Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Anesthesiology ; 125(2): 399-411, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27286478

ABSTRACT

Volatile general anesthetics continue to be an important part of clinical anesthesia worldwide. The impact of volatile anesthetics on the immune system has been investigated at both mechanistic and clinical levels, but previous studies have returned conflicting findings due to varied protocols, experimental environments, and subject species. While many of these studies have focused on the immunosuppressive effects of volatile anesthetics, compelling evidence also exists for immunoactivation. Depending on the clinical conditions, immunosuppression and activation due to volatile anesthetics can be either detrimental or beneficial. This review provides a balanced perspective on the anesthetic modulation of innate and adaptive immune responses as well as indirect effectors of immunity. Potential mechanisms of immunomodulation by volatile anesthetics are also discussed. A clearer understanding of these issues will pave the way for clinical guidelines that better account for the impact of volatile anesthetics on the immune system, with the ultimate goal of improving perioperative management.


Subject(s)
Anesthetics, Inhalation/pharmacology , Immunologic Factors/pharmacology , Adaptive Immunity/drug effects , Animals , Humans , Immunity, Innate/drug effects
2.
Nanomedicine ; 9(2): 222-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23123732

ABSTRACT

Macrophage carriage, release, and antitumor activities of polymeric nanoformulated paclitaxel (PTX) were developed as a novel delivery system for malignant glioma. To achieve this goal, the authors synthesized PTX-loaded nanoformulations (nano-PTX), then investigated their uptake, release, and toxicological properties. Chemosensitivity was significant in U87 cells (P < 0.05) at concentrations from 10(-4) to 10(-8) M following 72 hours' exposure to bone-marrow-derived macrophages (BMM)-nano-PTX in comparison with treatment with nano-PTX alone. The most significant reductions in U87 cell viability (P < 0.05) were observed in the transwell cocultures containing BMM-nano-PTX. Limited toxicity to BMM was observed at the same concentrations. BMM functions were tested by analysis of microtubules and actin filaments, as the cytoarchitecture, demonstrating a similar cytoskeleton pattern before and after nano-PTX was loaded into cells. This data indicate that nanoformulations of PTX facilitate cell uptake, delay toxicity, and show improved therapeutic efficacy by BMM-nano-PTX delivery. FROM THE CLINICAL EDITOR: In this study the delivery, release, and antitumor activity of polymeric nanoformulated paclitaxel carried by macrophages are described as a novel and efficient system for treatment of resistant malignant glioma.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems/methods , Glioma/drug therapy , Macrophages/cytology , Paclitaxel/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Nanostructures/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology
3.
ACS Chem Neurosci ; 14(23): 4115-4127, 2023 12 06.
Article in English | MEDLINE | ID: mdl-37967214

ABSTRACT

Cardiac arrest is one of the most dangerous health problems in the world. Outcome prognosis is largely based on cerebral performance categories determined by neurological evaluations. Few systemic tests are currently available to predict survival to hospital discharge. Here, we present the results from the preclinical studies of cardiac arrest and resuscitation (CAR) in mice to identify signatures of circulating immune cells as blood-derived biomarkers to predict outcomes after CAR. Two flow cytometry panels for circulating blood lymphocytes and myeloid-derived cells, respectively, were designed to correlate with neuroinflammation and neuronal and dendritic losses in the selectively vulnerable regions of bilateral hippocampi. We found that CD4+CD25+ regulatory T cells, CD11b+CD11c- and CD11b+Ly6C+Ly6G+ myeloid-derived cells, and cells positive for the costimulatory molecules CD80 and CD86 in the blood were correlated with activation of microglia and astrocytosis, and CD4+CD25+ T cells are additionally correlated with neuronal and dendritic losses. A fingerprint pattern of blood T cells and monocytes is devised as a diagnostic tool to predict CAR outcomes. Blood tests aimed at identifying these immunocyte patterns in cardiac arrest patients will guide future clinical trials to establish better prognostication tools to avoid unnecessary early withdrawal from life-sustaining treatment.


Subject(s)
Encephalitis , Heart Arrest , Humans , Mice , Animals , Myeloid Cells , Biomarkers , Prognosis
4.
Stroke ; 43(3): 884-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22156696

ABSTRACT

BACKGROUND AND PURPOSE: Erythropoietin (EPO) has been demonstrated to possess significant neuroprotective effects in stroke. We determined if the nano-drug form of human recombinant EPO (PLGA-EPO nanoparticles [PLGA-EPO-NP]) can enhance neuroprotection at lower dosages versus human recombinant EPO (r-EPO). METHODS: Established neonatal rat model of unilateral ischemic stroke was used to compare r-EPO, PLGA-EPO-NP and phosphate-buffered saline, given by daily intraperitoneal injections, followed by infarction volume and Rotarod Performance Test assessment. RESULTS: PLGA-EPO-NP significantly reduced infarction volumes 72 hours after injury compared with the same concentrations of r-EPO. Functional deficits were significantly reduced by 300 U/kg PLGA-EPO-NP versus controls, with deficit attenuation apparent at significantly lower dosages of PLGA-EPO-NP versus r-EPO. CONCLUSIONS: PLGA-EPO-NP is neuroprotective and beneficial against deficits after brain ischemia, at significantly reduced dosages versus r-EPO.


Subject(s)
Brain Ischemia/drug therapy , Erythropoietin/pharmacology , Hypoxia, Brain/drug therapy , Neuroprotective Agents , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain Ischemia/pathology , Brain Ischemia/psychology , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Cerebral Infarction/psychology , Erythropoietin/administration & dosage , Excipients , Female , Hypoxia, Brain/pathology , Hypoxia, Brain/psychology , Injections, Intraperitoneal , Lactic Acid , Male , Nanoparticles , Nervous System Diseases/prevention & control , Nervous System Diseases/psychology , Organ Size/physiology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Postural Balance/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Rotarod Performance Test
5.
J Immunol ; 184(2): 746-56, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19966207

ABSTRACT

When the nervous system is infected with HIV-1, it commonly results in neuroinflammation leading to overt neuronal dysfunction and subsequent cognitive and behavioral impairments. The multifaceted disease process, now referred to as HIV-1-associated neurocognitive disorders (HAND), provides a range of molecular targets for adjunctive therapies. One is CEP-1347, an inhibitor of mixed lineage kinases that elicits neuroprotective and anti-inflammatory responses in models of neurodegenerative diseases. Since HAND is associated with inflammatory encephalopathy induced by virus infection and mononuclear phagocytes (perivascular macrophages and microglia) immune activation, we investigated whether CEP-1347 could ameliorate disease in laboratory models of HAND. We now demonstrate that CEP-1347 reduces the levels of secreted proinflammatory cytokines and chemokines in HIV-1-infected human macrophages and attenuates dose-dependent neurotoxicity in rodent cortical neurons. CEP-1347-treated mice readily achieve therapeutic drug levels in peripheral blood. HIV-1 encephalitis (HIVE) mice, where human virus-infected monocyte-derived macrophages are stereotactically injected into the basal ganglia of CB17 severe combined immunodeficient mice, received daily intraperitoneal injections of CEP-1347. Here, CEP-1347 treatment of HIVE mice showed a dose-dependent reduction in microgliosis. Dendritic integrity and neuronal loss were sustained and prevented, respectively. These results demonstrate that CEP-1347 elicits anti-inflammatory and neuroprotective responses in an HIVE model of human disease and as such warrants further study as an adjunctive therapy for human disease.


Subject(s)
AIDS Dementia Complex/drug therapy , Carbazoles/administration & dosage , Carbazoles/therapeutic use , AIDS Dementia Complex/prevention & control , Animals , Basal Ganglia/virology , Cerebral Cortex/pathology , Chemotherapy, Adjuvant , Cytokines/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gliosis/prevention & control , Humans , Inflammation/prevention & control , Macrophages/metabolism , Macrophages/virology , Mice , Neurons/virology , Neuroprotective Agents/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Treatment Outcome
6.
Nanomedicine ; 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22306157

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

7.
Front Immunol ; 13: 754557, 2022.
Article in English | MEDLINE | ID: mdl-35663976

ABSTRACT

Delivery of plasmid DNA to transfect human primary macrophages is extremely difficult, especially for genetic engineering. Engineering macrophages is imperative for the treatment of many diseases including infectious diseases, cancer, neurological diseases, and aging. Unfortunately, plasmid does not cross the nuclear membranes of terminally differentiated macrophages to integrate the plasmid DNA (pDNA) into their genome. To address this issue, we have developed a core-shell nanoparticle (NP) using our newly created cationic lipid to deliver the anti-inflammatory cytokine IL-4 pDNA (IL-4pDNA-NPs). Human blood monocyte-derived macrophages (MDM) were effectively transfected with IL-4pDNA-NPs. IL-4pDNA-NPs were internalized in MDM within 30 minutes and delivered into the nucleus within 2 hours. Exogenous IL-4 expression was detected within 1 - 2 days and continued up to 30 days. Functional IL-4 expression led to M2 macrophage polarization in vitro and in an in vivo mouse model of inflammation. These data suggest that these NPs can protect pDNA from degradation by nucleases once inside the cell, and can transport pDNA into the nucleus to enhance gene delivery in macrophages in vitro and in vivo. In this research, we developed a new method to deliver plasmids into the nucleus of monocytes and macrophages for gene-editing. Introducing IL-4 pDNA into macrophages provides a new gene therapy solution for the treatment of various diseases.


Subject(s)
Gene Editing , Monocytes , Animals , DNA/metabolism , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , Macrophages/metabolism , Mice , Monocytes/metabolism
8.
Sci Rep ; 12(1): 2417, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35165339

ABSTRACT

Programmed death ligand 1 (PD-L1) plays a key role in glioblastoma multiforme (GBM) immunosuppression, vitality, proliferation, and migration, and is therefore a promising target for treating GBM. CRISPR/Cas9-mediated genomic editing can delete both cell surface and intracellular PD-L1. This systemic deliverable genomic PD-L1 deletion system can be used as an effective anti-GBM therapy by inhibiting tumor growth and migration, and overcoming immunosuppression. To target PD-L1 for CRISPR/Cas9 gene editing, we first identified two single guide RNA (sgRNA) sequences located on PD-L1 exon 3. The first sgRNA recognizes the forward strand of human PD-L1 near the beginning of exon 3 that allows editing by Cas9 at approximately base pair 82 (g82). The second sgRNA recognizes the forward strand of exon 3 that directs cutting at base pair 165 (g165). A homology-directed repair template (HDR) combined with the dual-sgRNAs was used to improve PD-L1 knockout specificity and efficiency. sgRNAs g82 and g165 were cloned into the multiplex CRISPR/Cas9 assembly system and co-transfected with the HDR template in human U87 GBM cells (g82/165 + HDR). T7E1 analysis suggests that the dual-sgRNA CRISPR/Cas9 strategy with a repair template was capable of editing the genomic level of PD-L1. This was further confirmed by examining PD-L1 protein levels by western blot and immunofluorescence assays. Western blot analysis showed that the dual-sgRNAs with the repair template caused a 64% reduction of PD-L1 protein levels in U87 cells, while immunostaining showed a significant reduction of intracellular PD-L1. PD-L1 deletion inhibited proliferation, growth, invasion and migration of U87 cells, indicating intracellular PD-L1 is necessary for tumor progression. Importantly, U87 cells treated with g82/165 + HDR polarized tumor-associated macrophages (TAM) toward an M1 phenotype, as indicated by an increase in TNF-α and a decrease in IL-4 secretions. This was further confirmed with flow cytometry that showed an increase in the M1 markers Ly6C + and CD80 +, and a decrease in the M2 marker CD206 + both in vitro and in vivo. Utilizing dual-sgRNAs and an HDR template with the CRISPR/Cas9 gene-editing system is a promising avenue for the treatment of GBM.


Subject(s)
B7-H1 Antigen/genetics , Cell Polarity , Glioblastoma/genetics , Glioblastoma/physiopathology , Tumor-Associated Macrophages/cytology , B7-H1 Antigen/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Exons , Gene Editing , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , Neoplasm Invasiveness , RNA, Guide, Kinetoplastida , Tumor-Associated Macrophages/metabolism
9.
J Immunol ; 183(1): 661-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19535632

ABSTRACT

Antiretroviral therapy (ART) shows variable blood-brain barrier penetration. This may affect the development of neurological complications of HIV infection. In attempts to attenuate viral growth for the nervous system, cell-based nanoformulations were developed with the focus on improving drug pharmacokinetics. We reasoned that ART carriage could be facilitated within blood-borne macrophages traveling across the blood-brain barrier. To test this idea, an HIV-1 encephalitis (HIVE) rodent model was used where HIV-1-infected human monocyte-derived macrophages were stereotactically injected into the subcortex of severe combined immunodeficient mice. ART was prepared using indinavir (IDV) nanoparticles (NP, nanoART) loaded into murine bone marrow macrophages (BMM, IDV-NP-BMM) after ex vivo cultivation. IDV-NP-BMM was administered i.v. to mice resulting in continuous IDV release for 14 days. Rhodamine-labeled IDV-NP was readily observed in areas of HIVE and specifically in brain subregions with active astrogliosis, microgliosis, and neuronal loss. IDV-NP-BMM treatment led to robust IDV levels and reduced HIV-1 replication in HIVE brain regions. We conclude that nanoART targeting to diseased brain through macrophage carriage is possible and can be considered in developmental therapeutics for HIV-associated neurological disease.


Subject(s)
Brain/virology , Encephalitis, Viral/drug therapy , HIV-1/drug effects , Indinavir/administration & dosage , Macrophages/transplantation , Macrophages/virology , Nanocapsules/administration & dosage , Severe Combined Immunodeficiency/drug therapy , Animals , Biological Availability , Bone Marrow Cells/pathology , Bone Marrow Cells/virology , Brain/pathology , Cell Movement , Cells, Cultured , Disease Models, Animal , Drug Administration Schedule , Encephalitis, Viral/metabolism , HIV-1/growth & development , Humans , Indinavir/pharmacokinetics , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Severe Combined Immunodeficiency/virology , Virus Replication/drug effects
10.
Curr Gene Ther ; 21(4): 349-360, 2021.
Article in English | MEDLINE | ID: mdl-33573550

ABSTRACT

BACKGROUND: Successful delivery of gene-editing tools using nano-carriers is dependent on the ability of nanoparticles to pass through the cellular membrane, move through the cytoplasm, and cross the nuclear envelope to enter the nucleus. It is critical that intracellular nanoparticles interact with the cytoskeletal network to move toward the nucleus, and must escape degradation pathways including lysosomal digestion. Without efficient intracellular transportation and nuclear entry, nanoparticles-based gene-editing cannot be effectively used for targeted genomic modification. OBJECTIVE: We have developed nanoparticles with a low molecular weight branched polyethylenimine lipid shell and a PLGA core that can effectively deliver plasmid DNA to macrophages for gene editing while limiting toxicity. METHODS: Core-shell nanoparticles were synthesized by a modified solvent evaporation method and were loaded with plasmid DNA. Confocal microscopy was used to visualize the internalization, intracellular distribution and cytoplasmic transportation of plasmid DNA loaded nanoparticles (pDNA-NPs) in bone marrow-derived macrophages. RESULTS: Core-shell nanoparticles had a high surface charge of +56 mV and narrow size distribution. When loaded with plasmid DNA for transfection, the nanoparticles increased in size from 150 nm to 200 nm, and the zeta potential decreased to +36 mV, indicating successful encapsulation. Further, fluorescence microscopy revealed that pDNA-NPs crossed the cell membrane and interacted with actin filaments. Intracellular tracking of pDNA-NPs showed successful separation of pDNA- NPs from lysosomes, allowing entry into the nucleus at 2 hours, with further nuclear ingress up to 5 hours. Bone marrow-derived macrophages treated with pDNA/GFP-NPs exhibited high GFP expression with low cytotoxicity. CONCLUSION: Together, this data suggests pDNA-NPs are an effective delivery system for macrophage gene-editing.


Subject(s)
Gene Editing , Nanoparticles , Cytoplasm/genetics , DNA/genetics , Macrophages , Plasmids/genetics , Polyethyleneimine , Transfection
11.
J Neuroimmunol ; 200(1-2): 41-52, 2008 Aug 30.
Article in English | MEDLINE | ID: mdl-18653244

ABSTRACT

Blood-borne macrophage ingress into brain in HIV-1 associated neurocognitive disorders governs the tempo of disease. We used superparamagnetic iron-oxide particles loaded into murine bone marrow-derived macrophages (BMM) injected intravenously into HIV-1 encephalitis mice to quantitatively assess BMM entry into diseased brain regions. Magnetic resonance imaging tests were validated by histological coregistration and enhanced image processing. The demonstration of robust BMM migration into areas of focal encephalitis provide 'proof of concept' for the use of MRI to monitor macrophage ingress into brain.


Subject(s)
Blood-Brain Barrier/physiopathology , Encephalitis/etiology , Encephalitis/pathology , HIV Infections/complications , Macrophages/physiology , Animals , Blood-Brain Barrier/pathology , Cell Movement/physiology , Disease Models, Animal , Encephalitis/virology , Glial Fibrillary Acidic Protein/metabolism , Imaging, Three-Dimensional , Macrophages/pathology , Magnetic Resonance Imaging , Male , Mice , Mice, SCID , Vimentin/metabolism
12.
Aging Dis ; 9(3): 412-425, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29896429

ABSTRACT

Although a direct link has long been suspected between systemic immune responses and neuronal injuries after stroke, it is unclear which immune cells play an important role. A question remains as to whether the blood brain barrier (BBB) is transiently disrupted after circulatory arrest to allow peripheral immune cells to enter brain parenchyma. Here, we developed a clinically relevant cardiac arrest and resuscitation model in mice to investigate the BBB integrity using noninvasive magnetic resonance imaging. Changes in immune signals in the brain and periphery were assayed by immunohistochemistry and flow cytometry. Quantitative variance maps from T1-weighted difference images before and after blood-pool contrast clearance revealed BBB disruptions immediately after resuscitation and one day after reperfusion. Time profiles of hippocampal CA1 neuronal injuries correlated with the morphological changes of microglia activation. Cytotoxic T cells, CD11b+CD11c+ dendritic cells, and CD11b+CD45+hi monocytes and macrophages were significantly increased in the brain three days after cardiac arrest and resuscitation, suggesting direct infiltration of these cells following the BBB disruption. Importantly, these immune cell changes were coupled with a parallel increase in the same subset of immune cell populations in the bone marrow and blood. We conclude that neurovascular breakdown during the initial reperfusion phase contributes to the systemic immune cell invasion and subsequent neuropathogenesis affecting the long-term outcome after cardiac arrest and resuscitation.

13.
J Biomed Nanotechnol ; 14(10): 1785-1795, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30041724

ABSTRACT

Convenient methods for the preparation of gene delivery platforms based on branched low molecular weight polyethylenimine (PEI) were described. Firstly, PEI lipids, with a low molecular weight PEI headgroup and hexadecyl chain tail group, were prepared through a highly efficient ring-opening reaction of glycidyl hexadecyl ether (EpoxyC16) by amine from PEI. Then, the PEI lipids were used as a component of cationic liposomes and as a surfactant for the preparation of poly(D,L-lactide-co-glycolide) (PLGA) nanoparticle (NP) via solvent extraction/evaporation method. As potential effective gene delivery platforms, their preparation, size, size distribution, toxicities, plasmid DNA loading, in vitro transfection and intracellular trafficking were studied. Both facile platforms showed less toxicity and higher transfection efficacy when compared to high molecular weight PEI in vitro, and may have further versatile applications in the gene delivery field.


Subject(s)
Polyethyleneimine/chemistry , Cell Survival , DNA , Drug Carriers , Molecular Weight , Particle Size , Plasmids , Transfection
14.
J Leukoc Biol ; 80(5): 1165-74, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16908517

ABSTRACT

We posit that the same mononuclear phagocytes (MP) [bone marrow (BM) and blood monocytes, tissue macrophages, microglia, and dendritic cells] which serve as targets, reservoirs, and vehicles for HIV dissemination, can be used as vehicles for antiretroviral therapy (ART). Toward this end, BM macrophages (BMM) were used as carriers for nanoparticle-formulated indinavir (NP-IDV), and the cell distribution was monitored by single photon emission computed tomography (SPECT), transverse relation time (T2)* weighted magnetic resonance imaging (MRI), histology, and gamma-scintillation spectrometry. BMM labeled with super paramagnetic iron oxide and/or 111indium oxine were infused i.v. into naïve mice. During the first 7 h, greater than 86% of cell label was recorded within the lungs. On Days 1, 3, 5, and 7, less than 10% of BMM were in lungs, and 74-81% and 13-18% were in liver and spleen, respectively. On a tissue volume basis, as determined by SPECT and MRI, BMM densities in spleen and liver were significantly greater than other tissues. Migration into the lymph nodes on Days 1 and 7 accounted for 1.5-2% of the total BMM. Adoptive transfer of BMM loaded with NP-IDV produced drug levels in lymphoid and nonlymphoid tissues that exceeded reported therapeutic concentrations by 200- to 350-fold on Day 1 and remained in excess of 100- to 300-fold on Day 14. These data show real-time kinetics and destinations of macrophage trafficking and demonstrate the feasibility of monitoring macrophage-based, nanoformulated ART.


Subject(s)
Drug Carriers/chemistry , Indinavir/chemistry , Macrophages/physiology , Magnetic Resonance Imaging/methods , Nanoparticles/chemistry , Tomography, Emission-Computed, Single-Photon/methods , Adoptive Transfer , Animals , Bone Marrow/chemistry , Cell Movement/physiology , Dextrans , Drug Carriers/pharmacokinetics , Drug Delivery Systems , Feasibility Studies , Ferrosoferric Oxide , Indinavir/pharmacokinetics , Indium Radioisotopes , Iron/pharmacokinetics , Macrophages/drug effects , Macrophages/transplantation , Magnetite Nanoparticles , Male , Mice , Mice, Inbred BALB C , Organometallic Compounds/pharmacokinetics , Oxides/pharmacokinetics , Oxyquinoline/analogs & derivatives , Oxyquinoline/pharmacokinetics , Tissue Distribution
15.
Oncotarget ; 8(4): 6564-6578, 2017 Jan 24.
Article in English | MEDLINE | ID: mdl-28036254

ABSTRACT

Nanoparticles containing mixed lipid monolayer shell, biodegradable polymer core and rabies virus glycoprotein (RVG) peptide as brain targeting ligand, were developed for brain targeted delivery of paclitaxel (PTX) to treat malignant glioma. RVG conjugated PTX loaded NPs (RVG-PTX-NPs) had the desirable size (~140 nm), narrow size distribution and spherical shape. RVG-PTX-NPs showed poor uptake by neurons and selective targeting to the brain tumor associated macrophages (TAMs) with controlled release and tumor specific toxicity. In vivo studies revealed that RVG-PTX-NPs were significant to cross the blood-brain barrier (BBB) and had specific targeting to the brain. Most importantly, RVG-PTX-NPs showed effectiveness for anti-glioma therapy on human glioma of mice model. We concluded that RVG-PTX-NPs provided an effective approach for brain-TAMs targeted delivery for the treatment of glioma.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Carriers , Glioma/drug therapy , Macrophages/drug effects , Nanoparticles , Paclitaxel/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Capillary Permeability , Cell Line, Tumor , Coculture Techniques , Delayed-Action Preparations , Dose-Response Relationship, Drug , Drug Compounding , Drug Design , Glioma/metabolism , Glioma/pathology , Glycoproteins/metabolism , Humans , Lactic Acid/chemistry , Ligands , Lipids/chemistry , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred BALB C , Mice, SCID , Neurons/metabolism , Paclitaxel/chemistry , Paclitaxel/metabolism , Paclitaxel/toxicity , Peptide Fragments/metabolism , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Time Factors , Tissue Distribution , Viral Proteins/metabolism , Xenograft Model Antitumor Assays
16.
J Neurosci ; 25(37): 8375-85, 2005 Sep 14.
Article in English | MEDLINE | ID: mdl-16162919

ABSTRACT

Lithium (Li) has garnered considerable interest as a neuroprotective drug for a broad range of nervous system disorders. Its neuroprotective activities occur as a consequence of glycogen synthase kinase-3beta (GSK-3beta) inhibition leading to downstream blockade of beta-catenin and Tau phosphorylation. In the present study, we investigated Li-mediated neuroprotective mechanisms in laboratory and murine human immunodeficiency virus-1 (HIV-1) encephalitis (HIVE) models. In laboratory tests, Li protected neurons from neurotoxic secretions of HIV-1-infected monocyte-derived macrophages (MDMs). This neuroprotection was mediated, in part, through the phosphatidyl inositol 3-kinase/Akt and GSK-3beta pathways. To examine the effects of Li treatment in vivo, MDMs were injected into the basal ganglia of severe combined immunodeficient mice and then Li was administered (60 mg/kg/d). Seven days after MDM injection, mice were killed and CNS tissue was collected and subjected to immunocytochemical and Western blot assays for leukocyte and neural antigens, GSK-3beta, and key kinase substrates such as beta-catenin and Tau. Numbers of HIV-1 p24 antigen-positive MDMs were unaltered by Li treatment of HIVE mice. Similarly, the greatly increased extent of astrocyte and microglia activation in HIVE mice (10-fold and 16-fold, respectively, compared with unmanipulated controls) was also unaltered by Li. In contrast, Li restored HIVE-associated loss of microtubule-associated protein-2-positive neurites and synaptic density while reducing levels or activity of phospho-Tau Ser202, phospho-beta-catenin, and GSK-3beta. Electrophysiological recordings showed diminished long-term potentiation in hippocampal slices of HIVE mice that were restored by Li. Based on these data, the use of Li as an adjuvant for HIV-1-associated dementia is now being pursued.


Subject(s)
Encephalitis, Viral/prevention & control , HIV Infections/complications , HIV-1/drug effects , HIV-2/drug effects , Lithium/pharmacology , Abortion, Induced , Animals , Brain/embryology , Brain/pathology , Cells, Cultured , Disease Models, Animal , Fetus , Humans , Macrophages/virology , Male , Mice , Mice, SCID , Monocytes/virology , Neurons/cytology , Neurons/drug effects , Neurons/physiology
17.
Oncogene ; 22(34): 5221-8, 2003 Aug 14.
Article in English | MEDLINE | ID: mdl-12917623

ABSTRACT

Porphobilinogen deaminase (PBGD) is a rate-limiting enzyme of the heme biosynthesis pathway, whose level is elevated in various human tumors. PBGD was observed in both nuclear and cytoplasmic fractions of C6 glioma cells by immunostaining. During mitosis, chromatids were intensely stained for PBGD in comparison to the interphase chromatin. Using the yeast two-hybrid system, we identified RanBPM, the nuclear Ran-binding protein, as an interacting partner of PBGD. During butyrate-induced differentiation of C6, both nuclear and cytoplasmic PBGD levels declined as did Ran protein and its nucleotide exchange factor RCC1. N,N'-hexamethylene bis-acetamide-dependent differentiation resulted in an increase of the cytoplasmic PBGD, whereas nuclear PBGD, Ran protein and RCC1 remained unchanged. mRNA levels of PBGD remained unchanged during stimulation with both butyrate and N,N'-hexamethylene bis-acetamide. The enzymatic activity of PBGD and protoporphyrin IX synthesis in C6 cells were dependent on the differentiation induction agent. We conclude that PBGD possibly has a nuclear role in addition to its cytosolic enzymatic activity required for heme synthesis, which is related to cell transformation and differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Nucleus/metabolism , Glioma/metabolism , Hydroxymethylbilane Synthase/metabolism , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Animals , HeLa Cells , Humans , Molecular Sequence Data , Porphyrins/metabolism , Rats
18.
J Neurosci ; 23(27): 9162-70, 2003 Oct 08.
Article in English | MEDLINE | ID: mdl-14534250

ABSTRACT

Human immunodeficiency virus-1 (HIV-1) infection of the nervous system can result in neuroinflammatory events leading first to neuronal dysfunction then to cognitive and behavioral impairments in infected people. The multifaceted nature of the disease process, commonly called HIV-1-associated dementia (HAD), provides a number of adjunctive therapeutic opportunities. One proposed adjunctive therapy is sodium valproate (VPA), an anticonvulsant known to promote neurite outgrowth and increase beta-catenin through inhibiting glycogen synthase kinase 3beta activity and tau phosphorylation. We now show that VPA treatment of rat cortical neurons exposed to HIV-1 gp120 prevents resultant neurotoxic activities. This includes the induction of significant neurite outgrowth and microtubule-associated protein 2 (MAP-2) and neuron-specific nuclear protein (NeuN) antigens in affected neuronal cell bodies and processes. Similarly, VPA protects severe combined immunodeficient (SCID) mice against the neurodegeneration of HIV-1ADA infected monocyte-derived macrophages (MDMs). In SCID mice with HIV-1 MDM-induced encephalitis, VPA treatment significantly reduced neuronal phosphorylatedbeta-catenin and tau without affecting HIV-1 replication or glial activation. We conclude that VPA protects neurons against HIV-1 infected MDM neurotoxicity, possibly through its effects on the phosphorylation of tau and beta-catenin. The use of VPA as an adjuvant in treatment of human HAD is being pursued.


Subject(s)
AIDS Dementia Complex/drug therapy , HIV-1 , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Valproic Acid/therapeutic use , AIDS Dementia Complex/pathology , AIDS Dementia Complex/physiopathology , Animals , Basal Ganglia/pathology , Basal Ganglia/virology , Biomarkers/analysis , Cell Differentiation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , HIV Core Protein p24/analysis , HIV Core Protein p24/biosynthesis , HIV Envelope Protein gp120/toxicity , HIV-1/genetics , HIV-1/growth & development , HIV-1/isolation & purification , Humans , Macrophages/pathology , Macrophages/virology , Male , Mice , Mice, SCID , Monocytes/cytology , Monocytes/drug effects , Neuroglia/drug effects , Neuroglia/pathology , Neurons/pathology , Phosphorylation/drug effects , RNA, Viral/analysis , RNA, Viral/biosynthesis , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
19.
Curr HIV Res ; 2(1): 61-78, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15053341

ABSTRACT

Persons with advanced human immunodeficiency virus type one (HIV-1) infection seek medical advice for a wide range of neurological disorders including, but not limited to, peripheral neuropathy, toxoplasmosis, cryptococcal meningitis, cytomegalovirus retinitis progressive multifocal leukoencephalopathy, lymphoma and dementia. The diagnosis of HIV-1-associated dementia (HAD) induced as a direct consequence of HIV infection of the brain comes commonly by exclusion. Diagnostic decisions can often be clouded by concomitant depression, motor impairments, and lethargy that follow debilitating immune suppression and weight loss. Indeed, cognitive, motor and behavior abnormalities underlie a variety of neurological dysfunctions associated with advanced HIV-1 infection. Thus, even combinations of clinical, laboratory and neuroimaging tests [for example, magnetic resonance imaging (MRI), computed tomography (CT), single photon emission computed tomography (SPECT) and positron emission tomography (PET)] often fail to provide conclusive diagnostic information. Nonetheless, the recent development of quantitative MR spectroscopic imaging has improved diagnostic possibilities for HAD. We are pleased to discuss these developments as well as taking a forward look into what will soon be made available to improve neuroimaging diagnostic precision. New MR and SPECT testing are being developed in our laboratories and elsewhere both for animal model systems and in humans with HIV-1 disease. Such tests can facilitate dynamic measures of HIV-1 neuropathogenesis providing information for disease events that even 2 years ago were unattainable.


Subject(s)
AIDS Dementia Complex/pathology , Brain/pathology , Diagnostic Imaging/methods , HIV-1 , AIDS Dementia Complex/diagnosis , AIDS Dementia Complex/drug therapy , Animals , Brain/metabolism , Disease Models, Animal , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Mice , Tomography, Emission-Computed , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed
20.
Neurotox Res ; 6(7-8): 503-21, 2004.
Article in English | MEDLINE | ID: mdl-15639783

ABSTRACT

The human immunodeficiency virus-1 (HIV-1) commonly affects cognitive, behavioral and motor functions during the disease course. The neuropathogenesis of viral infection revolves around neurotoxins produced from infected and immune-activated mononuclear phagocytes (MP; perivascular macrophages and microglia). Direct infection of neurons occurs rarely, if at all. Neurologic disease arises in part as a consequence of MP metabolic dysfunction. Although the advent of highly active antiretroviral therapy (HAART) has attenuated the incidence and severity of neurologic disease, it, nonetheless, remains a common and disabling problem for those living with HIV-1 infection. Adjunctive therapies are currently designed to ameliorate clinical outcomes and are included in the therapeutic armamentarium. Anti-inflammatory drugs that inhibit cytokines, chemokines and interferons linked to neurodegenerative processes can significantly ameliorate neuronal function. HIV-1 neurotoxins have the unique ability to up-regulate glycogen synthase kinase-3beta (GSK-3beta) activity that in turn elicits neuronal apoptosis. GSK-3beta inhibitors are neuroprotective in animal models of Neuro AIDS. They are also currently in Phase 1 clinical trials designed for safety and tolerability in patients with HIV-1 infection. Neurotrophins are only beginning to be realized for their therapeutic potential in HIV-1 associated neurologic disease. This review article provides a broad overview of neuroprotective strategies for HIV-1 infection and details how such strategies act and may be implemented for treatment of human disease.


Subject(s)
AIDS Dementia Complex/drug therapy , HIV-1/pathogenicity , Neuroprotective Agents/therapeutic use , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL