphetamine withdrawal. These findings implicate ventral hippocampal corticosterone in playing an important role in driving neural systems involved in positive stress coping mechanisms in healthy conditions, whereas dysregulation of this system may contribute to relapse during withdrawal.


Subject(s)
Amphetamine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Corticosterone/metabolism , Dopamine/metabolism , Hippocampus/drug effects , Nucleus Accumbens/drug effects , Stress, Psychological/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Corticosterone/administration & dosage , Hippocampus/metabolism , Male , Nucleus Accumbens/metabolism , Rats, Sprague-Dawley , Stress, Psychological/chemically induced
4.
Front Pharmacol ; 8: 193, 2017.
Article in English | MEDLINE | ID: mdl-28443023

ABSTRACT

For metastases in the central nervous system, angiogenesis enhances metastatic potential and promotes progression. Primary factors which drive vessel growth are vascular endothelial growth factor (VEGF) and angiopoietin-2. Preclinical models show inhibition of either factor reduces metastases spread and inhibits growth. This work sets out to answer two questions in a preclinical mouse model. First, whether the combined inhibition of VEGF and angiopoietin-2, reduces passive permeability and limits drug uptake into brain metastases; and second, whether this inhibition reduces metastases burden in brain. We observed combinatorial inhibition of VEGF and angiopoietin-2, decreased (p < 0.05) angiogenesis and vascular branching in an aortic ring assay and decreased (p < 0.05) endothelial wound closure times. Using a brain metastases of breast cancer model (induced by intracardiac injections of brain seeking MDA-MB-231Br cells or 4T1Br cells), we observed, similar to VEGF, angiopoetin-2 expression correlates to increased angiogenesis (p < 0.05) and increased lesion permeability. To determine efficacy, animals were administered bevacizumab plus L1-10 (angiopoietin inhibitor) twice per week until neurological symptoms developed. Lesion permeability significantly decreased by ∼50% (p < 0.05) compared to untreated lesions, but remained ∼25% greater (p < 0.0%) than brain. In subsequent experiments, animals were administered similar regimens but sacrificed on day 32. The number of metastatic lesions developed was significantly (p < 0.001) reduced in the bevacizumab group (56%) and combination group (86%). Lesions' size was reduced in bevacizumab treated lesions (∼67%) and bevacizumab and L1-10 treated lesions (∼78%) developing area < 0.5 mm2. In summary, combinatorial inhibition of VEGF and angiopoietin reduces lesion permeability and brain metastatic burden.

5.
Cancer Res ; 77(2): 238-246, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27815391

ABSTRACT

Tumors residing in the central nervous system (CNS) compromise the blood-brain barrier (BBB) via increased vascular permeability, with the magnitude of changes dependent on the tumor type and location. Current studies determine penetrability of a cancer therapeutic by administering progressively larger molecules until cutoff is observed where little to no tumor accumulation occurs. However, decades-old experimental work and mathematical modeling document methods to calculate both the size of the vascular opening (pore) with solute permeability values. In this study, we updated this classic mathematical modeling approach with quantitative fluorescence microscopy in two preclinical tumor models, allowing simultaneous administration of multiple sized tracers to determine vascular permeability at a resolution of nearly one micron. We observed that three molecules ranging from 100 Da to 70 kDa permeated into a preclinical glioblastoma model at rates proportional to their diffusion in water. This suggests the solutes freely diffused from blood to glioma across vascular pores without steric restriction, which calculates to a pore size of >140 nm in diameter. In contrast, the calculated pore size of a brain metastasis of breast cancer was approximately 10-fold smaller than glioma vasculature. This difference explains why antibodies are effective against glioblastoma but generally fail in brain metastases of breast cancer. On the basis of our observations, we hypothesize that trastuzumab most likely fails in the treatment of brain metastases of breast cancer because of poor CNS penetration, while the similar sized antibody bevacizumab is effective in the same tumor type not because it penetrates the CNS degree better, but because it scavenges VEGF in the vascular compartment, which reduces edema and permeation. Cancer Res; 77(2); 238-46. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/drug therapy , Capillary Permeability/physiology , Models, Theoretical , Neoplasm Metastasis/drug therapy , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms/secondary , Diffusion , Female , Glioma/pathology , Male , Mice , Mice, Nude , Microscopy, Fluorescence , Rats , Rats, Inbred F344
6.
J Neurosci Methods ; 270: 124-131, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27321229

ABSTRACT

BACKGROUND: Measurement of vascular density has significant value in characterizing healthy and diseased tissue, particularly in brain where vascular density varies among regions. Further, an understanding of brain vessel size helps distinguish between capillaries and larger vessels like arterioles and venules. Unfortunately, few cutting edge methodologies are available to laboratories to rapidly quantify vessel density. NEW METHOD: We developed a rapid microscopic method, which quantifies the numbers and diameters of blood vessels in brain. Utilizing this method we characterized vascular density of five brain regions in both mice and rats, in two tumor models, using three tracers. RESULTS: We observed the number of sections/mm(2) in various brain regions: genu of corpus callosum 161±7, hippocampus 266±18, superior colliculus 300±24, frontal cortex 391±55, and inferior colliculus 692±18 (n=5 animals). Regional brain data were not significantly different between species (p>0.05) or when using different tracers (70kDa and 2000kDa Texas Red; p>0.05). Vascular density decreased (62-79%) in preclinical brain metastases but increased (62%) a rat glioma model. COMPARISON WITH EXISTING METHODS: Our values were similar (p>0.05) to published literature. We applied this method to brain-tumors and observed brain metastases of breast cancer to have a ∼2.5-fold reduction (p>0.05) in vessels/mm(2) compared to normal cortical regions. In contrast, vascular density in a glioma model was significantly higher (sections/mm(2) 736±84; p<0.05). CONCLUSIONS: In summary, we present a vascular density counting method that is rapid, sensitive, and uses fluorescence microscopy without antibodies.


Subject(s)
Brain/blood supply , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence/methods , Pattern Recognition, Automated/methods , Animals , Brain/anatomy & histology , Brain/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioma/pathology , Humans , Male , Mice, Transgenic , Neoplasm Metastasis/pathology , Rats, Inbred F344 , Rats, Sprague-Dawley
7.
Front Pharmacol ; 4: 136, 2013.
Article in English | MEDLINE | ID: mdl-24312053

ABSTRACT

The blood-brain barrier (BBB) is a specialized vascular interface that restricts the entry of many compounds into brain. This is accomplished through the sealing of vascular endothelial cells together with tight junction proteins to prevent paracellular diffusion. In addition, the BBB has a high degree of expression of numerous efflux transporters which actively extrude compounds back into blood. However, when a metastatic lesion develops in brain the vasculature is typically compromised with increases in passive permeability (blood-tumor barrier; BTB). What is not well documented is to what degree active efflux retains function at the BTB despite the changes observed in passive permeability. In addition, there have been previous reports documenting both increased and decreased expression of P-glycoprotein (P-gp) in lesion vasculature. Herein, we simultaneously administer a passive diffusion marker ((14)C-AIB) and a tracer subject to P-gp efflux (rhodamine 123) into a murine preclinical model of brain metastases of breast cancer. We observed that the metastatic lesions had similar expression (p > 0.05; n = 756-1214 vessels evaluated) at the BBB and the BTB. Moreover, tissue distribution of R123 was not significantly (p > 0.05) different between normal brain and the metastatic lesion. It is possible that the similar expression of P-gp on the BBB and the BTB contribute to this phenomenon. Additionally we observed P-gp expression at the metastatic cancer cells adjacent to the vasculature which may also contribute to reduced R123 uptake into the lesion. The data suggest that despite the disrupted integrity of the BTB, efflux mechanisms appear to be intact, and may be functionally comparable to the normal BBB. The BTB is a significant hurdle to delivering drugs to brain metastasis.

8.
Mol Cancer Ther ; 12(11): 2389-99, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002934

ABSTRACT

Brain (central nervous system; CNS) metastases pose a life-threatening problem for women with advanced metastatic breast cancer. It has recently been shown that the vasculature within preclinical brain metastasis model markedly restricts paclitaxel delivery in approximately 90% of CNS lesions. Therefore to improve efficacy, we have developed an ultra-small hyaluronic acid (HA) paclitaxel nanoconjugate (∼5 kDa) that can passively diffuse across the leaky blood-tumor barrier and then be taken up into cancer cells (MDA-MB-231Br) via CD44 receptor-mediated endocytocis. Using CD44 receptor-mediated endocytosis as an uptake mechanism, HA-paclitaxel was able to bypass p-glycoprotein-mediated efflux on the surface of the cancer cells. In vitro cytoxicity of the conjugate and free paclitaxel were similar in that they (i) both caused cell-cycle arrest in the G2-M phase, (ii) showed similar degrees of apoptosis induction (cleaved caspase), and (iii) had similar IC50 values when compared with paclitaxel in MTT assay. A preclinical model of brain metastases of breast cancer using intracardiac injections of Luc-2 transfected MDA-MB-231Br cells was used to evaluate in vivo efficacy of the nanoconjugate. The animals administered with HA-paclitaxel nanoconjugate had significantly longer overall survival compared with the control and the paclitaxel-treated group (P < 0.05). This study suggests that the small molecular weight HA-paclitaxel nanoconjugates can improve standard chemotherapeutic drug efficacy in a preclinical model of brain metastases of breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Hyaluronic Acid/pharmacology , Nanoconjugates , Paclitaxel/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/pathology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/pharmacokinetics , MCF-7 Cells , Mammary Neoplasms, Experimental , Mice, Nude , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use
9.
J Neurosci Methods ; 219(1): 188-95, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-23916719

ABSTRACT

Quantitative fluorescent microscopy is an emerging technology that has provided significant insight into cellular dye accumulation, organelle function, and tissue physiology. However, historically dyes have only been used to qualitatively or semi-quantitatively (fold change) determine changes in blood-brain barrier (BBB) integrity. Herein, we present a novel method to calculate the blood to brain transfer rates of the dyes rhodamine 123 and Texas red across the in situ BBB. We observed that rhodamine 123 is subject to p-glycoprotein mediated efflux at the rat BBB and can be increased nearly 20-fold with p-glycoprotein inhibition. However, Texas Red appears to not be subject to MRP2 mediated efflux at the rat BBB, agreeing with literature reports suggesting MRP2 may lack functionality at the normal rat BBB. Lastly, we present data demonstrating that once dyes have crossed the BBB, diffusion of the dye molecule is not as instantaneous as has been previously suggested. We propose that future work can now be completed to (1) match BBB transfer coefficients to interstitial diffusion constants and (2) use dyes with specific affinities to cellular organelles or that have specific properties (e.g., subject to efflux transporters) to more fully understand BBB physiology.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Algorithms , Animals , Autoradiography , Calcium Channel Blockers/pharmacology , Cells, Cultured , Data Interpretation, Statistical , Fluorescent Dyes , Green Fluorescent Proteins , Image Processing, Computer-Assisted , Kinetics , Linear Models , Male , Microscopy, Fluorescence , Perfusion , Propionates/pharmacology , Quinolines/pharmacology , Rats , Rats, Inbred F344 , Reference Standards , Rhodamine 123 , Verapamil/pharmacology , Xanthenes
10.
J Neurochem ; 115(6): 1495-507, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20950334

ABSTRACT

Endothelial tight junctions and efflux transporters of the blood-brain barrier (BBB) significantly limit brain accumulation of many drugs, including protease inhibitors such as saquinavir. The cholinergic agonist nicotine is one of the most commonly used drugs in the world and the incidence is even higher in the human immune deficiency virus population (∼ 70%). We examined the ability of nicotine and its primary metabolite cotinine to modify brain uptake of saquinavir in rats. Both nicotine and cotinine at pharmacological concentrations matching those in smokers, increased brain saquinavir uptake by two fold. Co-perfusion with nicotinic receptor antagonists and passive permeability markers showed that the effect was not caused by receptor activation or BBB permeability disruption. Transport inhibition studies demonstrated that brain saquinavir uptake is limited by multiple efflux transporters, P-glycoprotein (P-gp), breast cancer resistance protein and multidrug resistance-associated protein. In situ perfusion and in vitro experiments using a classical P-gp substrate rhodamine 123 linked the effect of nicotine to inhibition of BBB P-gp transport. The effect was confirmed in vivo in chronic 14 day nicotine administration animals. These data suggest nicotine increases antiretroviral drug exposure to brain and may represent a significant in vivo drug-drug interaction at the BBB. Although this may slightly benefit CNS antiretroviral efficacy, it may also expose the brain to potential serious neurotoxicity.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Cotinine/metabolism , HIV Protease Inhibitors/metabolism , Saquinavir/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Blood-Brain Barrier/drug effects , Brain/drug effects , Cell Line, Tumor , Cotinine/administration & dosage , Dose-Response Relationship, Drug , Drug Synergism , HIV Protease Inhibitors/administration & dosage , Male , Permeability/drug effects , Rats , Rats, Inbred F344 , Saquinavir/administration & dosage
11.
Clin Cancer Res ; 16(23): 5664-78, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20829328

ABSTRACT

PURPOSE: Brain metastases of breast cancer appear to be increasing in incidence, confer significant morbidity, and threaten to compromise gains made in systemic chemotherapy. The blood-tumor barrier (BTB) is compromised in many brain metastases; however, the extent to which this influences chemotherapeutic delivery and efficacy is unknown. Herein, we answer this question by measuring BTB passive integrity, chemotherapeutic drug uptake, and anticancer efficacy in vivo in two breast cancer models that metastasize preferentially to brain. EXPERIMENTAL DESIGN: Experimental brain metastasis drug uptake and BTB permeability were simultaneously measured using novel fluorescent and phosphorescent imaging techniques in immune-compromised mice. Drug-induced apoptosis and vascular characteristics were assessed using immunofluorescent microscopy. RESULTS: Analysis of over 2,000 brain metastases from two models (human 231-BR-Her2 and murine 4T1-BR5) showed partial BTB permeability compromise in greater than 89% of lesions, varying in magnitude within and between metastases. Brain metastasis uptake of ¹4C-paclitaxel and ¹4C-doxorubicin was generally greater than normal brain but less than 15% of that of other tissues or peripheral metastases, and only reached cytotoxic concentrations in a small subset (∼10%) of the most permeable metastases. Neither drug significantly decreased the experimental brain metastatic ability of 231-BR-Her2 tumor cells. BTB permeability was associated with vascular remodeling and correlated with overexpression of the pericyte protein desmin. CONCLUSIONS: This work shows that the BTB remains a significant impediment to standard chemotherapeutic delivery and efficacy in experimental brain metastases of breast cancer. New brain permeable drugs will be needed. Evidence is presented for vascular remodeling in BTB permeability alterations.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Blood-Brain Barrier/drug effects , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Disease Models, Animal , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Humans , Mice , Mice, Nude , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Permeability , Treatment Outcome , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 15(19): 6148-57, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789319

ABSTRACT

PURPOSE: As chemotherapy and molecular therapy improve the systemic survival of breast cancer patients, the incidence of brain metastases increases. Few therapeutic strategies exist for the treatment of brain metastases because the blood-brain barrier severely limits drug access. We report the pharmacokinetic, efficacy, and mechanism of action studies for the histone deactylase inhibitor vorinostat (suberoylanilide hydroxamic acid) in a preclinical model of brain metastasis of triple-negative breast cancer. EXPERIMENTAL DESIGN: The 231-BR brain trophic subline of the MDA-MB-231 human breast cancer cell line was injected into immunocompromised mice for pharmacokinetic and metastasis studies. Pharmacodynamic studies compared histone acetylation, apoptosis, proliferation, and DNA damage in vitro and in vivo. RESULTS: Following systemic administration, uptake of [(14)C]vorinostat was significant into normal rodent brain and accumulation was up to 3-fold higher in a proportion of metastases formed by 231-BR cells. Vorinostat prevented the development of 231-BR micrometastases by 28% (P = 0.017) and large metastases by 62% (P < 0.0001) compared with vehicle-treated mice when treatment was initiated on day 3 post-injection. The inhibitory activity of vorinostat as a single agent was linked to a novel function in vivo: induction of DNA double-strand breaks associated with the down-regulation of the DNA repair gene Rad52. CONCLUSIONS: We report the first preclinical data for the prevention of brain metastasis of triple-negative breast cancer. Vorinostat is brain permeable and can prevent the formation of brain metastases by 62%. Its mechanism of action involves the induction of DNA double-strand breaks, suggesting rational combinations with DNA active drugs or radiation.


Subject(s)
Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma/drug therapy , Carcinoma/pathology , DNA Breaks, Double-Stranded/drug effects , Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Enzyme Inhibitors/pharmacokinetics , Female , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylases , Humans , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured , Vorinostat , Xenograft Model Antitumor Assays
14.
Rev Rhum Mal Osteoartic ; 49(5): 385-91, 1982 Apr.
Article in French | MEDLINE | ID: mdl-7100807

ABSTRACT

Use of an immunoenzyme technique to seek two types of antibody (AB): antihistones and bicatenary anti-RNA, led to study of their distribution in different connective tissue and autoimmune diseases. Total antihistone AB and the H1 and H2b antifractions were seen almost exclusively in lupus (52.5 per cent of cases), principally during active phases of the disease since 87 per cent of active forms had antihistone AB as against 15 per cent of inactive forms. Antihistone AB would thus appear to be a valuable laboratory investigation in the diagnosis and surveillance of lupus. Bicatenary anti-RNA AB were more widely distributed, being found in DLE, RA, scleroderma and in various disorders with antinuclear antibodies. However they were slightly more common in active forms of lupus and their titres appeared to fall more quickly than those of antinuclear AB and anti-DNA AB at the time of clinical improvements. In RA, bicatenary anti-RNA AB were more commonly associated with inflammatory and progressive forms but further studies involving larger populations would be necessary to confirm these findings.


Subject(s)
Autoantibodies/analysis , Connective Tissue Diseases/immunology , Histones/immunology , RNA, Double-Stranded/immunology , Arthritis, Rheumatoid/immunology , Autoimmune Diseases/immunology , Connective Tissue Diseases/diagnosis , Enzyme-Linked Immunosorbent Assay , Humans , Lupus Erythematosus, Systemic/immunology , Scleroderma, Systemic/immunology
15.
Pathol Biol (Paris) ; 30(2): 73-8, 1982 Feb.
Article in French | MEDLINE | ID: mdl-6178071

ABSTRACT

The present paper describes a new ELISA type immunoenzymatic method for the detection of antibodies directed against nDNA, dDNA, and double stranded RNA (poly A, poly U and poly l, poly C). The antigens are adsorbed on glass beads but as some nucleic acids are difficult to adsorb on glass beads, polylysine was used to bind the antigens to the beads. The beads were saturated with sheep serum to get rid possible unspecific bindings. The sera are diluted before use with PBS containing sheep serum. The rate of antinucleic acid antibodies is obtained by subtraction of OD with antigen from OD without antigen. The technique was applied to human sera from patients with various autoimmune diseases (mostly disseminated lupus erythematosus). There is a rather good correlation between the determinations of anti-nDNA antibodies obtained with the ELISA and with indirect IF on Crithidia Luciliae.


Subject(s)
Autoantibodies/analysis , DNA/immunology , RNA/immunology , Adult , Autoimmune Diseases/immunology , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...