Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 324(2): L190-L198, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36625494

RESUMO

Pulmonary fibrosis is characterized by the accumulation of myofibroblasts in the lung and progressive tissue scarring. Fibroblasts exist across a spectrum of states, from quiescence in health to activated myofibroblasts in the setting of injury. Highly activated myofibroblasts have a critical role in the establishment of fibrosis as the predominant source of type 1 collagen and profibrotic mediators. Myofibroblasts are also highly contractile cells and can alter lung biomechanical properties through tissue contraction. Inhibiting signaling pathways involved in myofibroblast activation could therefore have significant therapeutic value. One of the ways myofibroblast activation occurs is through activation of the Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF) pathway, which signals through intracellular actin polymerization. However, concerns surrounding the pleiotropic and ubiquitous nature of these signaling pathways have limited the translation of inhibitory drugs. Herein, we demonstrate a novel therapeutic antifibrotic strategy using myofibroblast-targeted nanoparticles containing a MTRF/SRF pathway inhibitor (CCG-1423), which has been shown to block myofibroblast activation in vitro. Myofibroblasts were preferentially targeted via the angiotensin 2 receptor, which has been shown to be selectively upregulated in animal and human studies. These nanoparticles were nontoxic and accumulated in lung myofibroblasts in the bleomycin-induced mouse model of pulmonary fibrosis, reducing the number of these activated cells and their production of profibrotic mediators. Ultimately, in a murine model of lung fibrosis, a single injection of these drugs containing targeted nanoagents reduced fibrosis as compared with control mice. This approach has the potential to deliver personalized therapy by precisely targeting signaling pathways in a cell-specific manner, allowing increased efficacy with reduced deleterious off-target effects.


Assuntos
Fibrose Pulmonar , Fatores de Transcrição , Humanos , Animais , Camundongos , Fatores de Transcrição/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/prevenção & controle , Miofibroblastos/metabolismo , Fator de Resposta Sérica/metabolismo , Quinases Associadas a rho/metabolismo , Fibrose , Pulmão/metabolismo , Nanotecnologia , Diferenciação Celular
2.
J Am Soc Nephrol ; 31(5): 931-945, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32152232

RESUMO

BACKGROUND: Arteriovenous fistulas placed surgically for dialysis vascular access have a high primary failure rate resulting from excessive inward remodeling, medial fibrosis, and thrombosis. No clinically established pharmacologic or perisurgical therapies currently address this unmet need. Statins' induction of multiple anti-inflammatory and antithrombotic effects suggests that these drugs might reduce arteriovenous fistula failure. Yet, the in vivo physiologic and molecular effects of statins on fistula patency and maturation remain poorly understood. METHODS: We randomized 108 C57Bl/6J mice to receive daily atorvastatin 1.14 mg/kg or PBS (control) starting 7 days before end-to-side carotid artery-jugular vein fistula creation and for up to 42 days after fistula creation. We then assessed longitudinally the effects of statin therapy on primary murine fistula patency and maturation. We concomitantly analyzed the in vivo arteriovenous fistula thrombogenic and inflammatory macrophage response to statin therapy, using the fibrin-targeted, near-infrared fluorescence molecular imaging agent FTP11-CyAm7 and dextranated, macrophage-avid nanoparticles CLIO-VT680. RESULTS: In vivo molecular-structural imaging demonstrated that atorvastatin significantly reduced fibrin deposition at day 7 and macrophage accumulation at days 7 and 14, findings supported by histopathologic and gene-expression analyses. Structurally, atorvastatin promoted favorable venous limb outward remodeling, preserved arteriovenous fistula blood flow, and prolonged primary arteriovenous fistula patency through day 42 (P<0.05 versus control for all measures). CONCLUSIONS: These findings provide new in vivo evidence that statins improve experimental arteriovenous fistula patency and maturation, indicating that additional clinical evaluation of statin therapy in patients on dialysis undergoing arteriovenous fistula placement is warranted.


Assuntos
Derivação Arteriovenosa Cirúrgica , Atorvastatina/uso terapêutico , Fibrina/metabolismo , Macrófagos/efeitos dos fármacos , Grau de Desobstrução Vascular/efeitos dos fármacos , Animais , Atorvastatina/farmacologia , Artéria Carótida Interna , Colágeno/metabolismo , Feminino , Fibrose/prevenção & controle , Hemorreologia , Inflamação/prevenção & controle , Veias Jugulares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Imagem Molecular , Nanopartículas , RNA Mensageiro/biossíntese , Distribuição Aleatória , Trombose/prevenção & controle , Transcrição Gênica , Dispositivos de Acesso Vascular
3.
Eur Heart J ; 38(6): 447-455, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-26685129

RESUMO

AIMS: Fibrin deposition and absent endothelium characterize unhealed stents that are at heightened risk of stent thrombosis. Optical coherence tomography (OCT) is increasingly used for assessing stent tissue coverage as a measure of healed stents, but cannot precisely identify whether overlying tissue represents physiological neointima. Here we assessed and compared fibrin deposition and persistence on bare metal stent (BMS) and drug-eluting stent (DES) using near-infrared fluorescence (NIRF) molecular imaging in vivo, in combination with simultaneous OCT stent coverage. METHODS AND RESULTS: Rabbits underwent implantation of one BMS and one DES without overlap in the infrarenal aorta (N = 20 3.5 × 12 mm). At Days 7 and/or 28, intravascular NIRF-OCT was performed following the injection of fibrin-targeted NIRF molecular imaging agent FTP11-CyAm7. Intravascular NIRF-OCT enabled high-resolution imaging of fibrin overlying stent struts in vivo, as validated by histopathology. Compared with BMS, DES showed greater fibrin deposition and fibrin persistence at Days 7 and 28 (P < 0.01 vs. BMS). Notably, for edge stent struts identified as covered by OCT on Day 7, 92.8 ± 9.5% of DES and 55.8 ± 23.6% of BMS struts were NIRF fibrin positive (P < 0.001). At Day 28, 18.6 ± 10.6% (DES) and 5.1 ± 8.7% (BMS) of OCT-covered struts remained fibrin positive (P < 0.001). CONCLUSION: Intravascular NIRF fibrin molecular imaging improves the detection of unhealed stents, using clinically translatable technology that complements OCT. A sizeable percentage of struts deemed covered by OCT are actually covered by fibrin, particularly in DES, and therefore such stents might remain prothrombotic. These findings have implications for the specificity of standalone clinical OCT assessments of stent healing.

4.
Clin Rev Bone Miner Metab ; 16(4): 142-158, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30930699

RESUMO

Bone fractures create five problems that must be resolved: bleeding, risk of infection, hypoxia, disproportionate strain, and inability to bear weight. There have been enormous advancements in our understanding of the molecular mechanisms that resolve these problems after fractures, and in best clinical practices of repairing fractures. We put forth a modern, comprehensive model of fracture repair that synthesizes the literature on the biology and biomechanics of fracture repair to address the primary problems of fractures. This updated model is a framework for both fracture management and future studies aimed at understanding and treating this complex process. This model is based upon the fracture acute phase response (APR), which encompasses the molecular mechanisms that respond to injury. The APR is divided into sequential stages of "survival" and "repair." Early in convalescence, during "survival," bleeding and infection are resolved by collaborative efforts of the hemostatic and inflammatory pathways. Later, in "repair," avascular and biomechanically insufficient bone is replaced by a variable combination of intramembranous and endochondral ossification. Progression to repair cannot occur until survival has been ensured. A disproportionate APR-either insufficient or exuberant-leads to complications of survival (hemorrhage, thrombosis, systemic inflammatory response syndrome, infection, death) and/or repair (delayed- or non-union). The type of ossification utilized for fracture repair is dependent on the relative amounts of strain and vascularity in the fracture microenvironment, but any failure along this process can disrupt or delay fracture healing and result in a similar non-union. Therefore, incomplete understanding of the principles herein can result in mismanagement of fracture care or application of hardware that interferes with fracture repair. This unifying model of fracture repair not only informs clinicians how their interventions fit within the framework of normal biological healing but also instructs investigators about the critical variables and outputs to assess during a study of fracture repair.

5.
Circ Res ; 117(6): 502-512, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26199323

RESUMO

RATIONALE: The development of molecular imaging approaches that assess specific immunopathologic mechanisms can advance the study of myocarditis. OBJECTIVE: This study validates a novel molecular imaging tool that enables the in vivo visualization of granzyme B activity, a major effector of cytotoxic CD8+ T lymphocytes. METHODS AND RESULTS: We synthesized and optimized a fluorogenic substrate capable of reporting on granzyme B activity and examined its specificity ex vivo in mice hearts with experimental cytotoxic CD8+ T lymphocyte-mediated myocarditis using fluorescence reflectance imaging, validated by histological examination. In vivo experiments localized granzyme B activity in hearts with acute myocarditis monitored by fluorescent molecular tomography in conjunction with coregistered computed tomography imaging. A model anti-inflammatory intervention (dexamethasone administration) in vivo reduced granzyme B activity (vehicle versus dexamethasone: 504±263 versus 194±77 fluorescence intensities in hearts; P=0.002). CONCLUSIONS: Molecular imaging of granzyme B activity can visualize T cell-mediated myocardial injury and monitor the response to an anti-inflammatory intervention.


Assuntos
Granzimas/metabolismo , Miocardite/enzimologia , Miocardite/imunologia , Animais , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/imunologia , Ativação Enzimática/fisiologia , Corantes Fluorescentes/análise , Granzimas/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocardite/patologia
6.
Arterioscler Thromb Vasc Biol ; 35(1): 189-96, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25395614

RESUMO

OBJECTIVE: In vivo assessment of pathological endothelium within arteriovenous fistula (AVF) could provide new insights into inflow stenosis, a common cause of AVF primary failure in end-stage renal disease patients. Here we developed nanoparticle-based imaging strategies to assess pathological endothelium in vivo and elucidate its relationship to neointimal hyperplasia formation in AVF. APPROACH AND RESULTS: Jugular-carotid AVFs were created in C57BL/6 mice (n=38). Pathological endothelium in the AVF was visualized and quantified in vivo using dextranated magnetofluorescent nanoparticles (CLIO-VT680 [cross-linked iron oxide-VivoTag680]). At day 14, CLIO-VT680 was deposited in AVF, but only minimally in sham-operated arteries. Transmission electron microscopy revealed that CLIO-VT680 resided within endothelial cells and in the intimal extracellular space. Endothelial cells of AVF, but not control arteries, expressed vascular cell adhesion molecule-1 and showed augmented endothelial permeability near the anastomosis. Intravital microscopy demonstrated that CLIO-VT680 deposited most intensely near the AVF anastomosis (P<0.0001). The day 14 intravital microscopy CLIO-VT680 signal predicted the subsequent site and magnitude of AVF neointimal hyperplasia at day 42 (r=0.58, P<0.05). CLIO-VT680 deposition in AVF was further visualized by ex vivo MRI. CONCLUSIONS: AVF develop a pathological endothelial response that can be assessed in vivo via nanoparticle-enhanced imaging. AVF endothelium is activated and exhibits augmented permeability, offering a targeting mechanism for nanoparticle deposition and retention in pathological endothelium. The in vivo AVF nanoparticle signal identified and predicted subsequent inflow neointimal hyperplasia. This approach could be used to test therapeutic interventions aiming to restore endothelial health and to decrease early AVF failure caused by inflow stenosis.


Assuntos
Fístula Arteriovenosa/patologia , Artérias Carótidas/patologia , Dextranos , Endotélio Vascular/patologia , Corantes Fluorescentes , Veias Jugulares/patologia , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita , Microscopia de Fluorescência , Animais , Fístula Arteriovenosa/metabolismo , Fístula Arteriovenosa/fisiopatologia , Velocidade do Fluxo Sanguíneo , Permeabilidade Capilar , Artérias Carótidas/metabolismo , Artérias Carótidas/fisiopatologia , Artérias Carótidas/cirurgia , Artérias Carótidas/ultraestrutura , Proliferação de Células , Constrição Patológica , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Endotélio Vascular/cirurgia , Endotélio Vascular/ultraestrutura , Hiperplasia , Veias Jugulares/metabolismo , Veias Jugulares/fisiopatologia , Veias Jugulares/cirurgia , Veias Jugulares/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Neointima , Valor Preditivo dos Testes , Fluxo Sanguíneo Regional , Fatores de Tempo , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
Adv Mater ; 36(8): e2304615, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37934471

RESUMO

The spleen is an important mediator of both adaptive and innate immunity. As such, attempts to modulate the immune response provided by the spleen may be conducive to improved outcomes for numerous diseases throughout the body. Here, biomimicry is used to rationally design nanomaterials capable of splenic retention and immunomodulation for the treatment of disease in a distant organ, the postinfarct heart. Engineered senescent erythrocyte-derived nanotheranostic (eSENTs) are generated, demonstrating significant uptake by the immune cells of the spleen including T and B cells, as well as monocytes and macrophages. When loaded with suberoylanilide hydroxamic acid (SAHA), the nanoagents exhibit a potent therapeutic effect, reducing infarct size by 14% at 72 h postmyocardial infarction when given as a single intravenous dose 2 h after injury. These results are supportive of the hypothesis that RBC-derived biomimicry may provide new approaches for the targeted modulation of the pathological processes involved in myocardial infarction, thus further experiments to decisively confirm the mechanisms of action are currently underway. This novel concept may have far-reaching applicability for the treatment of a number of both acute and chronic conditions where the immune responses are either stimulated or suppressed by the splenic (auto)immune milieu.


Assuntos
Biomimética , Infarto do Miocárdio , Humanos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Coração , Imunidade Inata , Imunomodulação
8.
EuroIntervention ; 20(10): e669-e680, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38776143

RESUMO

BACKGROUND: Compared with thin-strut durable-polymer drug-eluting stents (DP-DES), ultrathin-strut biodegradable-polymer sirolimus-eluting stents (BP-SES) improve stent-related clinical outcomes in patients undergoing percutaneous coronary intervention (PCI). Reduced stent strut thickness is hypothesised to underlie these benefits, but this conjecture remains unproven. AIMS: We aimed to assess the impact of strut thickness on stent healing and clinical outcomes between ultrathin-strut and thin-strut BP-SES. METHODS: First, we performed a preclinical study of 8 rabbits implanted with non-overlapping thin-strut (diameter/thickness 3.5 mm/80 µm) and ultrathin-strut (diameter/thickness 3.0 mm/60 µm) BP-SES in the infrarenal aorta. On day 7, the rabbits underwent intravascular near-infrared fluorescence optical coherence tomography (NIRF-OCT) molecular-structural imaging of fibrin deposition and stent tissue coverage, followed by histopathological analysis. Second, we conducted an individual data pooled analysis of patients enrolled in the BIOSCIENCE and BIOSTEMI randomised PCI trials treated with ultrathin-strut (n=282) or thin-strut (n=222) BP-SES. The primary endpoint was target lesion failure (TLF) at 1-year follow-up, with a landmark analysis at 30 days. RESULTS: NIRF-OCT image analyses revealed that ultrathin-strut and thin-strut BP-SES exhibited similar stent fibrin deposition (p=0.49) and percentage of uncovered stent struts (p=0.63). Histopathological assessments corroÂborated these findings. In 504 pooled randomised trial patients, TLF rates were similar for those treated with ultrathin-strut or thin-strut BP-SES at 30-day (2.5% vs 1.8%; p=0.62) and 1-year follow-up (4.3% vs 4.7%; p=0.88). CONCLUSIONS: Ultrathin-strut and thin-strut BP-SES demonstrate similar early arterial healing profiles and 30-day and 1-year clinical outcomes.


Assuntos
Stents Farmacológicos , Intervenção Coronária Percutânea , Sirolimo , Tomografia de Coerência Óptica , Animais , Coelhos , Intervenção Coronária Percutânea/instrumentação , Intervenção Coronária Percutânea/métodos , Humanos , Sirolimo/uso terapêutico , Sirolimo/administração & dosagem , Sirolimo/farmacologia , Resultado do Tratamento , Desenho de Prótese , Doença da Artéria Coronariana/terapia , Doença da Artéria Coronariana/diagnóstico por imagem , Masculino , Implantes Absorvíveis , Feminino , Cicatrização
9.
Arterioscler Thromb Vasc Biol ; 32(11): 2616-24, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22995524

RESUMO

OBJECTIVE: Assessment of thrombus inflammation in vivo could provide new insights into deep vein thrombosis (DVT) resolution. Here, we develop and evaluate 2 integrated fluorescence molecular-structural imaging strategies to quantify DVT-related inflammation and architecture and to assess the effect of thrombus inflammation on subsequent DVT resolution in vivo. METHODS AND RESULTS: Murine DVT were created with topical 5% FeCl(3) application to thigh or jugular veins (n=35). On day 3, mice received macrophage and matrix metalloproteinase activity fluorescence imaging agents. On day 4, integrated assessment of DVT inflammation and architecture was performed using confocal fluorescence intravital microscopy. Day 4 analyses showed robust relationships among in vivo thrombus macrophages, matrix metalloproteinase activity, and fluorescein isothiocyanate-dextran deposition (r>0.70; P<0.01). In a serial 2-time point study, mice with DVT underwent intravital microscopy at day 4 and day 6. Analyses revealed that the intensity of thrombus inflammation at day 4 predicted the magnitude of DVT resolution at day 6 (P<0.05). In a second approach, noninvasive fluorescence molecular tomography-computed tomography was used and detected macrophages within jugular DVT (P<0.05 versus sham controls). CONCLUSIONS: Integrated fluorescence molecular-structural imaging demonstrates that the DVT-induced inflammatory response can be readily assessed in vivo and can inform the magnitude of thrombus resolution.


Assuntos
Inflamação/patologia , Microscopia Confocal , Microscopia de Fluorescência , Imagem Molecular/métodos , Trombose Venosa/patologia , Animais , Biomarcadores/metabolismo , Cloretos , Dextranos , Modelos Animais de Doenças , Veia Femoral/imunologia , Veia Femoral/metabolismo , Veia Femoral/patologia , Compostos Férricos , Fluoresceína-5-Isotiocianato/análogos & derivados , Corantes Fluorescentes , Inflamação/induzido quimicamente , Inflamação/diagnóstico por imagem , Inflamação/imunologia , Inflamação/metabolismo , Veias Jugulares/imunologia , Veias Jugulares/metabolismo , Veias Jugulares/patologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Flebografia , Prognóstico , Reprodutibilidade dos Testes , Veia Safena/imunologia , Veia Safena/metabolismo , Veia Safena/patologia , Índice de Gravidade de Doença , Fatores de Tempo , Tomografia Computadorizada por Raios X , Trombose Venosa/induzido quimicamente , Trombose Venosa/diagnóstico por imagem , Trombose Venosa/imunologia , Trombose Venosa/metabolismo
10.
Nano Lett ; 12(1): 203-8, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-22196766

RESUMO

Here we report a proof-of-concept for development of pancreatic islet-targeting nanoparticles for immunomodulatory therapy of autoimmune type 1 diabetes. Modified with a unique islet-homing peptide, these polymeric nanomaterials exhibit 3-fold greater binding to islet endothelial cells and a 200-fold greater anti-inflammatory effect through targeted islet endothelial cell delivery of an immunosuppressant drug. Our findings also underscore the need to carefully tailor drug loading and nanoparticle dosage to achieve maximal vascular targeting and immunosuppression.


Assuntos
Imunossupressores/administração & dosagem , Imunossupressores/farmacocinética , Imunoterapia/métodos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Nanocápsulas/química , Polímeros/química , Animais , Células Cultivadas , Camundongos
11.
J Org Chem ; 77(15): 6480-94, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22734444

RESUMO

The rational syntheses of meso-tetraaryl-3-oxo-2-oxaporphyrins 5, known as porpholactones, via MnO(4)(-)-mediated oxidations of the corresponding meso-tetraaryl-2,3-dihydroxychlorins (7) is detailed. Since chlorin 7 is prepared from the parent porphyrin 1, this amounts to a 2-step replacement of a pyrrole moiety in 1 by an oxazolone moiety. The stepwise reduction of the porpholactone 5 results in the formation of chlorin analogues, meso-tetraaryl-3-hydroxy-2-oxachlorin (11) and meso-tetraaryl-2-oxachlorins (12). The reactivity of 11 with respect to nucleophilic substitution by O-, N-, and S-nucleophiles is described. The profound photophysical consequences of the formal replacement of a pyrrole with an oxazolone (porphyrin-like chromophore) or (substituted) oxazole moiety (chlorin-like chromophore with, for the parent oxazolochlorin 12, red-shifted Q(x) band with enhanced oscillator strengths) are detailed and rationalized on the basis of SAC-CI and MNDO-PSDCI molecular orbital theory calculations. The single crystal X-ray structures of the porpholactones point at a minor steric interaction between the carbonyl oxygen and the flanking phenyl group. The essentially planar structures of all chromophores in all oxidation states prove that the observed optical properties originate from the intrinsic electronic properties of the chromophores and are not subject to conformational modulation.


Assuntos
Lactonas/síntese química , Cristalografia por Raios X , Lactonas/química , Modelos Moleculares , Estrutura Molecular , Oxirredução
12.
Adv Healthc Mater ; 11(5): e2101387, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34879180

RESUMO

Polymeric nanocarriers (PNCs) can be used to deliver therapeutic microRNAs (miRNAs) to solid cancers. However, the ability of these nanocarriers to specifically target tumors remains a challenge. Alternatively, extracellular vesicles (EVs) derived from tumor cells show homotypic affinity to parent cells, but loading sufficient amounts of miRNAs into EVs is difficult. Here, it is investigated whether uPAR-targeted delivery of nanococktails containing PNCs loaded with therapeutic antimiRNAs, and coated with uPA engineered extracellular vesicles (uPA-eEVs) can elicit synergistic antitumor responses. The uPA-eEVs coating on PNCs increases natural tumor targeting affinities, thereby enhancing the antitumor activity of antimiRNA nanococktails. The systemic administration of uPA-eEV-PNCs nanococktail shows a robust tumor tropism, which significantly enhances the combinational antitumor effects of antimiRNA-21 and antimiRNA-10b, and leads to significant tumor regression and extension of progression free survival for syngeneic 4T1 tumor-bearing mice. In addition, the uPA-eEV-PNCs-antimiRNAs nanococktail plus low dose doxorubicin results in a synergistic antitumor effect as evidenced by inhibition of tumor growth, reduction of lung metastases, and extension of survival of 4T1 tumor-bearing mice. The targeted combinational nanococktail strategy could be readily translated to the clinical setting by using autologous cancer cells that have flexibility for ex vivo expansion and genetic engineering.


Assuntos
Vesículas Extracelulares , MicroRNAs , Neoplasias de Mama Triplo Negativas , Animais , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Humanos , Camundongos , MicroRNAs/genética , Peptídeos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
13.
J Am Chem Soc ; 133(22): 8740-52, 2011 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-21534626

RESUMO

The synthesis and chiral resolution of free-base and Ni(II) complexes of a number of derivatives of meso-tetraphenylmorpholinochlorins, with and without direct ß-carbon-to-o-phenyl linkages to the flanking phenyl groups, is described. The morpholinochlorins, a class of stable chlorin analogues, were synthesized in two to three steps from meso-tetraphenylporphyrin. The conformations and the relative stereostructures of a variety of free-base and Ni(II) complexes of these morpholinochlorins were elucidated by X-ray diffractometry. Steric and stereoelectronic arguments explain the relative stereoarray of the morpholino-substituents, which differ in the free-base and Ni(II) complexes, and in the monoalkoxy, ß-carbon-to-o-phenyl linked morpholinochlorins, and the dialkoxy derivatives. The Ni(II) complexes were all found to be severely ruffled whereas the free-base chromophores are more planar. As a result of the helimeric distortion of their porphyrinoid chromophores, the ruffled macrocycles possess a stable inherent element of chirality. Most significantly, resolution of the racemic mixtures was achieved, both by classical methods via diastereomers and by HPLC on a chiral phase. Full CD spectra were recorded and modeled using quantum-chemical computational methods, permitting, for the first time, an assignment of the absolute configurations of the chromophores. The report expands the range of known pyrrole-modified porphyrins. Beyond this, it introduces large chiral porphyrinoid π-systems that exist in the form of two enantiomeric, stereochemically stable helimers that can be resolved. This forms the basis for possible future applications, for example, in molecular-recognition systems or in materials with chiroptic properties.

14.
Arterioscler Thromb Vasc Biol ; 30(10): 1933-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20689078

RESUMO

OBJECTIVE: To investigate the effects of pioglitazone (PIO), a peroxisome proliferator-activated receptor γ agonist, on plaque matrix metalloproteinase (MMP) and macrophage (Mac) responses in vivo in a molecular imaging study. METHODS AND RESULTS: In vitro, PIO suppressed MMP-9 protein expression in murine peritoneal Macs (P<0.05). To assess PIO's effects on plaque inflammation, nondiabetic apolipoprotein E(-/-) mice receiving a high-cholesterol diet (HCD) were administered an MMP-activatable fluorescence imaging agent and a spectrally distinct Mac-avid fluorescent nanoparticle. After 24 hours, mice underwent survival dual-target intravital fluorescence microscopy of carotid arterial plaques. These mice were then randomized to HCD or HCD plus 0.012% PIO for 8 weeks, followed by a second intravital fluorescence microscopy study of the same carotid plaque. In the HCD group, in vivo MMP and Mac target-to-background ratios increased similarly (P<0.01 versus baseline). In contrast, PIO reduced MMP and Mac target-to-background ratios (P<0.01) versus HCD. Changes in MMP and Mac signals correlated strongly (r ≥0.75). Microscopy demonstrated MMP and Mac reductions in PIO-treated mice and a PIO-modulated increase in plaque collagen. CONCLUSIONS: Serial optical molecular imaging demonstrates that plaque MMP and Mac activity in vivo intensify with hypercholesterolemia and are reduced by PIO therapy.


Assuntos
Doenças das Artérias Carótidas/tratamento farmacológico , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Doenças das Artérias Carótidas/patologia , Doenças das Artérias Carótidas/fisiopatologia , Colesterol na Dieta/administração & dosagem , Colágeno/metabolismo , Feminino , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/fisiopatologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Metaloproteinase 9 da Matriz , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Pioglitazona
15.
Drug Discov Today ; 26(5): 1200-1211, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33561512

RESUMO

Cardiovascular disease (CVD) and its sequelae have long been the leading causes of death and disability in the developed world. Although mortality associated with CVD has been decreasing, due in large part to novel therapeutic options, the rate of decrease has flattened. Thus, there is a great need to investigate alternate therapeutic strategies that can increase efficacy while decreasing adverse effects. Nanomaterials have been widely investigated and have emerged as promising tools for both therapeutic and diagnostic purposes in oncology; however, the potential of nanomaterials has not been extensively explored for cardiovascular medicine. In this review, we focus on recent developments in the field of nanomedicines targeted for CVDs, with a special emphasis on cell membrane-coated nanoparticles (NPs) and their applications.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Nanopartículas , Animais , Membrana Celular/química , Humanos , Nanomedicina/métodos , Nanoestruturas
16.
Drug Discov Today ; 26(4): 902-915, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33383213

RESUMO

Smart nanocarriers obtained from bacteria and viruses offer excellent biomimetic properties which has led to significant research into the creation of advanced biomimetic materials. Their versatile biomimicry has application as biosensors, biomedical scaffolds, immobilization, diagnostics, and targeted or personalized treatments. The inherent natural traits of biomimetic and bioinspired bacteria- and virus-derived nanovesicles show potential for their use in clinical vaccines and novel therapeutic drug delivery systems. The past few decades have seen significant progress in the bioengineering of bacteria and viruses to manipulate and enhance their therapeutic benefits. From a pharmaceutical perspective, biomimetics enable the safe integration of naturally occurring bacteria and virus particles to achieve high, stable rates of cellular transfection/infection and prolonged circulation times. In addition, biomimetic technologies can overcome safety concerns associated with live-attenuated and inactivated whole bacteria or viruses. In this review, we provide an update on the utilization of bacterial and viral particles as drug delivery systems, theranostic carriers, and vaccine/immunomodulation modalities.


Assuntos
Bioengenharia/tendências , Materiais Biomiméticos/farmacologia , Portadores de Fármacos/farmacologia , Descoberta de Drogas/tendências , Nanoestruturas/uso terapêutico , Fenômenos Fisiológicos Bacterianos , Biomimética , Sistemas de Liberação de Medicamentos/tendências , Humanos , Vacinas/farmacologia , Fenômenos Fisiológicos Virais
17.
ACS Sens ; 6(6): 2225-2232, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34056903

RESUMO

Platelets play a prominent role in multiple diseases, in particular arterial and venous thrombosis and also in atherosclerosis and cancer. To advance the in vivo study of the biological activity of this cell type from a basic experimental focus to a clinical focus, new translatable platelet-specific molecular imaging agents are required. Herein, we report the development of a near-infrared fluorescence probe based upon tirofiban, a clinically approved small-molecule glycoprotein IIb/IIIa inhibitor (GPIIb/IIIa). Through in vitro experiments with human platelets and in vivo ones in a murine model of deep-vein thrombosis, we demonstrate the avidity of the generated probe for activated platelets, with the added benefit of a short blood half-life, thereby enabling rapid in vivo visualization within the vasculature.


Assuntos
Plaquetas , Inibidores da Agregação Plaquetária , Animais , Humanos , Camundongos , Imagem Óptica , Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Tirofibana
18.
Small ; 6(18): 2041-9, 2010 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-20721949

RESUMO

The synthesis and utility of a multimodal theranostic nanoagent based upon magnetofluorescent nanoparticles for the treatment of inflammatory atherosclerosis is described. These particles are modified with near-infrared fluorophores and light-activated therapeutic moieties, which allow for the optical determination of agent localization and phototoxic activation at spectrally distinct wavelengths. The resulting agent is readily taken up by murine macrophages in vitro and is highly phototoxic, with an LD(50) of 430 pM. Intravenous administration results in the localization of the nanoagent within macrophage-rich atherosclerotic lesions that can be imaged by intravital fluorescence microscopy. Irradiation of the atheroma with 650 nm light activates the therapeutic component and results in eradication of inflammatory macrophages, which may induce lesion stabilization. Importantly, these agents display limited skin photosensitivity, are highly efficacious, and provide an integrated imaging and therapeutic nanoplatform for atherosclerosis.


Assuntos
Aterosclerose/terapia , Sistemas de Liberação de Medicamentos/métodos , Inflamação/terapia , Macrófagos/efeitos dos fármacos , Nanoestruturas/uso terapêutico , Fototerapia/métodos , Animais , Apolipoproteínas E/genética , Aterosclerose/complicações , Aterosclerose/patologia , Separação Celular/métodos , Células Cultivadas , Feminino , Inflamação/complicações , Inflamação/patologia , Luz , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Nanoestruturas/administração & dosagem , Nanoestruturas/efeitos da radiação
19.
Life Sci ; 260: 118482, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32971105

RESUMO

Cardiovascular disease (CVD) is the most common co-morbidity associated with COVID-19 and the fatality rate in COVID-19 patients with CVD is higher compared to other comorbidities, such as hypertension and diabetes. Preliminary data suggest that COVID-19 may also cause or worsen cardiac injury in infected patients through multiple mechanisms such as 'cytokine storm', endotheliosis, thrombosis, lymphocytopenia etc. Autopsies of COVID-19 patients reveal an infiltration of inflammatory mononuclear cells in the myocardium, confirming the role of the immune system in mediating cardiovascular damage in response to COVID-19 infection and also suggesting potential causal mechanisms for the development of new cardiac pathologies and/or exacerbation of underlying CVDs in infected patients. In this review, we discuss the potential underlying molecular mechanisms that drive COVID-19-mediated cardiac damage, as well as the short term and expected long-term cardiovascular ramifications of COVID-19 infection in patients.


Assuntos
Betacoronavirus/isolamento & purificação , Doenças Cardiovasculares/etiologia , Infecções por Coronavirus/complicações , Inflamação/etiologia , Pneumonia Viral/complicações , COVID-19 , Doenças Cardiovasculares/patologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , Humanos , Inflamação/patologia , Pandemias , Pneumonia Viral/transmissão , Pneumonia Viral/virologia , Prognóstico , SARS-CoV-2
20.
Int J Nanomedicine ; 15: 8437-8449, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33162754

RESUMO

BACKGROUND: Lipid polymer hybrid nanoparticles (LPHNPs) have been widely investigated in drug and gene delivery as well as in medical imaging. A knowledge of lipid-based surface engineering and its effects on how the physicochemical properties of LPHNPs affect the cell-nanoparticle interactions, and consequently how it influences the cytological response, is in high demand. METHODS: Herein, we have engineered antibiotic-loaded (doxycycline or vancomycin) LPHNPs with cationic and zwitterionic lipids and examined the effects on their physicochemical characteristics (size and charge), antibiotic entrapment efficiency, and the in vitro intracellular bacterial killing efficiency against Mycobacterium smegmatis or Staphylococcus aureus infected macrophages. RESULTS: The incorporation of cationic or zwitterionic lipids in the LPHNP formulation resulted in a size reduction in LPHNPs formulations and shifted the surface charge of bare NPs towards positive or neutral values. Also observed were influences on the drug incorporation efficiency and modulation of the drug release from the biodegradable polymeric core. The therapeutic efficacy of LPHNPs loaded with vancomycin was improved as its minimum inhibitory concentration (MIC) (2 µg/mL) versus free vancomycin (4 µg/mL). Importantly, our results show a direct relationship between the cationic surface nature of LPHNPs and its intracellular bacterial killing efficiency as the cationic doxycycline or vancomycin loaded LPHNPs reduced 4 or 3 log CFU respectively versus the untreated controls. CONCLUSION: In our study, modulation of surface charge in the nanomaterial formulation increased macrophage uptake and intracellular bacterial killing efficiency of LPHNPs loaded with antibiotics, suggesting alternate way for optimizing their use in biomedical applications.


Assuntos
Antibacterianos/farmacologia , Sistemas de Liberação de Medicamentos , Espaço Intracelular/microbiologia , Macrófagos/microbiologia , Nanopartículas/química , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Lipídeos/química , Macrófagos/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Mycobacterium smegmatis/efeitos dos fármacos , Tamanho da Partícula , Polímeros/química , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa