Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Surgery ; 175(4): 1189-1197, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38092635

RESUMO

BACKGROUND: A novel hydrophobically modified chitosan (hm-chitosan) polymer has been previously shown to improve survival in a non-compressible intra-abdominal bleeding model in swine. We performed a 28-day survival study to evaluate the safety of the hm-chitosan polymer in swine. METHODS: Female Yorkshire swine (40-50 kg) were used. A mild, non-compressible, closed-cavity bleeding model was created with splenic transection. The hm-chitosan polymer was applied intra-abdominally through an umbilical nozzle in the same composition and dose previously shown to improve survival. Animals were monitored intraoperatively and followed 28 days postoperatively for survival, signs of pain, and end-organ function. Gross pathological and microscopic evaluations were performed at the conclusion of the experiment. RESULTS: A total of 10 animals were included (hm-chitosan = 8; control = 2). The 2 control animals survived through 28 days, and 7 of the 8 animals from the hm-chitosan group survived without any adverse events. One animal from the hm-chitosan group required early termination of the study for signs of pain, and superficial colonic ulcers were found on autopsy. Laboratory tests showed no signs of end-organ dysfunction after exposure to hm-chitosan after 28 days. On gross pathological examination, small (<0.5 cm) peritoneal nodules were noticed in the hm-chitosan group, which were consistent with giant-cell foreign body reaction in microscopy, presumably related to polymer remnants. Microscopically, no signs of systemic polymer embolization or thrombosis were noticed. CONCLUSION: Prolonged intraperitoneal exposure to the hm-chitosan polymer was tolerated without any adverse event in the majority of animals. In the single animal that required early termination, the material did not appear to be associated with end-organ dysfunction in swine. Superficial colonic ulcers that would require surgical repair were identified in 1 out of 8 animals exposed to hm-chitosan.


Assuntos
Quitosana , Feminino , Animais , Suínos , Quitosana/efeitos adversos , Insuficiência de Múltiplos Órgãos , Úlcera , Hemorragia/etiologia , Hemorragia/terapia , Biopolímeros , Dor
2.
Bioorg Med Chem Lett ; 98: 129546, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37944866

RESUMO

Epigenetic proteins containing YEATS domains (YD) are an emerging target class in drug discovery. Described herein are the discovery and characterization efforts associated with PFI-6, a new chemical probe for the YD of MLLT1 (ENL/YEATS1) and MLLT3 (AF9/YEATS3). For hit identification, fragment-like mimetics of endogenous YD ligands (crotonylated histone-containing proteins), were synthesized via parallel medicinal chemistry (PMC) and screened for MLLT1 binding. Subsequent SAR studies led to iterative MLLT1/3 binding and selectivity improvements, culminating in the discovery of PFI-6. PFI-6 demonstrates good affinity and selectivity for MLLT1/3 vs. other human YD proteins (YEATS2/4) and engages MLLT3 in cells. Small-molecule X-ray co-crystal structures of two molecules, including PFI-6, bound to the YD of MLLT1/3 are also described. PFI-6 may be a useful tool molecule to better understand the biological effects associated with modulation of MLLT1/3.


Assuntos
Histonas , Proteínas Nucleares , Humanos , Proteínas Nucleares/metabolismo , Histonas/metabolismo , Domínios Proteicos , Descoberta de Drogas , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo
3.
Chem Res Toxicol ; 36(6): 934-946, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37148271

RESUMO

We recently disclosed SAR studies on systemically acting, amide-based inhibitors of diacylglycerol acyltransferase 2 (DGAT2) that addressed metabolic liabilities with the liver-targeted DGAT2 inhibitor PF-06427878. Despite strategic placement of a nitrogen atom in the dialkoxyaromatic ring in PF-06427878 to evade oxidative O-dearylation, metabolic intrinsic clearance remained high due to extensive piperidine ring oxidation as exemplified with compound 1. Piperidine ring modifications through alternate N-linked heterocyclic ring/spacer combination led to azetidine 2 that demonstrated lower intrinsic clearance. However, 2 underwent a facile cytochrome P450 (CYP)-mediated α-carbon oxidation followed by azetidine ring scission, resulting in the formation of ketone (M2) and aldehyde (M6) as stable metabolites in NADPH-supplemented human liver microsomes. Inclusion of GSH or semicarbazide in microsomal incubations led to the formation of Cys-Gly-thiazolidine (M3), Cys-thiazolidine (M5), and semicarbazone (M7) conjugates, which were derived from reaction of the nucleophilic trapping agents with aldehyde M6. Metabolites M2 and M5 were biosynthesized from NADPH- and l-cysteine-fortified human liver microsomal incubations with 2, and proposed metabolite structures were verified using one- and two-dimensional NMR spectroscopy. Replacement of the azetidine substituent with a pyridine ring furnished 8, which mitigated the formation of the electrophilic aldehyde metabolite, and was a more potent DGAT2 inhibitor than 2. Further structural refinements in 8, specifically introducing amide bond substituents with greater metabolic stability, led to the discovery of PF-06865571 (ervogastat) that is currently in phase 2 clinical trials for the treatment of nonalcoholic steatohepatitis.


Assuntos
Azetidinas , Diacilglicerol O-Aciltransferase , Humanos , Diacilglicerol O-Aciltransferase/metabolismo , Tiazolidinas/metabolismo , NADP/metabolismo , Glutationa/metabolismo , Microssomos Hepáticos/metabolismo , Piperidinas/metabolismo , Azetidinas/farmacologia , Azetidinas/metabolismo , Amidas/metabolismo
4.
Surgery ; 172(1): 421-426, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35184891

RESUMO

BACKGROUND: In military combat settings, noncompressible closed cavity exsanguination is the leading cause of potentially survivable deaths, with no effective treatment available at point of injury. The aim of this study was to assess whether an expanding foam based on hydrophobically modified chitosan (hm-chitosan) may be used as a locally injectable hemostatic agent for the treatment of noncompressible bleeding in a swine model. METHODS: A closed-cavity, grade V hepato-portal injury was created in all animals resulting in massive noncoagulopathic, noncompressible bleeding. Animals received either fluid resuscitation alone (control, n = 8) or fluid resuscitation plus intraperitoneal hm-chitosan agent through an umbilical port (experimental, n = 18). The experiment was terminated at 180 minutes or death (defined as end-tidal CO2 <8mmHg or mean arterial pressure [MAP] <15mmHg), whichever came first. RESULTS: All animals had profound hypotension and experienced a near-arrest from hypovolemic shock (mean MAP = 24 mmHg at 10 minutes). Mean survival time was higher than 150 minutes in the experimental arm versus 27 minutes in the control arm (P < .001). Three-hour survival was 72% in the experimental group and 0% in the control group (P = .002). Hm-chitosan stabilized rising lactate, preventing acute lethal acidosis. MAP improved drastically after deployment of the hm-chitosan and was preserved at 60 mmHg throughout the 3 hours. Postmortem examination was performed in all animals and the hepatoportal injuries were anatomically similar. CONCLUSION: Intraperitoneal administration of hm-chitosan-based foam for massive, noncompressible abdominal bleeding improves survival in a lethal, closed-cavity swine model. Chronic safety and toxicity studies are required.


Assuntos
Quitosana , Hemostáticos , Animais , Modelos Animais de Doenças , Hidratação/efeitos adversos , Hemorragia/etiologia , Hemorragia/terapia , Técnicas Hemostáticas , Hemostáticos/uso terapêutico , Humanos , Suínos
5.
J Med Chem ; 63(19): 10879-10896, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-32809824

RESUMO

Preclinical and clinical data suggest that acetyl-CoA carboxylase (ACC) inhibitors have the potential to rebalance disordered lipid metabolism, leading to improvements in nonalcoholic steatohepatitis (NASH). Consistent with these observations, first-in-human clinical trials with our ACC inhibitor PF-05175157 led to robust reduction of de novo lipogenesis (DNL), albeit with concomitant reductions in platelet count, which were attributed to the inhibition of fatty acid synthesis within bone marrow. Herein, we describe the design, synthesis, and evaluation of carboxylic acid-based ACC inhibitors with organic anion transporting polypeptide (OATP) substrate properties, which facilitated selective distribution of the compounds at the therapeutic site of action (liver) relative to the periphery. These efforts led to the discovery of clinical candidate PF-05221304 (12), which selectively inhibits liver DNL in animals, while demonstrating considerable safety margins against platelet reduction in a nonhuman primate model.


Assuntos
Acetil-CoA Carboxilase/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Inibidores Enzimáticos/farmacologia , Fígado/efeitos dos fármacos , Acetil-CoA Carboxilase/metabolismo , Animais , Inibidores Enzimáticos/uso terapêutico , Humanos , Lipogênese , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Especificidade por Substrato
6.
J Trauma Acute Care Surg ; 88(1): e1-e21, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31626024

RESUMO

Uncontrolled exsanguination remains the leading cause of death for trauma patients, many of whom die in the pre-hospital setting. Without expedient intervention, trauma-associated hemorrhage induces a host of systemic responses and acute coagulopathy of trauma. For this reason, health care providers and prehospital personal face the challenge of swift and effective hemorrhage control. The utilization of adjuncts to facilitate hemostasis was first recorded in 1886. Commercially available products haves since expanded to include topical hemostats, surgical sealants, and adhesives. The ideal product balances efficacy, with safety practicality and cost-effectiveness. This review of hemostasis provides a guide for successful implementation and simultaneously highlights future opportunities.


Assuntos
Hemorragia/terapia , Técnicas Hemostáticas/normas , Hemostáticos/administração & dosagem , Ferimentos e Lesões/complicações , Administração Tópica , Hemorragia/etiologia , Técnicas Hemostáticas/efeitos adversos , Técnicas Hemostáticas/tendências , Hemostáticos/efeitos adversos , Humanos , Guias de Prática Clínica como Assunto
7.
PLoS One ; 14(1): e0210297, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30601866

RESUMO

BACKGROUND: Canine hemangiosarcoma (cHSA) is a highly metastatic mesenchymal cancer that disseminates by hematogenous and direct implantation routes. Therapies for cHSA are generally ineffective, in part due to advanced clinical disease stage at the time of diagnosis. The validation of conventional molecular methods for detecting novel biomarkers preferentially expressed by cHSA could lead to more timely diagnosis, earlier therapeutic interventions, and improved outcomes. In humans, prostate-specific membrane antigen (PSMA) is a transmembrane protein overexpressed by prostate carcinoma and tumor-associated endothelium of various solid cancer histologies. Importantly, the preferential overexpression of PSMA by certain cancers has been leveraged for the development of diagnostic molecular imaging reagents and targeted therapeutics. Recently, PSMA has been qualitatively demonstrated to be expressed in cHSA cell lines, however, quantitative PSMA expressions and the potential utility of PSMA transcript identification in biologic fluids to support the presence of microscopic cHSA burden has not been reported. Therefore, this study sought to characterize the differential quantitative expressions of PSMA between cHSA and non-malignant tissues, and to determine the potential diagnostic utility of PCR-generated PSMA amplicons as a surrogate of rare cHSA cells dwelling within peritoneal and pericardial cavities. METHODS: Quantitative gene and protein expressions for PSMA were compared between one normal endothelial and six cHSA cell lines by RT-PCR, western blot analysis, and fluorescent microscopy. Additionally, gene and protein expressions of PSMA in normal canine tissues were characterized. Graded expressions of PSMA were determined in spontaneously-arising cHSA tumor samples and the feasibility of qualitative PCR as a molecular diagnostic to detect PSMA transcripts in whole blood from healthy dogs and hemorrhagic effusions from cHSA-bearing dogs were evaluated. RESULTS: PSMA gene and protein expressions were elevated (up to 6-fold) in cHSA cells compared with non-malignant endothelium. By immunohistochemistry, protein expressions of PSMA were detectable in all cHSA tissue samples evaluated. As predicted by human protein atlas data, PSMA's expression was comparably identified at substantial levels in select normal canine tissues including kidney, liver, and intestine. In young healthy pet dogs, PSMA amplicons could not be identified in circulating whole blood yet were detectable in hemorrhagic effusions collected from pet dogs with confirmed cHSA or PSMA-expressing cancer. CONCLUSIONS: PSMA is quantitatively overexpressed in cHSA compared to normal endothelium, but its protein expression is not restricted to only cHSA tumor tissues, as specific visceral organs also substantively express PSMA. Optimized qualitative PCR methods failed to amplify PSMA amplicons sufficiently for visible detection in circulating whole blood derived from healthy young dogs, yet PSMA transcripts were readily identifiable in hemorrhagic effusions collected from pet dogs with histologically confirmed cHSA or PSMA-expressing cancer. While preliminary, findings derived from a limited cohort of normal and diseased pet dogs provocatively raise the potential value of PSMA amplicon detection as an ancillary molecular diagnostic test for supporting the presence of microscopic cHSA disease burden within hemorrhagic body cavity effusions.


Assuntos
Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Doenças do Cão/genética , Doenças do Cão/metabolismo , Glutamato Carboxipeptidase II/genética , Glutamato Carboxipeptidase II/metabolismo , Hemangiossarcoma/veterinária , Animais , Líquido Ascítico/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Cães , Hemangiossarcoma/genética , Hemangiossarcoma/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Técnicas de Diagnóstico Molecular/métodos , Derrame Pericárdico/genética , Derrame Pericárdico/metabolismo , Derrame Pericárdico/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
8.
J Med Chem ; 61(6): 2372-2383, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29466005

RESUMO

Optimization of the pharmacokinetic (PK) properties of a series of activators of adenosine monophosphate-activated protein kinase (AMPK) is described. Derivatives of the previously described 5-aryl-indole-3-carboxylic acid clinical candidate (1) were examined with the goal of reducing glucuronidation rate and minimizing renal excretion. Compounds 10 (PF-06679142) and 14 (PF-06685249) exhibited robust activation of AMPK in rat kidneys as well as desirable oral absorption, low plasma clearance, and negligible renal clearance in preclinical species. A correlation of in vivo renal clearance in rats with in vitro uptake by human and rat renal organic anion transporters (human OAT/rat Oat) was identified. Variation of polar functional groups was critical to mitigate active renal clearance mediated by the Oat3 transporter. Modification of either the 6-chloroindole core to a 4,6-difluoroindole or the 5-phenyl substituent to a substituted 5-(3-pyridyl) group provided improved metabolic stability while minimizing propensity for active transport by OAT3.


Assuntos
Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Ativadores de Enzimas/síntese química , Ativadores de Enzimas/farmacologia , Indóis/síntese química , Indóis/farmacologia , Animais , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacocinética , Humanos , Indóis/farmacocinética , Absorção Intestinal , Rim/efeitos dos fármacos , Rim/enzimologia , Masculino , Modelos Moleculares , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Ratos , Ratos Wistar , Relação Estrutura-Atividade
9.
J Surg Res ; 207: 45-52, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27979487

RESUMO

BACKGROUND: Currently, the standard of care for treating severe hemorrhage in a military setting is Combat Gauze (CG). Previous work has shown that hydrophobically modified chitosan (hm-C) has significant hemostatic capability relative to its native chitosan counterpart. This work aims to evaluate gauze coated in hm-C relative to CG as well as ChitoGauze (ChG) in a lethal in vivo hemorrhage model. METHODS: Twelve Yorkshire swine were randomized to receive either hm-C gauze (n = 4), ChG (n = 4), or CG (n = 4). A standard hemorrhage model was used in which animals underwent a splenectomy before a 6-mm punch arterial puncture of the femoral artery. Thirty seconds of free bleeding was allowed before dressings were applied and compressed for 3 min. Baseline mean arterial pressure was preserved via fluid resuscitation. Experiments were conducted for 3 h after which any surviving animal was euthanized. RESULTS: hm-C gauze was found to be at least equivalent to both CG and ChG in terms of overall survival (100% versus 75%), number of dressing used (6 versus 7), and duration of hemostasis (3 h versus 2.25 h). Total post-treatment blood loss was lower in the hm-C gauze treatment group (4.7 mL/kg) when compared to CG (13.4 mL/kg) and ChG (12.1 mL/kg) groups. CONCLUSIONS: hm-C gauze outperformed both CG and ChG in a lethal hemorrhage model but without statistical significance for key endpoints. Future comparison of hm-C gauze to CG and ChG will be performed on a hypothermic, coagulopathic model that should allow for outcome significance to be differentiated under small treatment groups.


Assuntos
Bandagens , Quitosana/administração & dosagem , Hemorragia/terapia , Técnicas Hemostáticas/instrumentação , Hemostáticos/administração & dosagem , Ferimentos e Lesões/complicações , Administração Tópica , Animais , Quitosana/química , Quitosana/uso terapêutico , Feminino , Hemorragia/etiologia , Hemostáticos/química , Hemostáticos/uso terapêutico , Interações Hidrofóbicas e Hidrofílicas , Distribuição Aleatória , Suínos , Resultado do Tratamento
10.
J Med Chem ; 59(17): 8068-81, 2016 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-27490827

RESUMO

Adenosine monophosphate-activated protein kinase (AMPK) is a protein kinase involved in maintaining energy homeostasis within cells. On the basis of human genetic association data, AMPK activators were pursued for the treatment of diabetic nephropathy. Identification of an indazole amide high throughput screening (HTS) hit followed by truncation to its minimal pharmacophore provided an indazole acid lead compound. Optimization of the core and aryl appendage improved oral absorption and culminated in the identification of indole acid, PF-06409577 (7). Compound 7 was advanced to first-in-human trials for the treatment of diabetic nephropathy.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Ativadores de Enzimas/química , Indóis/química , Administração Oral , Adsorção , Animais , Cristalografia por Raios X , Cães , Ativadores de Enzimas/síntese química , Ativadores de Enzimas/farmacocinética , Ativadores de Enzimas/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Indazóis/síntese química , Indazóis/química , Indazóis/farmacologia , Indóis/síntese química , Indóis/farmacocinética , Indóis/farmacologia , Injeções Intravenosas , Macaca fascicularis , Masculino , Modelos Moleculares , Conformação Proteica , Ratos
11.
J Surg Res ; 193(1): 316-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25016441

RESUMO

BACKGROUND: Trauma represents a significant public health burden, and hemorrhage alone is responsible for 40% of deaths within the first 24 h after injury. Noncompressible hemorrhage accounts for the majority of hemorrhage-related deaths. Thus, materials which can arrest bleeding rapidly are necessary for improved clinical outcomes. This preliminary study evaluated several self-expanding hydrophobically modified chitosan (HM-CS) foams to determine their efficacy on a noncompressible severe liver injury under resuscitation. METHODS: Six HM-CS foam formulations (HM-CS1, HM-CS2, HM-CS3, HM-CS4, HM-CS5, and HM-CS6) of different graft types and densities were synthesized, characterized, and packaged into spray canisters using dimethyl ether as the propellant. Expansion profiles of the foams were evaluated in bench testing. Foams were then evaluated in vitro, interaction with blood cells was determined via microscopy, and cytotoxicity was assessed via live-dead cell assay on MCF7 breast cancer cells. For in vivo evaluation, rats underwent a 14 ± 3% hepatectomy. The animals were treated with either: (1) an HM-CS foam formulation, (2) CS foam, and (3) no treatment (NT). All animals were resuscitated with lactated Ringer solution. Survival, total blood loss, mean arterial pressures (MAP), and resuscitation volume were recorded for 60 min. RESULTS: Microscopy showed blood cells immobilizing into colonies within tight groups of adjacent foam bubbles. HM-CS foam did not display any toxic effects in vitro on MCF7 cells over a 72 h period studied. Application of HM-CS foam after hepatectomy decreased total blood loss (29.3 ± 7.8 mL/kg in HM-CS5 group versus 90.9 ± 20.3 mL/kg in the control group; P <0.001) and improved survival from 0% in controls to 100% in the HM-CS5 group (P <0.001). CONCLUSIONS: In this model of severe liver injury, spraying HM-CS foams directly on the injured liver surface decreased blood loss and increased survival. HM-CS formulations with the highest levels of hydrophobic modification (HM-CS4 and HM-CS5) resulted in the lowest total blood loss and highest survival rates. This pilot study suggests HM-CS foam may be useful as a hemostatic adjunct or solitary hemostatic intervention.


Assuntos
Quitosana/farmacologia , Hemorragia/etiologia , Hemorragia/terapia , Hemostáticos/farmacologia , Teste de Materiais , Ferimentos e Lesões/complicações , Animais , Bandagens , Bovinos , Quitosana/química , Modelos Animais de Doenças , Hemostáticos/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Fígado/irrigação sanguínea , Fígado/lesões , Células MCF-7 , Masculino , Projetos Piloto , Ratos Sprague-Dawley , Ressuscitação
12.
Biomater Sci ; 2(7): 1016-1023, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-32481975

RESUMO

Polymer hydrogels have long been used to hold and culture biological cells within their three-dimensional (3-D) matrices. Typically, in such cases, the cells are passively entrapped in a mesh of polymer chains. Here, we demonstrate an alternate approach where cells serve as active structural elements (crosslinks) within a polymer gel network. The polymers used in this context are hydrophobically modified (hm) derivatives of common biopolymers such as chitosan and alginate. We show that hm-polymers rapidly transform a liquid suspension of cells into an elastic gel. In contrast, the native biopolymers (without hydrophobes) do not cause such gelation. Gelation occurs because the hydrophobes on the polymer get embedded within the hydrophobic interiors of cell bilayer membranes. The polymer chains thus connect the cells into a 3-D sample-spanning network, with the cells serving as the junctions in the network. We demonstrate that a variety of cell types, including blood cells, endothelial cells, and breast cancer cells can be gelled by this approach. Cells gelled by hm-alginate are shown to remain viable within the network. Also, since the crosslinking mechanism is based on hydrophobic interactions, we show that the addition of supramolecules with hydrophobic binding pockets can reverse the gelation and release the cells. Cell-gels can be employed as injectable biomaterials since the connections in the network are susceptible to shear, but recover rapidly once shear is stopped. The overall approach provides a simple route towards the directed assembly of cell clusters and potentially to living tissue.

13.
Biotechnol Bioeng ; 110(2): 552-62, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22903626

RESUMO

We demonstrate that "nanofactory"-loaded biopolymer capsules placed in the midst of a bacterial population can direct bacterial communication. Quorum sensing (QS) is a process by which bacteria communicate through small-molecules, such as autoinducer-2 (AI-2), leading to collective behaviors such as virulence and biofilm formation. In our approach, a "nanofactory" construct is created, which comprises an antibody complexed with a fusion protein that produces AI-2. These nanofactories are entrapped within capsules formed by electrostatic complexation of cationic (chitosan) and anionic (sodium alginate) biopolymers. The chitosan capsule shell is crosslinked by tripolyphosphate (TPP) to confer structural integrity. The capsule shell is impermeable to the encapsulated nanofactories, but freely permeable to small molecules. In turn, the capsules are able to take in substrates from the external medium via diffusion, and convert these via the nanofactories into AI-2, which then diffuses out. The exported AI-2 is shown to stimulate QS responses in vicinal Escherichia coli. Directing bacterial population behavior has potential applications in next-generation antimicrobial therapy and pathogen detection. We also envision such capsules to be akin to artificial "cells" that can participate in native biological signaling and communicate in real-time with the human microbiome. Through such interaction capabilities, these "cells" may sense the health of the microbiome, and direct its function in a desired, host-friendly manner.


Assuntos
Bioengenharia/métodos , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Nanocápsulas , Nanotecnologia/métodos , Percepção de Quorum/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Alginatos/química , Animais , Anticorpos , Bovinos , Quitosana/química , Ácido Glucurônico/química , Proteínas de Fluorescência Verde/química , Ácidos Hexurônicos/química , Homosserina/análogos & derivados , Homosserina/química , Homosserina/farmacologia , Lactonas/química , Lactonas/farmacologia , Polifosfatos , Proteínas Recombinantes de Fusão/química , S-Adenosil-Homocisteína , Soroalbumina Bovina/química
14.
J Trauma Acute Care Surg ; 72(4): 899-907, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22491602

RESUMO

BACKGROUND: Chitosan is a functional biopolymer that has been widely used as a hemostat. Recently, its efficacy has been questioned due to clinical failures as a result of poor adhesiveness. The purpose of this study was to compare, in a severe groin injury model in swine, the hemostatic properties of an unmodified standard chitosan sponge with standard gauze dressing and a novel hydrophobically modified (hm) chitosan sponge. Previous studies have demonstrated that hm-chitosan provides greatly enhanced cellular adhesion and hemostatic effect via noncovalent insertion of hydrophobic pendant groups into cell membranes. METHODS: Twenty-four Yorkshire swine were randomized to receive hm-chitosan (n = 8), unmodified chitosan (n = 8), or standard Accu-Sorb gauze dressing (n = 8) for hemostatic control. A complex groin injury involving arterial puncture (4.4-mm punch) of the femoral artery was made after splenectomy. After 30 seconds of uncontrolled hemorrhage, the randomized dressing was applied and compression was held for 3 minutes. Fluid resuscitation was initiated to achieve and maintain the baseline mean arterial pressure and the wound was inspected for bleeding. Failure of hemostasis was defined as pooling of blood outside the wound. Animals were then monitored for 180 minutes and surviving animals were killed. RESULTS: Blood loss before treatment was similar between groups (p < 0.1). Compared with the hm-chitosan sponge group, which had no failures, the unmodified chitosan sponge group and the standard gauze group each had eight failures over the 180-minute observation period. For the unmodified chitosan sponge failures, six of which provided initial hemostasis, secondary rebleeding was observed 44 minutes ± 28 minutes after application. Standard gauze provided no initial hemostasis after the 3-minute compression interval. CONCLUSIONS: Hm-chitosan is superior to unmodified chitosan sponges (p < 0.001) or standard gauze for controlling bleeding from a lethal arterial injury. The hm-chitosan technology may provide an advantage over native chitosan-based dressings for control of active hemorrhage.


Assuntos
Artérias/lesões , Bandagens , Quitosana/uso terapêutico , Técnicas Hemostáticas/instrumentação , Tampões de Gaze Cirúrgicos , Adesividade , Animais , Modelos Animais de Doenças , Feminino , Artéria Femoral/lesões , Hemorragia/terapia , Suínos
15.
Bioconjug Chem ; 21(11): 1968-77, 2010 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-20964333

RESUMO

We demonstrate that multiphoton-absorption-induced luminescence (MAIL) is an effective means of monitoring the uptake of targeted nanoparticles into cells. Gold nanoparticles (AuNPs) with diameters of 4.5 and 16 nm were surface-functionalized with monocyclic RGDfK, an RGD peptide analogue that specifically targets the α(v)ß3 integrin, a membrane protein that is highly overexpressed in activated endothelial cells during tumor angiogenesis. To determine whether cyclic RGD can enhance the uptake of the functionalized AuNPs into activated endothelium, human umbilical vein endothelial cells (HUVECs) were used as a model system. MAIL imaging of HUVECs incubated with AuNPs demonstrates differential uptake of AuNPs functionalized with RGD analogues: RGDfK-modified nanoparticles are taken up by the HUVECs preferentially compared to AuNPs modified with linear RGD (GRGDSP) conjugates or with no surface conjugates. The luminescence counts observed for the AuNP-RGDfK conjugates are an order of magnitude greater than for AuNP-GRGDSP conjugates. Transmission electron microscopy shows that, once internalized, the AuNP-RGDfK conjugates remain primarily within endosomal and lysosomal vesicles in the cytoplasm of the cells. Significant aggregation of these particles was observed within the cells. MAIL imaging studies in the presence of specific uptake inhibitors indicate that AuNP-RGDfK conjugate uptake involves a specific binding event, with α(v)ß3 integrin-mediated endocytosis being an important uptake mechanism.


Assuntos
Células Endoteliais/metabolismo , Ouro/química , Ouro/metabolismo , Luminescência , Nanopartículas Metálicas/química , Imagem Molecular/métodos , Peptídeos Cíclicos/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Ouro/análise , Humanos , Nanopartículas Metálicas/análise , Tamanho da Partícula , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química , Soroalbumina Bovina/farmacologia , Azida Sódica/farmacologia , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA