Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Vaccines (Basel) ; 9(11)2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34835167

ABSTRACT

Development of a vaccine that can elicit robust HIV specific antibody responses in the mucosal compartments is desired for effective prevention of HIV via sexual transmission. However, the current mucosal vaccines have either poor immunogenicity when administered orally or invite safety concerns when administered intranasally. Sublingual immunization has received more attention in recent years based on its efficiency in inducing systemic and mucosal immune responses in both mucosal and extra-mucosal tissues. To facilitate the transport of the immunogen across the sub-mucosal epithelial barrier, we found that CD91, the receptor of C1q, is prevalently expressed in the sublingual mucosal lining, and thus, a modified chimeric C1q surface conjugated CD40L/HIV VLP was generated. The ability of this chimeric C1q/CD40L/HIV VLP to bind, cross the epithelial layer, access and activate the sub-mucosal layer dendritic cells (DCs), and ultimately induce enhanced mucosal and systemic immune responses against HIV is evaluated in this study. We found that C1q/CD40L/HIV VLPs have enhanced binding, increased transport across the epithelial layer, and upregulate DC activation markers as compared to CD40L/HIV VLPs alone. Mice immunized with C1q/CD40L/HIV VLPs by sublingual administration showed higher levels of IgA salivary antibodies against both HIV Gag and Env than mice immunized with CD40L/HIV VLPs. Moreover, sublingual immunization with C1q/CD40L/HIV VLPs induced more Env- and Gag-specific IFN-γ producing T cells than the CD40L/HIV VLPs group. Interestingly, C1q/CD40L/HIV VLP immunization can also induce more mucosal homing T cells than that in CD40L/HIV VLP group. Our data suggest that incorporation of C1q to CD40L/HIV VLPs is a promising novel strategy and that the sublingual immunization can be a favorite immunization route for HIV mucosal vaccines.

2.
Vaccines (Basel) ; 8(2)2020 Jun 06.
Article in English | MEDLINE | ID: mdl-32517277

ABSTRACT

Studies have shown that blockade of CTLA-4 promoted the expansion of germinal center B-cells in viral infection or immunization with model antigens. Few studies have evaluated the immunological consequences of CTLA-4 blockade during immunization against relevant vaccine candidates. Here, we investigated the effects of CTLA-4 blockade on HIV virus-like particles (VLPs) vaccination in a C57BL/6J mouse model. We found that CTLA-4 blockade during HIV VLP immunization resulted in increased CD4+ T-cell activation, promoted the expansion of HIV envelope (Env)-specific follicular helper T cell (Tfh) cells, and significantly increased HIV Gag- and Env-specific IgG with higher avidity and antibody-dependent cellular cytotoxicity (ADCC) capabilities. Furthermore, after only a single immunization, CTLA-4 blockade accelerated T-cell dependent IgG class switching and the induction of significantly high serum levels of the B-cell survival factor, A proliferation-inducing ligand (APRIL). Although no significant increase in neutralizing antibodies was observed, increased levels of class-switched Env- and Gag-specific IgG are indicative of increased polyclonal B-cell activation, which demonstrated the ability to mediate and enhance ADCC in this study. Altogether, our findings show that CTLA-4 blockade can increase the levels of HIV antigen-specific B-cell and antigen-specific Tfh cell activity and impact humoral immune responses when combined with a clinically relevant HIV VLP-based vaccine.

3.
PLoS One ; 14(3): e0213294, 2019.
Article in English | MEDLINE | ID: mdl-30921351

ABSTRACT

Clinical trials of EGFR inhibitors in combination with gemcitabine for the treatment of pancreatic ductal adenocarcinoma (PDAC) have generated mixed results partially due to the poorly defined effectiveness of EGFR inhibitors in PDAC. Here, we studied a panel of PDAC cell lines to compare the IC50s of the EGFR inhibitors gefitinib and cetuximab. We found that gefitinib induced biphasic inhibition in over 50% of PDAC cells, with the initial growth inhibition occurring at nanomolar concentrations and a second growth inhibition occurring outside the clinical range. In contrast to gefitinib, cetuximab produced a single phase growth inhibition in a subset of PDAC cells. Using this sensitivity data, we screened for correlations between cell morphology proteins and EGFR ligands to EGFR inhibitor sensitivity, and found that mesothelin and the EGFR ligand TGF-α have a strong correlation to gefitinib and cetuximab sensitivity. Analysis of downstream signaling pathways indicated that plc-γ1 and c-myc were consistently inhibited by EGFR inhibitor treatment in sensitive cell lines. While an inconsistent additive effect was observed with either cetuximab or gefitinib in combination with gemcitabine, the cell pathway data indicated consistent ERK activation, leading us to pursue EGFR inhibitors in combination with trametinib, a MEK1/2 inhibitor. Both cetuximab and gefitinib in combination with trametinib produced an additive effect in all EGFR sensitive cell lines. Our results indicate that mesothelin and TGF-α can predict PDAC sensitivity to EGFR inhibitors and a combination of EGFR inhibitors with trametinib could be a novel effective treatment for PDAC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/pathology , Drug Resistance, Neoplasm , GPI-Linked Proteins/metabolism , Pancreatic Neoplasms/pathology , Transforming Growth Factor alpha/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle , Cell Proliferation , Cetuximab/administration & dosage , ErbB Receptors/antagonists & inhibitors , GPI-Linked Proteins/genetics , Gefitinib/administration & dosage , Humans , Mesothelin , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Signal Transduction , Transforming Growth Factor alpha/genetics , Tumor Cells, Cultured
5.
Vaccine ; 34(48): 5886-5894, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27997339

ABSTRACT

Human Immunodeficiency Virus (HIV) Virus-Like Particles (VLPs) composed of HIVIIIB Gag and HIVBaL gp120/gp41 envelope are a pseudovirion vaccine capable of presenting antigens in their native conformations. To enhance the immunogenicity of the HIV Env antigen, VLPs were coupled to VesiVax Conjugatable Adjuvant Lipid Vesicles (CALV) containing one of four toll-like-receptor (TLR) ligands, each activating a receptor with distinct cellular localization and downstream pathways. C57BL/6 mice were vaccinated by intranasal prime followed by two sub-cheek boosts and their sera immunoglobulin and neutralizing potency were measured over a duration of 3months after vaccination. PBS control, VLPs alone, CALV+VLPs, and VLPs complexed with CALV and ligands for TLR2 (PAM3CAG), TLR3 (dsRNA), TLR4 (MPLA), or TLR7/8 (resiquimod) were evaluated based on antibody titer, IgG1 and IgG2c class switching, germinal center formation, T follicular cells and potency of neutralizing antibodies. Consistently, the TLR3 ligand dsRNA complexed to CALV and in combination with VLPs (CALV(dsRNA)+VLPs) induced the strongest response. CALV(dsRNA)+VLPs induced the highest titers against the recombinant vaccine antigens clade B Bal gp120 and pr55 Gag. Additionally, CALV(dsRNA)+VLPs induced cross-clade antibodies, represented by high titers of antibody to clade c 96ZM651 gp120. CALV(dsRNA)+VLPs induced predominantly IgG2c over IgG1, a response associated with T helper type 1 (Th1)-like cytokines. In turn, CALV(dsRNA)+VLP immunized mice generated the most potent neutralizing antibodies against HIV strain MN.3. Finally, at time of sacrifice, a significant increase in germinal center B cells and T follicular cells was detected in mice which received CALV(dsRNA)+VLPs compared to PBS. Our results indicate that CALV(dsRNA) is a superior adjuvant for HIV VLPs in generating a Th1-like immunoglobulin profile, while prolonging lymph node germinal centers, T follicular cells and generating neutralizing antibodies to a highly sensitive tier 1A variant of HIV.


Subject(s)
AIDS Vaccines/immunology , Adjuvants, Immunologic , HIV Infections/prevention & control , Immunity, Humoral , Toll-Like Receptor 3/immunology , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Neutralizing/blood , Gene Products, gag/immunology , Germinal Center/immunology , HIV Antibodies/blood , HIV Infections/immunology , HIV-1/immunology , Ligands , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 3/chemistry , Vaccines, Synthetic/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/chemistry
6.
Oncotarget ; 7(52): 87431-87448, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27911862

ABSTRACT

Semaphorin-3E (Sema3E) is a member of an axon guidance gene family, and has recently been reported to contribute to tumor progression and metastasis. However, its role in pancreatic cancer is yet unknown and uncharacterized. In this study, we showed that Sema3E is overexpressed in human pancreatic cancer, and that high Sema3E levels are associated with tumor progression and poor survival. Interestingly, we also observed Sema3E expression in the nucleus, even though Sema3E is reported to be a secreted protein. Overexpression of Sema3E in pancreatic cancer cells promoted cell proliferation and migration in vitro, and increased tumor incidence and growth in vivo. Conversely, knockout of Sema3E suppressed cancer cell proliferation and migration in vitro, and reduced tumor incidence and size in vivo. Moreover, Sema3E induced cell proliferation via acting through the MAPK/ERK pathway. Collectively, these results reveal an undiscovered role of Sema3E in promoting pancreatic cancer pathogenesis, suggesting that Sema3E may be a suitable prognostic marker and therapeutic target for pancreatic cancer.


Subject(s)
Pancreatic Neoplasms/pathology , Semaphorins/physiology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Humans , MAP Kinase Signaling System/physiology , Mice , Pancreatic Neoplasms/mortality , Semaphorins/analysis
7.
PLoS One ; 10(8): e0136862, 2015.
Article in English | MEDLINE | ID: mdl-26312747

ABSTRACT

HIV virus-like particles (VLPs) present the HIV envelope protein in its native conformation, providing an ideal vaccine antigen. To enhance the immunogenicity of the VLP vaccine, we sought to improve upon two components; the route of administration and the additional adjuvant. Using HIV VLPs, we evaluated sub-cheek as a novel route of vaccine administration when combined with other conventional routes of immunization. Of five combinations of distinct prime and boost sequences, which included sub-cheek, intranasal, and intradermal routes of administration, intranasal prime and sub-cheek boost (IN+SC) resulted in the highest HIV-specific IgG titers among the groups tested. Using the IN+SC regimen we tested the adjuvant VesiVax Conjugatable Adjuvant Lipid Vesicles (CALV) + monophosphoryl lipid A (MPLA) at MPLA concentrations of 0, 7.5, 12.5, and 25 µg/dose in combination with our VLPs. Mice that received 12.5 or 25 µg/dose MPLA had the highest concentrations of Env-specific IgG2c (20.7 and 18.4 µg/ml respectively), which represents a Th1 type of immune response in C57BL/6 mice. This was in sharp contrast to mice which received 0 or 7.5 µg MPLA adjuvant (6.05 and 5.68 µg/ml of IgG2c respectively). In contrast to IgG2c, MPLA had minor effects on Env-specific IgG1; therefore, 12.5 and 25 µg/dose of MPLA induced the optimal IgG1/IgG2c ratio of 1.3. Additionally, the percentage of germinal center B cells increased significantly from 15.4% in the control group to 31.9% in the CALV + 25 µg MPLA group. These mice also had significantly more IL-2 and less IL-4 Env-specific CD8+ T cells than controls, correlating with an increased percentage of Env-specific central memory CD4+ and CD8+ T cells. Our study shows the strong potential of IN+SC as an efficacious route of administration and the effectiveness of VLPs combined with MPLA adjuvant to induce Env specific Th1-oriented HIV-specific immune responses.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Immunization, Secondary/methods , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/immunology , Drug Administration Routes , Female , HIV-1/immunology , HIV-1/ultrastructure , Immunoglobulin G/immunology , Injections, Intradermal , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Mice, Inbred C57BL , Particle Size , Thy-1 Antigens/immunology , Toll-Like Receptor 4/immunology
9.
J Biol Chem ; 289(34): 23318-28, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24986863

ABSTRACT

Mutations in the cytosolic NADP(+)-dependent isocitrate dehydrogenase (IDH1) occur in several types of cancer, and altered cellular metabolism associated with IDH1 mutations presents unique therapeutic opportunities. By altering IDH1, these mutations target a critical step in reductive glutamine metabolism, the metabolic pathway that converts glutamine ultimately to acetyl-CoA for biosynthetic processes. While IDH1-mutated cells are sensitive to therapies that target glutamine metabolism, the effect of IDH1 mutations on reductive glutamine metabolism remains poorly understood. To explore this issue, we investigated the effect of a knock-in, single-codon IDH1-R132H mutation on the metabolism of the HCT116 colorectal adenocarcinoma cell line. Here we report the R132H-isobolome by using targeted (13)C isotopomer tracer fate analysis to trace the metabolic fate of glucose and glutamine in this system. We show that introduction of the R132H mutation into IDH1 up-regulates the contribution of glutamine to lipogenesis in hypoxia, but not in normoxia. Treatment of cells with a d-2-hydroxyglutarate (d-2HG) ester recapitulated these changes, indicating that the alterations observed in the knocked-in cells were mediated by d-2HG produced by the IDH1 mutant. These studies provide a dynamic mechanistic basis for metabolic alterations observed in IDH1-mutated tumors and uncover potential therapeutic targets in IDH1-mutated cancers.


Subject(s)
Cell Hypoxia , Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Neoplasms/enzymology , Cell Line, Tumor , Glycolysis , HCT116 Cells , Humans , Isocitrate Dehydrogenase/metabolism , Mitochondria/physiology , Neoplasms/pathology
10.
Brain Res ; 1551: 45-58, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24440774

ABSTRACT

Reactive astrogliosis is an essential feature of astrocytic response to all forms of central nervous system (CNS) injury and disease, which may benefit or harm surrounding neural and non-neural cells. Despite extensive study, its molecular triggers remain largely unknown in term of ischemic stroke. In the current study we investigated the role p38 mitogen-activated protein kinase (MAPK) in astrogliosis both in vitro and in vivo. In a mouse model of middle cerebral artery occlusion (MCAO), p38 MAPK activation was observed in the glia scar area, along with increased glial fibrillary acidic protein (GFAP) expression. In primary astrocyte cultures, hypoxia and scratch injury-induced astrogliosis was attenuated by both p38 inhibition and knockout of p38 MAPK. In addition, both knockout and inhibition of p38 MAPK also reduced astrocyte migration, but did not affect astrocyte proliferation. In a mouse model of permanent MCAO, no significant difference in motor function recovery and lesion volume was observed between conditional GFAP/p38 MAPK knockout mice and littermates. While a significant reduction of astrogliosis was observed in the GFAP/p38 knockout mice compared with the littermates. Our findings suggest that p38 MAPK signaling pathway plays an important role in the ischemic stroke-induced astrogliosis and thus may serve as a novel target to control glial scar formation.


Subject(s)
Astrocytes/enzymology , Brain Ischemia/enzymology , Gliosis/enzymology , Stroke/enzymology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Astrocytes/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Gliosis/etiology , Gliosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stroke/complications , Stroke/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Appl Toxicol ; 34(8): 903-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24037965

ABSTRACT

Cadmium is a toxic metal with no biological function in higher-order mammals. Humans are exposed to cadmium environmental contamination and the mechanism underlying the cadmium's cytotoxicity is unclear. To better understand this mechanism, we employed murine hippocampal HT-22 cells to test the in vitro effects of cadmium toxicity. Our study indicated that cadmium inhibits both mitochondria oxidative phosphorylation and glycolysis. In turn, this causes depolarization of mitochondrial membrane potential, increase of superoxide production and decrease of ATP generation. Furthermore, we demonstrated that the detrimental action of cadmium in bioenergetics could be mitigated by pyruvate, an intermediate metabolic product. Pyruvate decreased superoxide production, maintained mitochondrial membrane potential, restored glycolysis, mitigated the decrease in cellular ATP and attenuated cadmium cytotoxicity. Our study provides the first evidence that pyruvate might offer promising therapy for cadmium poisoning.


Subject(s)
Cadmium/toxicity , Hippocampus/cytology , Hippocampus/drug effects , Pyruvic Acid/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Glycolysis/drug effects , Hippocampus/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Oxidative Stress/drug effects , Phosphorylation/drug effects , Superoxides/antagonists & inhibitors , Superoxides/metabolism
12.
Brain Res ; 1503: 97-107, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23399685

ABSTRACT

Emerging evidence suggests a decline of ERß expression in various peripheral cancers. ERß has been proposed as a cancer brake that inhibits tumor proliferation. In the current study, we have identified ERß5 as the predominant isoform of ERß in human glioma and its expression was significantly increased in human glioma as compared with non-neoplastic brain tissue. Hypoxia and activation of hypoxia inducible factor (HIF) increased ERß transcription in U87 cells, suggesting elevated ERß expression in glioma might be induced by the hypoxic stress in the tumor. Over-expression of either ERß1 or ERß5 increased PTEN expression and inhibited activation of the PI3K/AKT/mTOR pathway. In addition, ERß5 inhibited the MAPK/ERK pathway. In U87 cells, ERß1 and ERß5 inhibit cell proliferation and reduced cells in the S+G2/M phase. Our findings suggest hypoxia induced ERß5 expression in glioma as a self-protective mechanism against tumor proliferation and that ERß5 might serve as a therapeutic target for the treatment of glioma.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glioma/metabolism , Glioma/pathology , Analysis of Variance , Cell Cycle , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Estrogen Receptor beta/classification , Estrogen Receptor beta/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Oxygen Consumption/physiology , RNA, Messenger , Signal Transduction/physiology , Transfection
13.
J Biol Chem ; 288(13): 9153-64, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23408428

ABSTRACT

Glioblastoma multiforme (GBM), like most cancers, possesses a unique bioenergetic state of aerobic glycolysis known as the Warburg effect. Here, we documented that methylene blue (MB) reverses the Warburg effect evidenced by the increasing of oxygen consumption and reduction of lactate production in GBM cell lines. MB decreases GBM cell proliferation and halts the cell cycle in S phase. Through activation of AMP-activated protein kinase, MB inactivates downstream acetyl-CoA carboxylase and decreases cyclin expression. Structure-activity relationship analysis demonstrated that toluidine blue O, an MB derivative with similar bioenergetic actions, exerts similar action in GBM cell proliferation. In contrast, two other MB derivatives, 2-chlorophenothiazine and promethazine, exert no effect on cellular bioenergetics and do not inhibit GBM cell proliferation. MB inhibits cell proliferation in both temozolomide-sensitive and -insensitive GBM cell lines. In a human GBM xenograft model, a single daily dosage of MB does not activate AMP-activated protein kinase signaling, and no tumor regression was observed. In summary, the current study provides the first in vitro proof of concept that reversal of Warburg effect might be a novel therapy for GBM.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Adenosine Triphosphate/metabolism , Animals , Annexin A5/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Energy Metabolism , Female , Humans , Lactates/metabolism , Methylene Blue/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Oxygen Consumption , Structure-Activity Relationship , Temozolomide , Tolonium Chloride/pharmacology
14.
PLoS One ; 7(10): e48279, 2012.
Article in English | MEDLINE | ID: mdl-23118969

ABSTRACT

Methylene blue (MB), the first lead chemical structure of phenothiazine and other derivatives, is commonly used in diagnostic procedures and as a treatment for methemoglobinemia. We have previously demonstrated that MB could function as an alternative mitochondrial electron transfer carrier, enhance cellular oxygen consumption, and provide protection in vitro and in rodent models of Parkinson's disease and stroke. In the present study, we investigated the structure-activity relationships of MB in vitro using MB and six structurally related compounds. MB reduces mitochondrial superoxide production via alternative electron transfer that bypasses mitochondrial complexes I-III. MB mitigates reactive free radical production and provides neuroprotection in HT-22 cells against glutamate, IAA and rotenone toxicity. Distinctly, MB provides no protection against direct oxidative stress induced by glucose oxidase. Substitution of a side chain at MB's 10-nitrogen rendered a 1000-fold reduction of the protective potency against glutamate neurototoxicity. Compounds without side chains at positions 3 and 7, chlorophenothiazine and phenothiazine, have distinct redox potentials compared to MB and are incapable of enhancing mitochondrial electron transfer, while obtaining direct antioxidant actions against glutamate, IAA, and rotenone insults. Chlorophenothiazine exhibited direct antioxidant actions in mitochondria lysate assay compared to MB, which required reduction by NADH and mitochondria. MB increased complex IV expression and activity, while 2-chlorphenothiazine had no effect. Our study indicated that MB could attenuate superoxide production by functioning as an alternative mitochondrial electron transfer carrier and as a regenerable anti-oxidant in mitochondria.


Subject(s)
Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Methylene Blue/analogs & derivatives , Methylene Blue/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Animals , Cell Line , Electron Transport/drug effects , Electron Transport Complex IV/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Thiazines/pharmacology
15.
PLoS One ; 7(10): e46585, 2012.
Article in English | MEDLINE | ID: mdl-23056355

ABSTRACT

By restoring mitochondrial function, methylene blue (MB) is an effective neuroprotectant in many neurological disorders (e.g., Parkinson's and Alzheimer's diseases). MB has also been proposed as a brain metabolic enhancer because of its action on mitochondrial cytochrome c oxidase. We used in vitro and in vivo approaches to determine how MB affects brain metabolism and hemodynamics. For in vitro, we evaluated the effect of MB on brain mitochondrial function, oxygen consumption, and glucose uptake. For in vivo, we applied neuroimaging and intravenous measurements to determine MB's effect on glucose uptake, cerebral blood flow (CBF), and cerebral metabolic rate of oxygen (CMRO(2)) under normoxic and hypoxic conditions in rats. MB significantly increases mitochondrial complex I-III activity in isolated mitochondria and enhances oxygen consumption and glucose uptake in HT-22 cells. Using positron emission tomography and magnetic resonance imaging (MRI), we observed significant increases in brain glucose uptake, CBF, and CMRO(2) under both normoxic and hypoxic conditions. Further, MRI revealed that MB dramatically increased CBF in the hippocampus and in the cingulate, motor, and frontoparietal cortices, areas of the brain affected by Alzheimer's and Parkinson's diseases. Our results suggest that MB can enhance brain metabolism and hemodynamics, and multimetric neuroimaging systems offer a noninvasive, nondestructive way to evaluate treatment efficacy.


Subject(s)
Brain/metabolism , Hemodynamics , Methylene Blue/metabolism , Cell Line , Humans , Mitochondria/metabolism
16.
Horm Behav ; 60(5): 617-24, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907204

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by dopamine neuron loss in the nigrostriatal pathway that shows greater incidence in men than women. The mechanisms underlying this gender bias remain elusive, although one possibility is that androgens may increase dopamine neuronal vulnerability to oxidative stress. Motor impairment can be modeled in rats receiving a unilateral injection of 6-hydroxydopamine (6-OHDA), a neurotoxin producing nigrostriatal degeneration. To investigate the role of androgens in PD, we compared young (2 months) and aged (24 months) male rats receiving gonadectomy (GDX) and their corresponding intact controls. One month after GDX, rats were unilaterally injected with 6-OHDA, and their motor impairment and asymmetry were assessed 2 weeks later using the cylinder test and the amphetamine-induced rotation test. Plasma samples were also collected to assess the concentration of testosterone and advanced oxidation protein products, a product of oxidative stress. GDX decreased lesion-induced asymmetry along with oxidative stress and increased amphetamine-induced rotations. These results show that GDX improves motor behaviors by decreasing motor asymmetry in 6-OHDA-treated rats, an effect that may be ascribed to increased release of striatal dopamine and decreased oxidative stress. Collectively, the data support the hypothesis that androgens may underlie the gender bias observed in PD.


Subject(s)
Adrenergic Agents/adverse effects , Androgens/pharmacology , Dopaminergic Neurons/drug effects , Motor Activity/drug effects , Neostriatum/drug effects , Oxidopamine/adverse effects , Substantia Nigra/drug effects , Amphetamine/pharmacology , Animals , Male , Orchiectomy , Oxidative Stress/drug effects , Rats , Rotation , Testosterone/blood
17.
J Biol Chem ; 286(18): 16504-15, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21454572

ABSTRACT

Neuroprotective strategies, including free radical scavengers, ion channel modulators, and anti-inflammatory agents, have been extensively explored in the last 2 decades for the treatment of neurological diseases. Unfortunately, none of the neuroprotectants has been proved effective in clinical trails. In the current study, we demonstrated that methylene blue (MB) functions as an alternative electron carrier, which accepts electrons from NADH and transfers them to cytochrome c and bypasses complex I/III blockage. A de novo synthesized MB derivative, with the redox center disabled by N-acetylation, had no effect on mitochondrial complex activities. MB increases cellular oxygen consumption rates and reduces anaerobic glycolysis in cultured neuronal cells. MB is protective against various insults in vitro at low nanomolar concentrations. Our data indicate that MB has a unique mechanism and is fundamentally different from traditional antioxidants. We examined the effects of MB in two animal models of neurological diseases. MB dramatically attenuates behavioral, neurochemical, and neuropathological impairment in a Parkinson disease model. Rotenone caused severe dopamine depletion in the striatum, which was almost completely rescued by MB. MB rescued the effects of rotenone on mitochondrial complex I-III inhibition and free radical overproduction. Rotenone induced a severe loss of nigral dopaminergic neurons, which was dramatically attenuated by MB. In addition, MB significantly reduced cerebral ischemia reperfusion damage in a transient focal cerebral ischemia model. The present study indicates that rerouting mitochondrial electron transfer by MB or similar molecules provides a novel strategy for neuroprotection against both chronic and acute neurological diseases involving mitochondrial dysfunction.


Subject(s)
Electron Transport Chain Complex Proteins/metabolism , Methylene Blue/pharmacology , Mitochondria/enzymology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/drug therapy , Animals , Cell Line , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glycolysis/drug effects , Male , Neurons/enzymology , Neurons/pathology , Oxygen Consumption/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/enzymology , Parkinson Disease, Secondary/pathology , Parkinson Disease, Secondary/physiopathology , Rats , Rats, Sprague-Dawley , Rotenone/adverse effects , Rotenone/pharmacology , Substantia Nigra/enzymology , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Uncoupling Agents/adverse effects , Uncoupling Agents/pharmacology
18.
Brain Res ; 1387: 141-8, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21376707

ABSTRACT

Matrix metalloproteinases (MMPs) are a group of proteinases that degrade components of the extracellular matrix (ECM). There is increasing evidence for a link between the activation of MMPs and Alzheimer's disease (AD) pathogenesis, in which both beneficial and detrimental actions of MMPs have been suggested. It has been demonstrated that MMPs could degrade amyloid ß (Aß) and play important roles in the extracellular Aß catabolism and clearance. On the other hand, MMPs could contribute to AD pathogenesis by compromising the blood brain barrier and promoting neurodegeneration. In the present study, we observed that oligomeric Aß regulates MMP2 expression in a paradoxical manner. In rat primary astrocyte cultures, oligomeric Aß down-regulated MMP2 transcription and reduced its extracellular activity. However, in a widely used mouse model for AD, immunohistochemistry demonstrated an increase of MMP2 expression in astrocytes surrounding senile plaques in APP/PS1 transgenic mice brains. Using real-time PCR, we found that the MMP2 mRNA level was elevated in APP/PS1 transgenic mice brains. In addition, elevated mRNA levels of MMP stimulating cytokines such as IL-1ß and TGFß were found in the brains of APP/PS1 mice. Our study suggests a complex regulation of MMP2 expression by oligomeric Aß in astrocytes. While oligomeric Aß directly down-regulates MMP2 expression and activation in astrocytes, it induces production of proinflammatory cytokines which could serve as strong stimulators for MMP2. Therefore, the ultimate outcome of the oligomeric Aß on MMP2 activation in astrocytes might be the combination of its direct inhibitory action on astrocyte MMP2 expression and the secondary action of inducing inflammatory cytokines.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Matrix Metalloproteinase 2/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Enzyme Activation/physiology , Gene Expression , Gene Expression Regulation , Immunohistochemistry , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
19.
J Neuroinflammation ; 7: 53, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20828397

ABSTRACT

BACKGROUND: Neuroinflammation is a complex process involving cells from the immune system and the central nerve system (CNS). Polymorphonuclear neutrophils (PMN) are the most abundant class of white blood cells, and typically the first type of leukocyte recruited to sites of inflammation. In the CNS, astrocytes are the most abundant glial cell population and participate in the local innate immune response triggered by a variety of insults. In the present study, we investigated the impacts of astrocytes on PMN function. METHODS: Primary astrocyte cultures were derived from postnatal C57BL/6 mice and primary neutrophils were isolated from 8 to 12 weeks old C57BL/6 mice. PMNs respiratory burst was analyzed by H2DCFDA assay. For phagocytosis assay, neutrophils were incubated with FITC-labeled E. coli and the phagocytosis of E coli was determined by flow cytometer. PMNs degranulation was determined by myeloperoxidase assay. Cytokine expression was determined by real-time PCR. To determine the involvement of different signaling pathway, protein lysates were prepared and western blots were conducted to assess the activation of Akt, Erk1/2, and p38. RESULTS: Using ex vivo neutrophils and primary astrocyte cultures, our study demonstrated that astrocytes differentially regulate neutrophil functions, depending upon whether the interactions between the two cell types are direct or indirect. Upon direct cell-cell contact, astrocytes attenuate neutrophil apoptosis, respiratory bust, and degranulation, while enhancing neutrophil phagocytic capability and pro-inflammatory cytokine expression. Through indirect interaction with neutrophils, astrocytes attenuate apoptosis and enhance necrosis in neutrophils, augment neutrophil phagocytosis and respiratory burst, and inhibit neutrophil degranulation. In addition, astrocytes could augment Akt, Erk1/2, and p38 activation in neutrophils. CONCLUSIONS: Astrocytes differentially regulate neutrophil functions through direct or indirect interactions between the two cell types. The diversified actions of astrocytes on neutrophils might provide protection against potential microbial infections given compromised blood-brain barrier integrity under certain neuropathological conditions. The complex actions of astrocytes on neutrophils could provide further insight to harness the inflammatory response to promote CNS repair.


Subject(s)
Astrocytes/metabolism , Cell Communication/immunology , Neutrophils/metabolism , Respiratory Burst/immunology , Analysis of Variance , Animals , Astrocytes/cytology , Astrocytes/immunology , Blotting, Western , Cells, Cultured , Coculture Techniques , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Neutrophil Activation/immunology , Neutrophils/cytology , Neutrophils/immunology , Phagocytosis/immunology , Reverse Transcriptase Polymerase Chain Reaction
20.
Neuropharmacology ; 56 Suppl 1: 213-25, 2009.
Article in English | MEDLINE | ID: mdl-18762202

ABSTRACT

Through a multidisciplinary approach involving experimental and computational studies, we address quantitative aspects of signaling mechanisms triggered in the cell by the receptor targets of hallucinogenic drugs, the serotonin 5-HT2A receptors. To reveal the properties of the signaling pathways, and the way in which responses elicited through these receptors alone and in combination with other serotonin receptors' subtypes (the 5-HT1AR), we developed a detailed mathematical model of receptor-mediated ERK1/2 activation in cells expressing the 5-HT1A and 5-HT2A subtypes individually, and together. In parallel, we measured experimentally the activation of ERK1/2 by the action of selective agonists on these receptors expressed in HEK293 cells. We show here that the 5-HT1AR agonist Xaliproden HCl elicited transient activation of ERK1/2 by phosphorylation, whereas 5-HT2AR activation by TCB-2 led to higher, and more sustained responses. The 5-HT2AR response dominated the MAPK signaling pathway when co-expressed with 5-HT1AR, and diminution of the response by the 5-HT2AR antagonist Ketanserin could not be rescued by the 5-HT1AR agonist. Computational simulations produced qualitative results in good agreement with these experimental data, and parameter optimization made this agreement quantitative. In silico simulation experiments suggest that the deletion of the positive regulators PKC in the 5-HT2AR pathway, or PLA2 in the combined 5-HT1A/2AR model greatly decreased the basal level of active ERK1/2. Deletion of negative regulators of MKP and PP2A in 5-HT1AR and 5-HT2AR models was found to have even stronger effects. Under various parameter sets, simulation results implied that the extent of constitutive activity in a particular tissue and the specific drug efficacy properties may determine the distinct dynamics of the 5-HT receptor-mediated ERK1/2 activation pathways. Thus, the mathematical models are useful exploratory tools in the ongoing efforts to establish a mechanistic understanding and an experimentally testable representation of hallucinogen-specific signaling in the cellular machinery, and can be refined with quantitative, function-related information.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hallucinogens/pharmacology , Models, Biological , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Cell Line, Transformed , Computer Simulation , Dose-Response Relationship, Drug , Humans , Protein Binding/drug effects , Radioligand Assay/methods , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT2A/genetics , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...