Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Chem Neurosci ; 14(23): 4115-4127, 2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-37967214

RESUMO

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.


Assuntos
Encefalite , Parada Cardíaca , Humanos , Camundongos , Animais , Células Mieloides , Biomarcadores , Prognóstico
2.
Front Immunol ; 13: 754557, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35663976

RESUMO

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.


Assuntos
Edição de Genes , Monócitos , Animais , DNA/metabolismo , Humanos , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/metabolismo , Camundongos , Monócitos/metabolismo
3.
Sci Rep ; 12(1): 2417, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35165339

RESUMO

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.


Assuntos
Antígeno B7-H1/genética , Polaridade Celular , Glioblastoma/genética , Glioblastoma/fisiopatologia , Macrófagos Associados a Tumor/citologia , Antígeno B7-H1/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células , Éxons , Edição de Genes , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Interleucina-4/genética , Interleucina-4/metabolismo , Invasividade Neoplásica , RNA Guia de Cinetoplastídeos , Macrófagos Associados a Tumor/metabolismo
4.
Curr Gene Ther ; 21(4): 349-360, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33573550

RESUMO

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.


Assuntos
Edição de Genes , Nanopartículas , Citoplasma/genética , DNA/genética , Macrófagos , Plasmídeos/genética , Polietilenoimina , Transfecção
5.
J Biomed Nanotechnol ; 14(10): 1785-1795, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30041724

RESUMO

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.


Assuntos
Polietilenoimina/química , Sobrevivência Celular , DNA , Portadores de Fármacos , Peso Molecular , Tamanho da Partícula , Plasmídeos , Transfecção
6.
Aging Dis ; 9(3): 412-425, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29896429

RESUMO

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.

7.
Oncotarget ; 8(4): 6564-6578, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-28036254

RESUMO

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.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Portadores de Fármacos , Glioma/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Nanopartículas , Paclitaxel/administração & dosagem , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/toxicidade , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Permeabilidade Capilar , Linhagem Celular Tumoral , Técnicas de Cocultura , Preparações de Ação Retardada , Relação Dose-Resposta a Droga , Composição de Medicamentos , Desenho de Fármacos , Glioma/metabolismo , Glioma/patologia , Glicoproteínas/metabolismo , Humanos , Ácido Láctico/química , Ligantes , Lipídeos/química , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos SCID , Neurônios/metabolismo , Paclitaxel/química , Paclitaxel/metabolismo , Paclitaxel/toxicidade , Fragmentos de Peptídeos/metabolismo , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Fatores de Tempo , Distribuição Tecidual , Proteínas Virais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Anesthesiology ; 125(2): 399-411, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27286478

RESUMO

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.


Assuntos
Anestésicos Inalatórios/farmacologia , Fatores Imunológicos/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Humanos , Imunidade Inata/efeitos dos fármacos
9.
Nanomedicine ; 9(2): 222-32, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23123732

RESUMO

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.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Glioma/tratamento farmacológico , Macrófagos/citologia , Paclitaxel/administração & dosagem , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Nanoestruturas/química , Paclitaxel/farmacocinética , Paclitaxel/farmacologia
10.
Colloids Surf B Biointerfaces ; 102: 604-10, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23104031

RESUMO

Vitamin E succinate (TOS) modified pluronic micelles (PF-TOS micelles) were prepared to be used as a vehicle for paclitaxel (PTX). The average size of resultant PTX-loaded PF-TOS micelles (PF-TOS-PTX-micelles) was about 58 nm. PF-TOS-PTX-micelles showed enhanced encapsulation efficiency and better stability as compared to the non-modified controls. Fluorescence microscopy and high performance liquid chromatography studies showed that PTX-loaded PF-TOS micelles had excellent cellular uptake ability by human glioma U87 cells. Cytotoxicity assay revealed that PTX-loaded PF-TOS micelles demonstrated higher anti-cancer activity to U87 cells than that of the non-modified PF micelles. Pharmacokinetic studies indicated the longer systemic circulation time and slower plasma elimination rate in PTX-loaded PF-TOS micelles than that of the non-modified controls. In vivo tissue distribution studies showed that PTX-loaded PF-TOS micelles were preferably accumulated in spleen and liver. Taken together, the results evidently indicated that PF-TOS micelles improve the hydrophobic chemotherapeutic drugs delivery by means of efficient encapsulation and stabilization for anti-tumor therapy.


Assuntos
Portadores de Fármacos/química , Micelas , Paclitaxel/química , Ácido Succínico/química , Vitamina E/análogos & derivados , Vitamina E/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Portadores de Fármacos/efeitos adversos , Estabilidade de Medicamentos , Humanos , Camundongos
11.
Transl Neurodegener ; 1(1): 15, 2012 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23210607

RESUMO

BACKGROUND: Neurogenesis, including the proliferation, migration and differentiation of neural progenitor cells (NPCs), is impaired in HIV-1 associated dementia (HAD). We previously demonstrated HIV-1-infected macrophages (HIV-MDM) regulate stromal cell-derived factor 1 (SDF-1) production in astrocytes through Interleukin-1ß (IL-1ß). Chemokines are known to induce NPC migration; however, it remains unclear how chemokines produced in inflammation regulate NPC migration. METHODS: The secretion of SDF-1 and Monocyte chemotactic preotein-1 (MCP-1) in astrocytes upon IL-1ß stimulation was measured by ELISA assay. Human NPCs were injected parallel along with IL-1ß, SDF-1 or MCP-1 intracranially into basal ganglion 1 mm apart in SCID mice, and immunofluorescent staining was used to study the survival and migration of injected human NPCs. RESULTS: SDF-1 and MCP-1 are secreted by astrocytes upon IL-1ß stimulation in a time-dependent manner. Injected human NPCs survived in SCID mice and migrated towards sites of IL-1ß, SDF-1 and MCP-1 injection. CONCLUSIONS: In conclusion, chemokines SDF-1 or MCP-1 secreted by astrocytes in the presence of IL-1ß injection are attractive to NPCs injected into SCID mouse brains, suggesting that SDF-1 and MCP-1 play important roles in NPC migration during neuroinflammation.

12.
Nanomedicine ; 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22306157

RESUMO

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.

13.
Stroke ; 43(3): 884-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22156696

RESUMO

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.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Eritropoetina/farmacologia , Hipóxia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Isquemia Encefálica/patologia , Isquemia Encefálica/psicologia , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Infarto Cerebral/psicologia , Eritropoetina/administração & dosagem , Excipientes , Feminino , Hipóxia Encefálica/patologia , Hipóxia Encefálica/psicologia , Injeções Intraperitoneais , Ácido Láctico , Masculino , Nanopartículas , Doenças do Sistema Nervoso/prevenção & controle , Doenças do Sistema Nervoso/psicologia , Tamanho do Órgão/fisiologia , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Equilíbrio Postural/fisiologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Teste de Desempenho do Rota-Rod
14.
Arthritis Res Ther ; 12(5): R170, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20836843

RESUMO

INTRODUCTION: The purpose of the present manuscript is to test the hypothesis that arthrotropic localization and synovial cell internalization account for the unique capacity of N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer-dexamethasone conjugate (P-Dex, a macromolecular prodrug of dexamethasone) to induce sustained amelioration of joint inflammation and inhibition of tissue damage in an animal model of inflammatory arthritis. METHODS: Rats with adjuvant-induced arthritis (AA) were treated with P-Dex, free dexamethasone, saline or HPMA homopolymer. To define the biodistribution of P-Dex, conjugates with different imaging labels were given to AA rats and analyzed. Isolated joint tissues were evaluated by fluorescence-activated cell sorting (FACS) and immunohistochemical staining. Cellular uptake of P-Dex and its effects on apoptosis and production of proinflammatory cytokines were examined using human monocyte-macrophages and fibroblasts. RESULTS: A single systemic administration of P-Dex completely suppressed AA for >20 days. Magnetic resonance imaging demonstrated higher HPMA copolymer influx into the inflamed joints than the normal joints. Immunohistochemistry and FACS analyses of arthritic joints revealed extensive uptake of the polymer conjugate by synovial fibroblasts and myeloid lineage cells. The capacity of P-Dex to suppress inflammation was confirmed in monocyte-macrophage cultures in which P-Dex treatment resulted in suppression of lipopolysaccharide-induced IL-6 and TNFα release. Similarly, TNFα-induced expression of matrix metalloproteinases (MMP1 and MMP3) in synovial fibroblasts from a rheumatoid arthritis patient was suppressed by P-Dex. P-Dex showed no detectable effect on monocyte apoptosis. CONCLUSIONS: P-Dex provides superior and sustained amelioration of AA compared with an equivalent dose of free dexamethasone. The arthrotropism and local retention of P-Dex is attributed to the enhanced vascular permeability in arthritic joints and the internalization of P-Dex by synovial cells. The uptake and processing of P-Dex by macrophages and fibroblasts, and downregulation of proinflammatory mediators, provides an explanation for the sustained anti-inflammatory efficacy of P-Dex in this model of inflammatory arthritis.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Dexametasona/farmacologia , Metacrilatos/farmacologia , Pró-Fármacos/farmacologia , Adjuvantes Imunológicos/toxicidade , Animais , Antirreumáticos/metabolismo , Antirreumáticos/farmacocinética , Antirreumáticos/farmacologia , Artrite Experimental/induzido quimicamente , Artrite Experimental/imunologia , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/imunologia , Permeabilidade Capilar/efeitos dos fármacos , Separação Celular , Dexametasona/metabolismo , Dexametasona/farmacocinética , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Humanos , Imuno-Histoquímica , Metacrilatos/metabolismo , Metacrilatos/farmacocinética , Microscopia Confocal , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacocinética , Ratos , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Distribuição Tecidual
15.
J Immunol ; 184(2): 746-56, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19966207

RESUMO

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.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Carbazóis/administração & dosagem , Carbazóis/uso terapêutico , Complexo AIDS Demência/prevenção & controle , Animais , Gânglios da Base/virologia , Córtex Cerebral/patologia , Quimioterapia Adjuvante , Citocinas/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Gliose/prevenção & controle , Humanos , Inflamação/prevenção & controle , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Neurônios/virologia , Fármacos Neuroprotetores/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Resultado do Tratamento
16.
Nanomedicine (Lond) ; 4(8): 903-17, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19958227

RESUMO

BACKGROUND: Factors limiting the efficacy of conventional antiretroviral therapy for HIV-1 infection include treatment adherence, pharmacokinetics and penetration into viral sanctuaries. These affect the rate of viral mutation and drug resistance. In attempts to bypass such limitations, nanoparticles containing ritonavir, indinavir and efavirenz (described as nanoART) were manufactured to assess macrophage-based drug delivery. METHODS: NanoART were made by high-pressure homogenization of crystalline drug with various surfactants. Size, charge and shape of the nanoparticles were assessed. Monocyte-derived macrophage nanoART uptake, drug release, migration and cytotoxicity were determined. Drug levels were measured by reverse-phase high-performance liquid chromatography. RESULTS: Efficient monocyte-derived macrophage cytoplasmic vesicle uptake in less than 30 min based on size, charge and coating was observed. Antiretroviral drugs were released over 14 days and showed dose-dependent reduction in progeny virion production and HIV-1 p24 antigen. Cytotoxicities resulting from nanoART carriage were limited. CONCLUSION: These results support the continued development of macrophage-mediated nanoART carriage for HIV-1 disease.


Assuntos
Benzoxazinas/farmacocinética , Inibidores da Protease de HIV/farmacocinética , Indinavir/farmacocinética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Monócitos/citologia , Ritonavir/farmacocinética , Alcinos , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/uso terapêutico , Benzoxazinas/administração & dosagem , Benzoxazinas/síntese química , Benzoxazinas/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ciclopropanos , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/administração & dosagem , Inibidores da Protease de HIV/uso terapêutico , Humanos , Indinavir/administração & dosagem , Indinavir/uso terapêutico , Macrófagos/virologia , Microscopia de Força Atômica , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Nanopartículas/química , Nanopartículas/ultraestrutura , Ritonavir/administração & dosagem , Ritonavir/uso terapêutico
17.
J Neuroimmunol ; 213(1-2): 47-59, 2009 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-19541372

RESUMO

The neuroregulatory activities of PMS-601, a platelet activating factor antagonist, were investigated in laboratory and animal models of HIV-1 encephalitis (HIVE). For the former, PMS-601 reduced monocyte-derived macrophage pro-inflammatory secretions, multinucleated giant cell (MGC) formation, and neuronal loss independent of antiretroviral responses. PMS-601 treatment of HIVE severe combined immunodeficient mice showed reduced microgliosis, MGCs and neurodegeneration. These observations support the further development of PMS-601 as an adjunctive therapy for HIV-1 associated neurocognitive disorders.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Piperazinas/farmacologia , Fator de Ativação de Plaquetas/antagonistas & inibidores , Complexo AIDS Demência/imunologia , Complexo AIDS Demência/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Células Cultivadas , Transtornos Cognitivos/imunologia , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Quinase 2 de Adesão Focal/efeitos dos fármacos , Quinase 2 de Adesão Focal/metabolismo , Células Gigantes/efeitos dos fármacos , Células Gigantes/imunologia , Células Gigantes/metabolismo , Gliose/tratamento farmacológico , Gliose/imunologia , Gliose/metabolismo , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Mediadores da Inflamação/agonistas , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos SCID , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Piperazinas/uso terapêutico , Fator de Ativação de Plaquetas/metabolismo
18.
J Immunol ; 183(1): 661-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535632

RESUMO

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.


Assuntos
Encéfalo/virologia , Encefalite Viral/tratamento farmacológico , HIV-1/efeitos dos fármacos , Indinavir/administração & dosagem , Macrófagos/transplante , Macrófagos/virologia , Nanocápsulas/administração & dosagem , Imunodeficiência Combinada Severa/tratamento farmacológico , Animais , Disponibilidade Biológica , Células da Medula Óssea/patologia , Células da Medula Óssea/virologia , Encéfalo/patologia , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Esquema de Medicação , Encefalite Viral/metabolismo , HIV-1/crescimento & desenvolvimento , Humanos , Indinavir/farmacocinética , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Imunodeficiência Combinada Severa/virologia , Replicação Viral/efeitos dos fármacos
19.
J Neuroimmune Pharmacol ; 4(3): 317-27, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19462247

RESUMO

HIV-1-associated neurocognitive disorders (HAND) remains a significant source of morbidity in the era of wide spread use of highly active antiretroviral therapy. Disease is precipitated by low levels of viral growth and glial immune activation within the central nervous system. Blood borne macrophage and microglia affect a proinflammatory response and release viral proteins that affects neuronal viability and leads to death of nerve cells. Increasing evidence supports the notion that HAND is functional channelopathy, but proof of this concept remains incomplete. Based on their role in learning and memory processes, we now posit that voltage-gated potassium (K(v)) channels could be a functional substrate for disease. This was tested in the severe combined immunodeficient (SCID) mouse model of HIV-1 encephalitis (HIVE) by examining whether the K(v) channel blocker, 4-aminopyridine (4-AP), could affect behavioral, electrophysiological, and morphological measures of learning and memory. HIVE SCID mice showed impaired spatial memory in radial arm water maze tests. Electrophysiology studies revealed a reduction of long-term potentiation (LTP) in the CA1 region of the hippocampus. Importantly, systemic administration of 4-AP blocked HIV-1-associated reduction of LTP and improved animal performance in the radial arm water maze. These results support the importance of K(v) channel dysfunction in disease but, more importantly, provide a potential target for adjunctive therapies for HAND.


Assuntos
4-Aminopiridina/farmacologia , Complexo AIDS Demência/tratamento farmacológico , Complexo AIDS Demência/psicologia , HIV-1 , Memória/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Percepção Espacial/efeitos dos fármacos , Complexo AIDS Demência/patologia , Complexo AIDS Demência/virologia , Animais , Encéfalo/patologia , Separação Celular , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Aprendizagem/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos SCID , Microscopia Eletrônica , Monócitos/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Natação/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/fisiologia
20.
PLoS One ; 4(2): e4343, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19183814

RESUMO

BACKGROUND: We posit that the same mononuclear phagocytes (MP) that serve as target cells and vehicles for a host of microbial infections can be used to improve diagnostics and drug delivery. We also theorize that physical and biological processes such as particle shape, size, coating and opsonization that affect MP clearance of debris and microbes can be harnessed to facilitate uptake of nanoparticles (NP) and tissue delivery. METHODS: Monocytes and monocyte-derived macrophages (MDM) were used as vehicles of superparamagnetic iron oxide (SPIO) NP and immunoglobulin (IgG) or albumin coated SPIO for studies of uptake and distribution. IgG coated SPIO was synthesized by covalent linkage and uptake into monocytes and MDM investigated related to size, time, temperature, concentration, and coatings. SPIO and IgG SPIO were infused intravenously into naïve mice. T(2) measures using magnetic resonance imaging (MRI) were used to monitor tissue distribution in animals. RESULTS: Oxidation of dextran on the SPIO surface generated reactive aldehyde groups and permitted covalent linkage to amino groups of murine and human IgG and F(ab')(2) fragments and for Alexa Fluor(R) 488 hydroxylamine to form a Schiff base. This labile intermediate was immediately reduced with sodium cyanoborohydride in order to stabilize the NP conjugate. Optical density measurements of the oxidized IgG, F(ab')(2), and/or Alexa Fluor(R) 488 SPIO demonstrated approximately 50% coupling yield. IgG-SPIO was found stable at 4 degrees C for a period of 1 month during which size and polydispersity index varied little from 175 nm and 200 nm, respectively. In vitro, NP accumulated readily within monocyte and MDM cytoplasm after IgG-SPIO exposure; whereas, the uptake of native SPIO in monocytes and MDM was 10-fold less. No changes in cell viability were noted for the SPIO-containing monocytes and MDM. Cell morphology was not changed as observed by transmission electron microscopy. Compared to unconjugated SPIO, intravenous injection of IgG-SPIO afforded enhanced and sustained lymphoid tissue distribution over 24 hours as demonstrated by MRI. CONCLUSIONS: Facilitated uptake of coated SPIO in monocytes and MDM was achieved. Uptake was linked to particle size and was time and concentration dependent. The ability of SPIO to be rapidly taken up and distributed into lymphoid tissues also demonstrates feasibility of macrophage-targeted nanoformulations for diagnostic and drug therapy.


Assuntos
Compostos Férricos/farmacocinética , Macrófagos/metabolismo , Nanopartículas Metálicas , Monócitos/metabolismo , Animais , Portadores de Fármacos/farmacocinética , Humanos , Fígado/metabolismo , Magnetismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Baço/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...