Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Nitric Oxide ; 148: 13-22, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38642795

RESUMO

Endothelial dysfunction, underlying the vascular complications of diabetes and other cardiovascular disorders, may result from uncoupling of endothelial nitric oxide synthase (eNOS) activity due to decreased levels of tetrahydrobiopterin (BH4), a critical co-factor for eNOS. Some clinical trials attempting to deliver exogenous BH4 as a potential therapeutic strategy in vascular disease states have failed due to oxidation of BH4 in the circulation. We sought to develop a means of protecting BH4 from oxidation while delivering it to dysfunctional endothelial cells. Polymeric and solid lipid nanoparticles (NPs) loaded with BH4 were delivered by injection or oral gavage, respectively, to streptozotocin-induced diabetic rats. BH4 was measured in coronary endothelial cells and endothelium-dependent vascular reactivity was assessed in vascular rings. Lymphatic uptake of orally delivered lipid NPs was verified by sampling mesenteric lymph. BH4-loaded polymeric NPs maintained nitric oxide production by cultured endothelial cells under conditions of oxidative stress. BH4-loaded NPs, delivered via injection or ingestion, increased coronary endothelial BH4 concentration and improved endothelium-dependent vasorelaxation in diabetic rats. Pharmacodynamics assessment indicated peak concentration of solid lipid NPs in the systemic bloodstream 6 hours after ingestion, with disappearance noted by 48 hours. These studies support the feasibility of utilizing NPs to deliver BH4 to dysfunctional endothelial cells to increase nitric oxide bioavailability. BH4-loaded NPs could provide an innovative tool to restore redox balance in blood vessels and modulate eNOS-mediated vascular function to reverse or retard vascular disease in diabetes.


Assuntos
Biopterinas , Diabetes Mellitus Experimental , Endotélio Vascular , Nanopartículas , Animais , Biopterinas/análogos & derivados , Biopterinas/farmacologia , Biopterinas/administração & dosagem , Biopterinas/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Masculino , Nanopartículas/química , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Ratos , Ratos Sprague-Dawley , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo
2.
J Pharmacol Exp Ther ; 370(3): 902-910, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30940690

RESUMO

Poor cellular uptake, rapid degradation in the presence of serum, and inefficient transfection are some of the major barriers in achieving therapeutic efficacy of naked small interfering RNAs (siRNAs). We investigated the efficacy of the polyplex formulated using our synthesized polymer, polyethylene glycol (PEG)-modified l-arginine oligo(-alkylaminosiloxane) that is grafted with poly(ethyleneimine) (PEI) for siRNA delivery. We hypothesized that the polyplex formulated using the polymer with a balanced composition of PEI for siRNA condensation and its protection, PEG for polyplex stability and to minimize the PEI-associated toxicity, and with arginine facilitating cellular uptake would overcome the aforementioned issues with siRNA delivery. We tested our hypothesis using antiluciferase siRNA in luciferase-expressing metastatic breast cancer cells (MDA-MB-231-Luc-D3H2LN) and anti-ABCB1 siRNA against an efflux membrane protein, ABCB1, in doxorubicin (DOX)-resistant breast cancer cells (MCF-7/Adr). The results demonstrated that the polyplex at an optimal nucleotide/polymer ratio is stable in the presence of excess polyanions, has no cellular toxicity, and protects siRNA from RNase degradation. Transfection of MDA-MB-231-Luc-D3H2LN cells with antiluciferase siRNA polyplex showed almost complete knockdown of luciferase expression. In MCF-7/Adr cells, transfection with anti-ABCB1 siRNA effectively downregulated its target efflux protein, ABCB1; increased cellular uptake of DOX; and enhanced its cytotoxic effect. However, the cotreatment did not completely overcome drug resistance, suggesting that further optimization is needed and/or a mechanism(s) other than the efflux protein ABCB1 may be involved in drug resistance. In conclusion, our polyplex is effective for siRNA delivery and can be explored for different therapeutic applications.


Assuntos
Arginina/química , Neoplasias da Mama/terapia , Polietilenoimina/química , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Subfamília B de Transportador de Cassetes de Ligação de ATP/efeitos dos fármacos , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Composição de Medicamentos , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Estrutura Molecular , RNA Interferente Pequeno/farmacocinética
3.
Curr Opin Urol ; 29(4): 458-465, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30985344

RESUMO

PURPOSE OF REVIEW: The present review highlights regenerative electrical stimulation (RES) as potential future treatment options for patients with nerve injuries leading to urological dysfunction, such as urinary incontinence, voiding dysfunction or erectile dysfunction. Additionally, it will highlight the mechanism of nerve injury and regeneration as well as similarities and differences between RES and current electrical stimulation treatments in urology, functional electrical stimulation (FES) and neuromodulation. RECENT FINDINGS: It has been demonstrated that RES upregulates brain-derived neurotrophic factor (BDNF) and its receptor to facilitate neuroregeneration, facilitating accurate reinnervation of muscles by motoneurons. Further, RES upregulates growth factors in glial cells. Within the past 2 years, RES of the pudendal nerve upregulated BDNF in Onuf's nucleus, the cell bodies of motoneurons that course through the pudendal nerve and accelerated functional recovery in an animal model of stress urinary incontinence. Additionally, electrical stimulation of the vaginal tissue in an animal model of stress urinary incontinence accelerated functional recovery. SUMMARY: RES has great potential but future research is needed to expand the potential beneficial effects of RES in the field of urology.


Assuntos
Terapia por Estimulação Elétrica/métodos , Doenças Urogenitais Masculinas/terapia , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/terapia , Animais , Feminino , Humanos , Masculino , Doenças Urogenitais Masculinas/etiologia , Modelos Animais , Traumatismos dos Nervos Periféricos/complicações
4.
J Urol ; 194(4): 1155-61, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25796115

RESUMO

PURPOSE: Nanoparticles, which are submicroscopic particles typically ranging from 100 to 300 nm, are interesting as potential treatment of testicular disorders because they can be engineered to allow delivery to privileged tissues, such as across the blood-brain barrier or theoretically the blood-testis barrier. We compared the effects of anatomical and/or ligand targeting on testicular nanoparticle uptake in a rat model. MATERIALS AND METHODS: A total of 48 rats were divided into 6 groups, including a control group and groups that received intra-arterial injection of unconjugated nanoparticles with and without saline flush, intravenous injection of unconjugated nanoparticles, intra-arterial injection of follicle stimulating hormone conjugated nanoparticles, intravenous injection of follicle stimulating hormone conjugated nanoparticles and intra-arterial injection of transactivating transcriptor conjugated nanoparticles. A dose response curve was assessed for intra-arterially injected unconjugated nanoparticles. Using high performance liquid chromatography and histological analysis we determined nanoparticle uptake by the testicle at 4 hours. RESULTS: Intra-arterial injection resulted in a 5.8-fold increase in uptake compared to intravenous injection at 35 mg/kg of unconjugated nanoparticles (3.7 vs 0.6 µg nanoparticles per gm testicle, p = 0.04). Anatomical targeting failed to improve testicular uptake in FSH conjugated nanoparticles (intra-arterial vs intravenous injection 0.33 vs 0.38 µg FSH nanoparticles per gm testicular tissue, p = 0.73). On fluorescence microscopy nanoparticles were noted in the testicular interstitium and seminiferous tubules, and absent from the testicular vasculature. CONCLUSIONS: Arterial injection for anatomical targeting of nanoparticles to the testis is feasible, improves unconjugated nanoparticle delivery to testicular tissue and enables nanoparticles to cross the gonadal vascular endothelium and the blood-testis barrier.


Assuntos
Sistemas de Liberação de Medicamentos , Hormônio Foliculoestimulante/administração & dosagem , Nanopartículas/administração & dosagem , Testículo , Animais , Sistemas de Liberação de Medicamentos/métodos , Masculino , Ratos , Ratos Sprague-Dawley
5.
Mol Pharm ; 12(2): 621-9, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25591125

RESUMO

In this study, we formulated polyplexes with different compositions for codelivery of DNA and small-interfering RNA (siRNA). Since DNA and siRNA have distinctive and complementary morphological characteristics (DNA is long and winding and siRNA is short and rigid), we hypothesized that their codelivery using polyplex would enhance each other's transfection. To test this hypothesis, cationic polymer branched polyethylenimine (bPEI) as a standard transfecting agent and its derivative arginine-rich oligopeptide-grafted bPEI modified with polyethylene glycol (P(SiDAAr)5P3), synthesized in our laboratory, were used as carriers for transfection. Polyplexes at different nucleic acid to polymer weight ratios were characterized for transfection in breast cancer sensitive (MCF-7) and resistant (MCF-7/Adr) cell lines. Gene silencing effect of polyplexes was determined in MDA-MB-231-luc-D3H2LN cell line. The results demonstrated that the polyplexes formed with derivative P(SiDAAr)5P3 show significantly lower toxicity compared to polyplexes formed using bPEI. Further, codelivery resulted in 20-fold higher DNA transfection and 2-fold higher siRNA transfection as compared to the respective single nucleotide delivery. DNA transfection was ∼100-fold lower in resistant MCF-7/Adr cells than in sensitive MCF-7 cells. Confocal imaging and flow cytometry data demonstrated that enhanced transfection does not solely depend on DNA's cellular uptake, suggesting that other mechanisms contribute to increased transfection. DNA-co-siRNA delivery could be a promising therapeutic approach to achieve synergistic effects because it can simultaneously target and interfere with multiple regulatory levels in a cell to halt and reverse disease progression.


Assuntos
Arginina/química , DNA/administração & dosagem , DNA/química , Polietilenoimina/química , Polímeros/química , RNA Interferente Pequeno/análise , RNA Interferente Pequeno/química , Linhagem Celular , Citometria de Fluxo , Humanos , Microscopia Eletrônica de Transmissão
6.
Langmuir ; 31(42): 11564-73, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26439800

RESUMO

Cell-membrane lipid composition can greatly influence biophysical properties of cell membranes, affecting various cellular functions. We previously showed that lipid synthesis becomes altered in the membranes of resistant breast cancer cells (MCF-7/ADR); they form a more rigid, hydrophobic lipid monolayer than do sensitive cell membranes (MCF-7). These changes in membrane lipids of resistant cells, attributed to epigenetic aberration, significantly affected drug transport and endocytic function, thus impacting the efficacy of anticancer drugs. The present study's objective was to determine the effects of the epigenetic drug, 5-aza-2'-deoxycytidine (DAC), delivered in sustained-release nanogels (DAC-NGs), on the composition and biophysical properties of membrane lipids of resistant cells. Resistant and sensitive cells were treated with DAC in solution (DAC-sol) or DAC-NGs, and cell-membrane lipids were isolated and analyzed for lipid composition and biophysical properties. In resistant cells, we found increased formation of cholesterol-sphingomyelin (CHOL-SM) rafts with culturing time, whereas DAC treatment reduced their formation. In general, the effect of DAC-NGs was greater in changing the lipid composition than with DAC-sol. DAC treatment also caused a rise in levels of certain phospholipids and neutral lipids known to increase membrane fluidity, while reducing the levels of certain lipids known to increase membrane rigidity. Isotherm data showed increased lipid membrane fluidity following DAC treatment, attributed to decrease levels of CHOL-SM rafts (lamellar beta [Lß] structures or ordered gel) and a corresponding increase in lipids that form lamellar alpha-structures (Lα, liquid crystalline phase). Sensitive cells showed marginal or insignificant changes in lipid profile following DAC-treatment, suggesting that epigenetic changes affecting lipid biosynthesis are more specific to resistant cells. Since membrane fluidity plays a major role in drug transport and endocytic function, treatment of resistant cells with epigenetic drugs with altered lipid profile could facilitate anticancer drug transport to overcome acquired drug resistance in a combination therapy.


Assuntos
Colesterol/química , Lipídeos de Membrana/química , Esfingomielinas/química , Neoplasias da Mama/metabolismo , Feminino , Humanos
7.
Langmuir ; 30(25): 7522-32, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24911361

RESUMO

To be effective for cytoplasmic delivery of therapeutics, nanoparticles (NPs) taken up via endocytic pathways must efficiently transport across the cell membrane and subsequently escape from the secondary endosomes. We hypothesized that the biomechanical and thermodynamic interactions of NPs with plasma and endosomal membrane lipids are involved in these processes. Using model plasma and endosomal lipid membranes, we compared the interactions of cationic NPs composed of poly(D,L-lactide-co-glycolide) modified with the dichain surfactant didodecyldimethylammonium bromide (DMAB) or the single-chain surfactant cetyltrimethylammonium bromide (CTAB) vs anionic unmodified NPs of similar size. We validated our hypothesis in doxorubicin-sensitive (MCF-7, with relatively fluid membranes) and resistant breast cancer cells (MCF-7/ADR, with rigid membranes). Despite their cationic surface charges, DMAB- and CTAB-modified NPs showed different patterns of biophysical interaction: DMAB-modified NPs induced bending of the model plasma membrane, whereas CTAB-modified NPs condensed the membrane, thereby resisted bending. Unmodified NPs showed no effects on bending. DMAB-modified NPs also induced thermodynamic instability of the model endosomal membrane, whereas CTAB-modified and unmodified NPs had no effect. Since bending of the plasma membrane and destabilization of the endosomal membrane are critical biophysical processes in NP cellular uptake and endosomal escape, respectively, we tested these NPs for cellular uptake and drug efficacy. Confocal imaging showed that in both sensitive and resistant cells DMAB-modified NPs exhibited greater cellular uptake and escape from endosomes than CTAB-modified or unmodified NPs. Further, paclitaxel-loaded DMAB-modified NPs induced greater cytotoxicity even in resistant cells than CTAB-modified or unmodified NPs or drug in solution, demonstrating the potential of DMAB-modified NPs to overcome the transport barrier in resistant cells. In conclusion, biomechanical interactions with membrane lipids are involved in cellular uptake and endosomal escape of NPs. Biophysical interaction studies could help us better understand the role of membrane lipids in cellular uptake and intracellular trafficking of NPs.


Assuntos
Lipídeos de Membrana/química , Nanopartículas/química , Cetrimônio , Compostos de Cetrimônio/química , Humanos , Células MCF-7 , Termodinâmica
8.
Adv Exp Med Biol ; 811: 73-91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24683028

RESUMO

Understanding the interactions of nanoparticles (NPs) with cells and how these interactions influence their cellular uptake is essential to exploring the biomedical applications of NPs, particularly for drug delivery. Various factors, whether differences in physical properties of NPs or variations in cell-membrane characteristics, influence NP-cell interactions and uptake processes. NP-cell membrane interactions may also influence intracellular trafficking of NPs, their sorting into different intracellular compartments, cellular retention, and hence the efficacy of encapsulated therapeutics. A crucial consideration is whether such interactions might cause any toxicity, starting with how NPs interact in transit with the biological environment prior to their interactions with targeted cells and tissues. Understanding the effects of various NP characteristics on cellular and biological processes could help in designing NPs that are efficient but also nontoxic.


Assuntos
Células/metabolismo , Endocitose , Nanopartículas/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Sistemas de Liberação de Medicamentos , Humanos , Nanopartículas/química , Nanopartículas/toxicidade , Tamanho da Partícula , Propriedades de Superfície
9.
Mol Pharm ; 10(1): 337-52, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23215027

RESUMO

Epigenetic alterations such as aberrant DNA methylation and histone modifications contribute substantially to both the cause and maintenance of drug resistance. These epigenetic changes lead to silencing of tumor suppressor genes involved in key DNA damage-response pathways, making drug-resistant cancer cells nonresponsive to conventional anticancer drug therapies. Our hypothesis is that treating drug-resistant cells with epigenetic drugs could restore the sensitivity to anticancer drugs by reactivating previously silenced genes. To test our hypothesis, we used drug-resistant breast cancer cells (MCF-7/ADR) and two epigenetic drugs that act via different mechanisms--5-aza-2'-deoxycytidine (decitabine, DAC), a demethylating agent, and suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor--in combination with doxorubicin. We show that the sequential treatment of resistant cells, first with an epigenetic drug (DAC), and then with doxorubicin, induces a highly synergistic effect, thus reducing the IC(50) of doxorubicin by several thousand fold. The sequential treatment caused over 90% resistant cells to undergo G2/M cell cycle arrest, determined to be due to upregulation of p21(WAF1/CIP1) expression, which is responsible for cell-cycle regulation. The induction of p21(WAF1/CIP1) correlated well with the depletion of DNA methyltransferase1 (DNMT1), an enzyme that promotes methylation of DNA, suggesting that the p21(WAF1/CIP1) gene may have been methylated and hence is inactive in MCF-7/ADR cells. Microarray analysis shows expression of several tumor suppressor genes and downregulation of tumor promoter genes, particularly in sequentially treated resistant cells. Sequential treatment was found to be significantly more effective than simultaneous treatment, and DAC was more effective than SAHA in overcoming doxorubicin resistance. Synergistic effect with sequential treatment was also seen in drug-sensitive breast cancer cells, but the effect was significantly more pronounced in resistant cells. In conclusion, the sequential treatment of an epigenetic drug in combination with doxorubicin induces a highly synergistic effect that overcomes doxorubicin resistance in breast cancer cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinógenos/farmacologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Decitabina , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Epigênese Genética , Epigenômica/métodos , Feminino , Fase G2/efeitos dos fármacos , Fase G2/genética , Genes Supressores de Tumor , Humanos , Ácidos Hidroxâmicos/farmacologia , Internacionalidade , Células MCF-7 , Vorinostat
10.
Biomedicines ; 11(3)2023 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-36979759

RESUMO

Stroke is a leading cause of death, long-term disability, and socioeconomic costs, highlighting the urgent need for effective treatment. During acute phase, intravenous administration of recombinant tissue plasminogen activator (tPA), a thrombolytic agent, and endovascular thrombectomy (EVT), a mechanical intervention to retrieve clots, are the only FDA-approved treatments to re-establish cerebral blood flow. Due to a short therapeutic time window and high potential risk of cerebral hemorrhage, a limited number of acute stroke patients benefit from tPA treatment. EVT can be performed within an extended time window, but such intervention is performed only in patients with occlusion in a larger, anatomically more proximal vasculature and is carried out at specialty centers. Regardless of the method, in case of successful recanalization, ischemia-reperfusion injury represents an additional challenge. Further, tPA disrupts the blood-brain barrier integrity and is neurotoxic, aggravating reperfusion injury. Nanoparticle-based approaches have the potential to circumvent some of the above issues and develop a thrombolytic agent that can be administered safely beyond the time window for tPA treatment. Different attributes of nanoparticles are also being explored to develop a multifunctional thrombolytic agent that, in addition to a thrombolytic agent, can contain therapeutics such as an anti-inflammatory, antioxidant, neuro/vasoprotective, or imaging agent, i.e., a theragnostic agent. The focus of this review is to highlight these advances as they relate to cerebrovascular conditions to improve clinical outcomes in stroke patients.

11.
J Control Release ; 357: 498-510, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37059400

RESUMO

Bone metastasis at an advanced disease stage is common in most solid tumors and is untreatable. Overexpression of receptor activator of nuclear factor κB ligand (RANKL) in tumor-bone marrow microenvironment drives a vicious cycle of tumor progression and bone resorption. Biodegradable nanoparticles (NPs), designed to localize in the tumor tissue in bone marrow, were evaluated in a prostate cancer model of bone metastasis. The combination treatment, encapsulating docetaxel, an anticancer drug (TXT-NPs), and Denosumab, a monoclonal antibody that binds to RANKL (DNmb-NPs), administered intravenously regressed the tumor completely, preventing bone resorption, without causing any mortality. With TXT-NPs alone treatment, after an initial regression, the tumor relapsed and acquired resistance, whereas DNmb-NPs alone treatment was ineffective. Only in the combination treatment, RANKL was not detected in the tumor tibia, thus negating its role in tumor progression and bone resorption. The combination treatment was determined to be safe as the vital organ tissue showed no increase in inflammatory cytokine or the liver ALT/AST levels, and animals gained weight. Overall, dual drug treatment acted synergistically to modulate the tumor-bone microenvironment with encapsulation enhancing their therapeutic potency to achieve tumor regression.


Assuntos
Neoplasias Ósseas , Reabsorção Óssea , Nanopartículas , Masculino , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Reabsorção Óssea/prevenção & controle , Combinação de Medicamentos , Microambiente Tumoral
12.
J Control Release ; 364: 109-123, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37866402

RESUMO

Traumatic spinal cord injury exacerbates disability with time due to secondary injury cascade triggered largely by overproduction of reactive oxygen species (ROS) at the lesion site, causing oxidative stress. This study explored nanoparticles containing antioxidant enzymes (antioxidant NPs) to neutralize excess ROS at the lesion site and its impact. When tested in a rat contusion model of spinal cord injury, a single dose of antioxidant NPs, administered intravenously three hours after injury, effectively restored the redox balance at the lesion site, interrupting the secondary injury progression. This led to reduced spinal cord tissue inflammation, apoptosis, cavitation, and inhibition of syringomyelia. Moreover, the treatment reduced scar tissue forming collagen at the lesion site, protected axons from demyelination, and stimulated lesion healing, with further analysis indicating the formation of immature neurons. The ultimate effect of the treatment was improved motor and sensory functions and rapid post-injury weight loss recovery. Histological analysis revealed activated microglia in the spinal cord displaying rod-shaped anti-inflammatory and regenerative phenotype in treated animals, contrasting with amoeboid inflammatory and degenerative phenotype in untreated control. Overall data suggest that restoring redox balance at the lesion site shifts the dynamics in the injured spinal cord microenvironment from degenerative to regenerative, potentially by promoting endogenous repair mechanisms. Antioxidant NPs show promise to be developed as an early therapeutic intervention in stabilizing injured spinal cord for enhanced recovery.


Assuntos
Nanopartículas , Traumatismos da Medula Espinal , Ratos , Animais , Antioxidantes/uso terapêutico , Antioxidantes/farmacologia , Espécies Reativas de Oxigênio , Traumatismos da Medula Espinal/tratamento farmacológico , Medula Espinal/patologia , Recuperação de Função Fisiológica
13.
Mol Pharm ; 9(9): 2730-42, 2012 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-22817326

RESUMO

In our recent studies exploring the biophysical characteristics of resistant cell lipids, and the role they play in drug transport, we demonstrated the difference of drug-resistant breast cancer cells from drug-sensitive cells in lipid composition and biophysical properties, suggesting that cancer cells acquire a drug-resistant phenotype through the alteration of lipid synthesis to inhibit intracellular drug transport to protect from cytotoxic effect. In cancer cells, epigenetic changes (e.g., DNA hypermethylation) are essential to maintain this drug-resistant phenotype. Thus, altered lipid synthesis may be linked to epigenetic mechanisms of drug resistance. We hypothesize that reversing DNA hypermethylation in resistant cells with an epigenetic drug could alter lipid synthesis, changing the cell membrane's biophysical properties to facilitate drug delivery to overcome drug resistance. Herein we show that treating drug-resistant breast cancer cells (MCF-7/ADR) with the epigenetic drug 5-aza-2'-deoxycytidine (decitabine) significantly alters cell lipid composition and biophysical properties, causing the resistant cells to acquire biophysical characteristics similar to those of sensitive cell (MCF-7) lipids. Following decitabine treatment, resistant cells demonstrated increased sphingomyelinase activity, resulting in a decreased sphingomyelin level that influenced lipid domain structures, increased membrane fluidity, and reduced P-glycoprotein expression. Changes in the biophysical characteristics of resistant cell lipids facilitated doxorubicin transport and restored endocytic function for drug delivery with a lipid-encapsulated form of doxorubicin, enhancing the drug efficacy. In conclusion, we have established a new mechanism for efficacy of an epigenetic drug, mediated through changes in lipid composition and biophysical properties, in reversing cancer drug resistance.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/genética , Metabolismo dos Lipídeos/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Neoplasias da Mama/tratamento farmacológico , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Metilação de DNA/efeitos dos fármacos , Decitabina , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética , Epigenômica/métodos , Feminino , Humanos , Células MCF-7
14.
Antioxidants (Basel) ; 11(2)2022 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-35204290

RESUMO

Free radicals are formed as a part of normal metabolic activities but are neutralized by the endogenous antioxidants present in cells/tissue, thus maintaining the redox balance. This redox balance is disrupted in certain neuropathophysiological conditions, causing oxidative stress, which is implicated in several progressive neurodegenerative diseases. Following neuronal injury, secondary injury progression is also caused by excessive production of free radicals. Highly reactive free radicals, mainly the reactive oxygen species (ROS) and reactive nitrogen species (RNS), damage the cell membrane, proteins, and DNA, which triggers a self-propagating inflammatory cascade of degenerative events. Dysfunctional mitochondria under oxidative stress conditions are considered a key mediator in progressive neurodegeneration. Exogenous delivery of antioxidants holds promise to alleviate oxidative stress to regain the redox balance. In this regard, natural and synthetic antioxidants have been evaluated. Despite promising results in preclinical studies, clinical translation of antioxidants as a therapy to treat neurodegenerative diseases remains elusive. The issues could be their low bioavailability, instability, limited transport to the target tissue, and/or poor antioxidant capacity, requiring repeated and high dosing, which cannot be administered to humans because of dose-limiting toxicity. Our laboratory is investigating nanoparticle-mediated delivery of antioxidant enzymes to address some of the above issues. Apart from being endogenous, the main advantage of antioxidant enzymes is their catalytic mechanism of action; hence, they are significantly more effective at lower doses in detoxifying the deleterious effects of free radicals than nonenzymatic antioxidants. This review provides a comprehensive analysis of the potential of antioxidant therapy, challenges in their clinical translation, and the role nanoparticles/drug delivery systems could play in addressing these challenges.

15.
FASEB J ; 23(5): 1384-95, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19124559

RESUMO

Excessive production of reactive oxygen species (ROS) after cerebral ischemia and reperfusion is implicated in brain damage through different cellular and molecular mechanisms, and it is further aggravated by impaired cellular antioxidant defense systems under ischemic conditions. Therapeutic strategies based on exogenous delivery of the native form of superoxide dismutase (SOD), a free radical scavenger, are limited because of its short half-life (approximately 6 min) in vivo and poor permeability across the blood-brain-barrier (BBB). We encapsulated SOD in biodegradable poly(D,L-lactide co-glycolide) nanoparticles (SOD-NPs) and tested their efficacy in a rat focal cerebral ischemia-reperfusion injury model. We hypothesized that localized brain delivery of SOD-NPs would sustain the protective effect of SOD by neutralizing the deleterious effects of ROS formed following ischemia-reperfusion. SOD-NPs were administered at the time of reperfusion via the intracarotid route to maximize their localization in the brain. Animals receiving SOD-NPs (10,000 U of SOD/kg) demonstrated a 65% reduction in infarct volume, whereas an equivalent dose of SOD in solution (SOD-Sol) increased it by 25% over saline control (P<0.001; data at 6 h following reperfusion). Control NPs alone or mixed with SOD-Sol were ineffective in reducing infract volume, with results similar to saline control, indicating the protective effect of the encapsulated enzyme. SOD-NPs maintained BBB integrity, thereby preventing edema, reduced the level of ROS formed following reperfusion, and protected neurons from undergoing apoptosis. Animals treated with SOD-NPs demonstrated greater survival than those with saline control (75% vs. 0% at 28 days) and later regained most vital neurological functions. SOD-NPs may be an effective treatment option in conjunction with a thrombolytic agent for stroke patients.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Nanopartículas , Traumatismo por Reperfusão/tratamento farmacológico , Superóxido Dismutase/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Infarto Cerebral/prevenção & controle , Ácido Láctico , Masculino , Nanopartículas/administração & dosagem , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/prevenção & controle , Superóxido Dismutase/administração & dosagem
16.
Mol Pharm ; 7(6): 1880-98, 2010 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-20866097

RESUMO

Next to surgical resection, tumor ablation is a commonly used intervention in the treatment of solid tumors. Tumor ablation methods include thermal therapies, photodynamic therapy, and reactive oxygen species (ROS) producing agents. Thermal therapies induce tumor cell death via thermal energy and include radiofrequency, microwave, high intensity focused ultrasound, and cryoablation. Photodynamic therapy and ROS producing agents cause increased oxidative stress in tumor cells leading to apoptosis. While these therapies are safe and viable alternatives when resection of malignancies is not feasible, they do have associated limitations that prevent their widespread use in clinical applications. To improve the efficacy of these treatments, nanoparticles are being studied in combination with nonsurgical ablation regimens. In addition to better thermal effect on tumor ablation, nanoparticles can deliver anticancer therapeutics that show a synergistic antitumor effect in the presence of heat and can also be imaged to achieve precision in therapy. Understanding the molecular mechanism of nanoparticle-mediated tumor ablation could further help engineer nanoparticles of appropriate composition and properties to synergize the ablation effect. This review aims to explore the various types of nonsurgical tumor ablation methods currently used in cancer treatment and potential improvements by nanotechnology applications.


Assuntos
Nanomedicina/métodos , Nanopartículas/uso terapêutico , Neoplasias/terapia , Humanos
17.
Mol Pharm ; 7(6): 2334-48, 2010 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-20958074

RESUMO

Understanding the role of lipids in drug transport is critical in cancer chemotherapy to overcome drug resistance. In this study, we isolated lipids from doxorubicin-sensitive (MCF-7) and -resistant (MCF-7/ADR) breast cancer cells to characterize the biophysical properties of membrane lipids (particularly lipid packing and membrane fluidity) and to understand the role of the interaction of cell membrane lipids with drug/nanocarrier on drug uptake and efficacy. Resistant cell membrane lipids showed significantly different composition and formed more condensed, less fluid monolayers than did lipids from sensitive cells. Doxorubicin, used as a model anticancer agent, showed a strong hydrophobic interaction with resistant cell membrane lipids but significantly less interaction, as well as a different pattern of interaction (i.e., ionic), with sensitive ones. The threshold intracellular doxorubicin concentration required to produce an antiproliferative effect was similar for both sensitive and resistant cell lines, suggesting that drug transport is a major barrier in determining drug efficacy in resistant cells. In addition to the biophysical characteristics of resistant cell membrane lipids, lipid-doxorubicin interactions appear to decrease intracellular drug transport via diffusion as the drug is trapped in the lipid bilayer. The rigid nature of resistant cell membranes also seems to influence endosomal functions that inhibit drug uptake when a liposomal formulation of doxorubicin is used. In conclusion, biophysical properties of resistant cell membrane lipids significantly influence drug transport, and hence drug efficacy. A better understanding of the mechanisms of cancer drug resistance is vital to developing more effective therapeutic interventions. In this regard, biophysical interaction studies with cell membrane lipids might be helpful to improve drug transport and efficacy through drug discovery and/or drug delivery approaches by overcoming the lipid barrier in resistant cells.


Assuntos
Neoplasias da Mama/metabolismo , Doxorrubicina/química , Doxorrubicina/metabolismo , Resistencia a Medicamentos Antineoplásicos , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Células Tumorais Cultivadas
18.
Pharm Res ; 27(11): 2283-95, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20845067

RESUMO

PURPOSE: Polyethylene glycol (PEG) functionalized magnetic nanoparticles (MNPs) were tested as a drug carrier system, as a magnetic resonance imaging (MRI) agent, and for their ability to conjugate to an antibody. METHODS: An iron oxide core coated with oleic acid (OA) and then with OA-PEG forms a water-dispersible MNP formulation. Hydrophobic doxorubicin partitions into the OA layer for sustained drug delivery. The T(1) and T(2) MRI contrast properties were determined in vitro and the circulation of the MNPs was measured in mouse carotid arteries. An N-hydroxysuccinimide group (NHS) on the OA-PEG-80 was used to conjugate the amine functional group on antibodies for active targeting in the human MCF-7 breast cancer cell line. RESULTS: The optimized formulation had a mean hydrodynamic diameter of 184 nm with an ~8 nm iron-oxide core. The MNPs enhance the T(2) MRI contrast and have a long circulation time in vivo with 30% relative concentration 50 min post-injection. Doxorubicin-loaded MNPs showed sustained drug release and dose-dependent antiproliferative effects in vitro; the drug effect was enhanced with transferrin antibody-conjugated MNPs. CONCLUSION: PEG-functionalized MNPs could be developed as a targeted drug delivery system and MRI contrast agent.


Assuntos
Sistemas de Liberação de Medicamentos , Imageamento por Ressonância Magnética , Magnetismo , Nanopartículas , Polietilenoglicóis , Animais , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos
19.
Int J Nanomedicine ; 15: 3639-3647, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32547019

RESUMO

PURPOSE: Astrocyte dysfunction is a hallmark of central nervous system injury or infection. As a primary contributor to neurodegeneration, astrocytes are an ideal therapeutic target to combat neurodegenerative conditions. Gene therapy has arisen as an innovative technique that provides excellent prospect for disease intervention. Poly (lactide-co-glycolide) (PLGA) and polyethylenimine (PEI) are polymeric nanoparticles commonly used in gene delivery, each manifesting their own set of advantages and disadvantages. As a clinically approved polymer by the Federal Drug Administration, well characterized for its biodegradability and biocompatibility, PLGA-based nanoparticles (PLGA-NPs) are appealing for translational gene delivery systems. However, our investigations revealed PLGA-NPs were ineffective at facilitating exogenous gene expression in primary human astrocytes, despite their success in other cell lines. Furthermore, PEI polymers illustrate high delivery efficiency but induce cytotoxicity. The purpose of this study is to develop viable and biocompatible NPsystem for astrocyte-targeted gene therapy. MATERIALS AND METHODS: Successful gene expression by PLGA-NPs alone or in combination with arginine-modified PEI polymers (AnPn) was assessed by a luciferase reporter gene encapsulated in PLGA-NPs. Cytoplasmic release and nuclear localization of DNA were investigated using fluorescent confocal imaging with YOYO-labeled plasmid DNA (pDNA). NP-mediated cytotoxicity was assessed via lactate dehydrogenase in primary human astrocytes and neurons. RESULTS: Confocal imaging of YOYO-labeled pDNA confirmed PLGA-NPs delivered pDNA to the cytoplasm in a dose and time-dependent manner. However, co-staining revealed pDNA delivered by PLGA-NPs did not localize to the nucleus. The addition of AnPn significantly improved nuclear localization of pDNA and successfully achieved gene expression in primary human astrocytes. Moreover, these formulations were biocompatible with both astrocytes and neurons. CONCLUSION: By co-transfecting two polymeric NPs, we developed an improved system for gene delivery and expression in primary human astrocytes. These findings provide a basis for a biocompatible and clinically translatable method to regulate astrocyte function during neurodegenerative diseases and disorders.


Assuntos
Arginina/química , Astrócitos/metabolismo , Técnicas de Transferência de Genes , Nanopartículas/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , DNA/genética , Células HEK293 , Humanos , Tamanho da Partícula , Plasmídeos/genética , Polietilenoimina , Transfecção
20.
J Control Release ; 317: 300-311, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31805339

RESUMO

In spinal cord injury (SCI), the initial damage leads to a rapidly escalating cascade of degenerative events, known as secondary injury. Loss of mitochondrial homeostasis after SCI, mediated primarily by oxidative stress, is considered to play a crucial role in the proliferation of secondary injury cascade. We hypothesized that effective exogenous delivery of antioxidant enzymes - superoxide dismutase (SOD) and catalase (CAT), encapsulated in biodegradable nanoparticles (nano-SOD/CAT) - at the lesion site would protect mitochondria from oxidative stress, and hence the spinal cord from secondary injury. Previously, in a rat contusion model of severe SCI, we demonstrated extravasation and retention of intravenously administered nanoparticles specifically at the lesion site. To test our hypothesis, a single dose of nano-SOD/CAT in saline was administered intravenously 6 h post-injury, and the spinal cords were analyzed one week post-treatment. Mitochondria isolated from the affected region of the spinal cord of nano-SOD/CAT-treated animals demonstrated significantly reduced mitochondrial reactive oxygen species (ROS) activities, increased mitochondrial membrane potential, reduced calcium levels, and also higher adenosine triphosphate (ATP) production capacity than those isolated from the spinal cords of untreated control or SOD/CAT solution treated animals. Although the treatment did not achieve the same mitochondrial function as in the spinal cords of sham control animals, it significantly attenuated mitochondrial dysfunction following SCI. Further, immunohistochemical analyses of the spinal cords of treated animals showed significantly lower ROS, cleaved caspase-3, and cytochrome c activities, leading to reduced spinal cord neuronal cell apoptosis and smaller lesion area than in untreated animals. These results imply that the treatment significantly attenuated progression of secondary injury that was also reflected from less weight loss and improved locomotive recovery of treated vs. untreated animals. In conclusion, nano-SOD/CAT mitigated activation of cascade of degenerating factors by protecting mitochondria and hence the spinal cord from secondary injury. An effective treatment during the acute phase following SCI could potentially have a positive long-term impact on neurological and functional recovery.


Assuntos
Nanopartículas , Traumatismos da Medula Espinal , Animais , Antioxidantes/metabolismo , Apoptose , Mitocôndrias/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA