Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Int J Mol Sci ; 23(15)2022 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-35955534

RESUMO

A key aspect of cytokine-induced changes as observed in sepsis is the dysregulated activation of endothelial cells (ECs), initiating a cascade of inflammatory signaling leading to leukocyte adhesion/migration and organ damage. The therapeutic targeting of ECs has been hampered by concerns regarding organ-specific EC heterogeneity and their response to inflammation. Using in vitro and in silico analysis, we present a comprehensive analysis of the proteomic changes in mouse lung, liver and kidney ECs following exposure to a clinically relevant cocktail of proinflammatory cytokines. Mouse lung, liver and kidney ECs were incubated with TNF-α/IL-1ß/IFN-γ for 4 or 24 h to model the cytokine-induced changes. Quantitative label-free global proteomics and bioinformatic analysis performed on the ECs provide a molecular framework for the EC response to inflammatory stimuli over time and organ-specific differences. Gene Ontology and PANTHER analysis suggest why some organs are more susceptible to inflammation early on, and show that, as inflammation progresses, some protein expression patterns become more uniform while additional organ-specific proteins are expressed. These findings provide an in-depth understanding of the molecular changes involved in the EC response to inflammation and can support the development of drugs targeting ECs within different organs. Data are available via ProteomeXchange (identifier PXD031804).


Assuntos
Células Endoteliais , Doenças Vasculares , Animais , Citocinas/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Inflamação/metabolismo , Camundongos , Proteômica , Fator de Necrose Tumoral alfa/metabolismo , Doenças Vasculares/metabolismo
2.
FASEB J ; 34(2): 2691-2702, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31908006

RESUMO

All drugs recently developed in rodent models to treat inflammatory disease have failed in clinical trials. We therefore used our novel biomimetic microfluidic assay (bMFA) to determine whether the response of murine cells to inflammatory activation or anti-inflammatory treatment is predictive of the response in human cells. Under physiologically relevant flow conditions, permeability and transendothelial electrical resistance (TEER) of human or mouse lung microvascular endothelial cells (HLMVEC or MLMVEC), and neutrophil-endothelial cell interaction was measured. The differential impact of a protein kinase C-delta TAT peptide inhibitor (PKCδ-i) was also quantified. Permeability of HLMVEC and MLMVEC was similar under control conditions but tumor necrosis factor α (TNF-α) and PKCδ-i had a significantly higher impact on permeability of HLMVEC. TEER across HLMVEC was significantly higher than MLMVEC, but PKCδ-i returned TEER to background levels only in human cells. The kinetics of N-formylmethionyl-leucyl-phenylalanine (fMLP)-mediated neutrophil migration was significantly different between the two species and PKCδ-i was significantly more effective in attenuating human neutrophil migration. However, human and mouse neutrophil adhesion patterns to microvascular endothelium were not significantly different. Surprisingly, while intercellular adhesion molecule 1 (ICAM-1) was significantly upregulated on activated HLMVEC, it was not significantly upregulated on activated MLMVEC. Responses to activation and anti-inflammatory treatment in mice may not always be predictive of their response in humans.


Assuntos
Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Endotélio Vascular/metabolismo , Neutrófilos/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Sepse/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
Int J Mol Sci ; 22(15)2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34360536

RESUMO

The endothelium is the inner layer of all blood vessels and it regulates hemostasis. It also plays an active role in the regulation of the systemic inflammatory response. Systemic inflammatory disease often results in alterations in vascular endothelium barrier function, increased permeability, excessive leukocyte trafficking, and reactive oxygen species production, leading to organ damage. Therapeutics targeting endothelium inflammation are urgently needed, but strong concerns regarding the level of phenotypic heterogeneity of microvascular endothelial cells between different organs and species have been expressed. Microvascular endothelial cell heterogeneity in different organs and organ-specific variations in endothelial cell structure and function are regulated by intrinsic signals that are differentially expressed across organs and species; a result of this is that neutrophil recruitment to discrete organs may be regulated differently. In this review, we will discuss the morphological and functional variations in differently originated microvascular endothelia and discuss how these variances affect systemic function in response to inflammation. We will review emerging in vivo and in vitro models and techniques, including microphysiological devices, proteomics, and RNA sequencing used to study the cellular and molecular heterogeneity of endothelia from different organs. A better understanding of microvascular endothelial cell heterogeneity will provide a roadmap for developing novel therapeutics to target the endothelium.


Assuntos
Anti-Inflamatórios/farmacologia , Endotélio Vascular/efeitos dos fármacos , Inflamação/tratamento farmacológico , Animais , Humanos
4.
Small ; 15(51): e1905375, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31762158

RESUMO

Targeting pharmaceuticals through the endothelial barrier is crucial for drug delivery. In this context, cavitation-assisted permeation shows promise for effective and reversible opening of intercellular junctions. A vessel-on-a-chip is exploited to investigate and quantify the effect of ultrasound-excited microbubbles-stable cavitation-on endothelial integrity. In the vessel-on-a-chip, the endothelial cells form a complete lumen under physiological shear stress, resulting in intercellular junctions that exhibit barrier functionality. Immunofluorescence microscopy is exploited to monitor vascular integrity following vascular endothelial cadherin staining. It is shown that microbubbles amplify the ultrasound effect, leading to the formation of interendothelial gaps that cause barrier permeabilization. The total gap area significantly increases with pressure amplitude compared to the control. Gap opening is fully reversible with gap area distribution returning to the control levels 45 min after insonication. The proposed integrated platform allows for precise and repeatable in vitro measurements of cavitation-enhanced endothelium permeability and shows potential for validating irradiation protocols for in vivo applications.


Assuntos
Células Endoteliais/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Microbolhas , Microscopia Confocal , Microscopia de Fluorescência
5.
FASEB J ; : fj201701099, 2018 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-29897816

RESUMO

In the event of a radiologic catastrophe, endothelial cell and neutrophil dysfunction play important roles in tissue injury. Clinically available therapeutics for radiation-induced vascular injury are largely supportive. PKCδ was identified as a critical regulator of the inflammatory response, and its inhibition was shown to protect critical organs during sepsis. We used a novel biomimetic microfluidic assay (bMFA) to interrogate the role of PKCδ in radiation-induced neutrophil-endothelial cell interaction and endothelial cell function. HUVECs formed a complete lumen in bMFA and were treated with 0.5, 2, or 5 Gy ionizing radiation (IR). At 24 h post-IR, the cells were treated with a PKCδ inhibitor for an additional 24 h. Under physiologic shear flow, the role of PKCδ on endothelium function and neutrophil adherence/migration was determined. PKCδ inhibition dramatically attenuated IR-induced endothelium permeability increase and significantly decreased neutrophil migration across IR-treated endothelial cells. Moreover, neutrophil adhesion to irradiated endothelial cells was significantly decreased after PKCδ inhibition in a flow-dependent manner. PKCδ inhibition downregulated IR-induced P-selectin, intercellular adhesion molecule 1, and VCAM-1 but not E-selectin overexpression. PKCδ is an important regulator of neutrophil-endothelial cell interaction post-IR, and its inhibition can serve as a potential radiation medical countermeasure.-Soroush, F., Tang, Y., Zaidi, H. M., Sheffield, J. B., Kilpatrick, L. E., Kiani, M. F. PKCδ inhibition as a novel medical countermeasure for radiation-induced vascular damage.

6.
Int J Mol Sci ; 20(6)2019 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-30917487

RESUMO

Protein Kinase C (PKC) is a family composed of phospholipid-dependent serine/threonine kinases that are master regulators of inflammatory signaling. The activity of different PKCs is context-sensitive and these kinases can be positive or negative regulators of signaling pathways. The delta isoform (PKCδ) is a critical regulator of the inflammatory response in cancer, diabetes, ischemic heart disease, and neurodegenerative diseases. Recent studies implicate PKCδ as an important regulator of the inflammatory response in sepsis. PKCδ, unlike other members of the PKC family, is unique in its regulation by tyrosine phosphorylation, activation mechanisms, and multiple subcellular targets. Inhibition of PKCδ may offer a unique therapeutic approach in sepsis by targeting neutrophil-endothelial cell interactions. In this review, we will describe the overall structure and function of PKCs, with a focus on the specific phosphorylation sites of PKCδ that determine its critical role in cell signaling in inflammatory diseases such as sepsis. Current genetic and pharmacological tools, as well as in vivo models, that are used to examine the role of PKCδ in inflammation and sepsis are presented and the current state of emerging tools such as microfluidic assays in these studies is described.


Assuntos
Proteína Quinase C-delta/metabolismo , Sepse/metabolismo , Transdução de Sinais , Regulação Alostérica , Animais , Humanos , Neutrófilos/metabolismo , Fosforilação , Proteína Quinase C-delta/química
7.
J Neuroinflammation ; 15(1): 309, 2018 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-30400800

RESUMO

BACKGROUND: Neuroinflammation often develops in sepsis leading to activation of cerebral endothelium, increased permeability of the blood-brain barrier (BBB), and neutrophil infiltration. We have identified protein kinase C-delta (PKCδ) as a critical regulator of the inflammatory response and demonstrated that pharmacologic inhibition of PKCδ by a peptide inhibitor (PKCδ-i) protected endothelial cells, decreased sepsis-mediated neutrophil influx into the lung, and prevented tissue damage. The objective of this study was to elucidate the regulation and relative contribution of PKCδ in the control of individual steps in neuroinflammation during sepsis. METHODS: The role of PKCδ in mediating human brain microvascular endothelial (HBMVEC) permeability, junctional protein expression, and leukocyte adhesion and migration was investigated in vitro using our novel BBB on-a-chip (B3C) microfluidic assay and in vivo in a rat model of sepsis induced by cecal ligation and puncture (CLP). HBMVEC were cultured under flow in the vascular channels of B3C. Confocal imaging and staining were used to confirm tight junction and lumen formation. Confluent HBMVEC were pretreated with TNF-α (10 U/ml) for 4 h in the absence or presence of PKCδ-i (5 µM) to quantify neutrophil adhesion and migration in the B3C. Permeability was measured using a 40-kDa fluorescent dextran in vitro and Evans blue dye in vivo. RESULTS: During sepsis, PKCδ is activated in the rat brain resulting in membrane translocation, a step that is attenuated by treatment with PKCδ-i. Similarly, TNF-α-mediated activation of PKCδ and its translocation in HBMVEC are attenuated by PKCδ-i in vitro. PKCδ inhibition significantly reduced TNF-α-mediated hyperpermeability and TEER decrease in vitro in activated HBMVEC and rat brain in vivo 24 h after CLP induced sepsis. TNF-α-treated HBMVEC showed interrupted tight junction expression, whereas continuous expression of tight junction protein was observed in non-treated or PKCδ-i-treated cells. PKCδ inhibition also reduced TNF-α-mediated neutrophil adhesion and migration across HBMVEC in B3C. Interestingly, while PKCδ inhibition decreased the number of adherent neutrophils to baseline (no-treatment group), it significantly reduced the number of migrated neutrophils below the baseline, suggesting a critical role of PKCδ in regulating neutrophil transmigration. CONCLUSIONS: The BBB on-a-chip (B3C) in vitro assay is suitable for the study of BBB function as well as screening of novel therapeutics in real-time. PKCδ activation is a key signaling event that alters the structural and functional integrity of BBB leading to vascular damage and inflammation-induced tissue damage. PKCδ-TAT peptide inhibitor has therapeutic potential for the prevention or reduction of cerebrovascular injury in sepsis-induced vascular damage.


Assuntos
Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiopatologia , Proteína Quinase C-delta/metabolismo , Sepse/patologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Humanos , Masculino , Infiltração de Neutrófilos/efeitos dos fármacos , Infiltração de Neutrófilos/fisiologia , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C-delta/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteína da Zônula de Oclusão-1/metabolismo
8.
Microvasc Res ; 99: 19-25, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25708050

RESUMO

Particle adhesion in vivo is highly dependent on the microvascular environment comprising of unique anatomical, geometrical, physiological fluid flow conditions and cell-particle and cell-cell interactions. Hence, proper design of vascular-targeted drug carriers that efficiently deliver therapeutics to the targeted cells or tissue at effective concentrations must account for these complex conditions observed in vivo. In this study, we build upon our previous results with the goal of characterizing the effects of bifurcations and their corresponding angle on adhesion of functionalized particles and neutrophils to activated endothelium. Our hypothesis is that adhesion is significantly affected by the type of biochemical interactions between particles and vessel wall as well as the presence of bifurcations and their corresponding angle. Here, we investigate adhesion of functionalized particles (2 µm and 7 µm microparticles) to protein coated channels as well as adhesion of human neutrophils to human endothelial cells under various physiological flow conditions in microfluidic bifurcating channels comprising of different contained angles (30°, 60°, 90°, or 120°). Our findings indicate that both functionalized particle and neutrophil adhesion propensity increase with a larger bifurcation angle. Moreover, the difference in the adhesion patterns of neutrophils and rigid, similar sized (7 µm) particles is more apparent in the junction regions with a larger contained angle. By selecting the right particle size range, enhanced targeted binding of vascular drug carriers can be achieved along with a higher efficacy at optimal drug dosage. Hence, vascular drug particle design needs to be tailored to account for higher binding propensity at larger bifurcation angles.


Assuntos
Vasos Sanguíneos/patologia , Adesão Celular , Microcirculação , Microvasos/fisiologia , Biotina/química , Portadores de Fármacos , Células Endoteliais/citologia , Humanos , Dispositivos Lab-On-A-Chip , Microesferas , Modelos Anatômicos , Neutrófilos/citologia , Tamanho da Partícula , Poliestirenos/química , Resistência ao Cisalhamento , Estresse Mecânico
9.
Anal Chem ; 86(16): 8344-51, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25135319

RESUMO

Current in vitro models of the leukocyte adhesion cascade cannot be used for real-time studies of the entire leukocyte adhesion cascade, including rolling, adhesion, and migration in a single assay. In this study, we have developed and validated a novel bioinspired microfluidic assay (bMFA) and used it to test the hypothesis that blocking of specific steps in the adhesion/migration cascade significantly affects other steps of the cascade. The bMFA consists of an endothelialized microvascular network in communication with a tissue compartment via a 3 µm porous barrier. Human neutrophils in bMFA preferentially adhered to activated human endothelial cells near bifurcations with rolling and adhesion patterns in close agreement with in vivo observations. Treating endothelial cells with monoclonal antibodies to E-selectin or ICAM-1 or treating neutrophils with wortmannin reduced rolling, adhesion, and migration of neutrophils to 60%, 20%, and 18% of their respective control values. Antibody blocking of specific steps in the adhesion/migration cascade (e.g., mAb to E-selectin) significantly downregulated other steps of the cascade (e.g., migration). This novel in vitro assay provides a realistic human cell based model for basic science studies, identification of new treatment targets, selection of pathways to target validation, and rapid screening of candidate agents.


Assuntos
Comunicação Celular , Endotélio Vascular/citologia , Leucócitos/citologia , Técnicas Analíticas Microfluídicas/métodos , Adulto , Adesão Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Migração e Rolagem de Leucócitos , Neutrófilos/citologia
10.
Analyst ; 139(12): 3039-47, 2014 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-24761431

RESUMO

Extracellular matrix (ECM) is a key component and regulator of many biological tissues including aorta. Several aortic pathologies are associated with significant changes in the composition of the matrix, especially in the content, quality and type of aortic structural proteins, collagen and elastin. The purpose of this study was to develop an infrared spectroscopic methodology that is comparable to biochemical assays to quantify collagen and elastin in aorta. Enzymatically degraded porcine aorta samples were used as a model of ECM degradation in abdominal aortic aneurysm (AAA). After enzymatic treatment, Fourier transform infrared (FTIR) spectra of the aortic tissue were acquired by an infrared fiber optic probe (IFOP) and FTIR imaging spectroscopy (FT-IRIS). Collagen and elastin content were quantified biochemically and partial least squares (PLS) models were developed to predict collagen and elastin content in aorta based on FTIR spectra. PLS models developed from FT-IRIS spectra were able to predict elastin and collagen content of the samples with strong correlations (RMSE of validation = 8.4% and 11.1% of the range respectively), and IFOP spectra were successfully used to predict elastin content (RMSE = 11.3% of the range). The PLS regression coefficients from the FT-IRIS models were used to map collagen and elastin in tissue sections of degraded porcine aortic tissue as well as a human AAA biopsy tissue, creating a similar map of each component compared to histology. These results support further application of FTIR spectroscopic techniques for evaluation of AAA tissues.


Assuntos
Aorta/metabolismo , Colágeno/análise , Elastina/análise , Matriz Extracelular/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Animais , Técnicas In Vitro , Suínos
11.
Nanomedicine ; 10(8): 1711-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24941463

RESUMO

Rebuilding of infarcted myocardium by mesenchymal stem cells (MSCs) has not been successful because of poor cell survival due in part to insufficient blood supply after myocardial infarction (MI). We hypothesize that targeted delivery of vascular endothelial growth factor (VEGF) to MI can help regenerate vasculature in support of MSC therapy in a rat model of MI. VEGF-encapsulated immunoliposomes targeting overexpressed P-selectin in MI tissue were infused by tail vein immediately after MI. One week later, MSCs were injected intramyocardially. The cardiac function loss was moderated slightly by targeted delivery of VEGF or MSC treatment. Targeted VEGF+MSC combination treatment showed highest attenuation in cardiac function loss. The combination treatment also increased blood vessel density (80%) and decreased collagen content in post-MI tissue (33%). Engraftment of MSCs in the combination treatment group was significantly increased and the engrafted cells contributed to the restoration of blood vessels. FROM THE CLINICAL EDITOR: VEGF immunoliposomes targeting myocardial infarction tissue resulted in significantly higher attenuation of cardiac function loss when used in combination with mesenchymal stem cells. MSCs were previously found to have poor ability to restore cardiac tissue, likely as a result of poor blood supply in the affected areas. This new method counterbalances that weakness by the known effects of VEGF, as demonstrated in a rat model.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/terapia , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Vasos Sanguíneos/efeitos dos fármacos , Colágeno/metabolismo , Modelos Animais de Doenças , Transplante de Células-Tronco Mesenquimais , Ratos
12.
Front Immunol ; 15: 1341752, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38524125

RESUMO

Purpose: Sepsis is a clinical syndrome defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis is a highly heterogeneous syndrome with distinct phenotypes that impact immune function and response to infection. To develop targeted therapeutics, immunophenotyping is needed to identify distinct functional phenotypes of immune cells. In this study, we utilized our Organ-on-Chip assay to categorize sepsis patients into distinct phenotypes using patient data, neutrophil functional analysis, and proteomics. Methods: Following informed consent, neutrophils and plasma were isolated from sepsis patients in the Temple University Hospital ICU (n=45) and healthy control donors (n=7). Human lung microvascular endothelial cells (HLMVEC) were cultured in the Organ-on-Chip and treated with buffer or cytomix ((TNF/IL-1ß/IFNγ). Neutrophil adhesion and migration across HLMVEC in the Organ-on-Chip were used to categorize functional neutrophil phenotypes. Quantitative label-free global proteomics was performed on neutrophils to identify differentially expressed proteins. Plasma levels of sepsis biomarkers and neutrophil extracellular traps (NETs) were determined by ELISA. Results: We identified three functional phenotypes in critically ill ICU sepsis patients based on ex vivo neutrophil adhesion and migration patterns. The phenotypes were classified as: Hyperimmune characterized by enhanced neutrophil adhesion and migration, Hypoimmune that was unresponsive to stimulation, and Hybrid with increased adhesion but blunted migration. These functional phenotypes were associated with distinct proteomic signatures and differentiated sepsis patients by important clinical parameters related to disease severity. The Hyperimmune group demonstrated higher oxygen requirements, increased mechanical ventilation, and longer ICU length of stay compared to the Hypoimmune and Hybrid groups. Patients with the Hyperimmune neutrophil phenotype had significantly increased circulating neutrophils and elevated plasma levels NETs. Conclusion: Neutrophils and NETs play a critical role in vascular barrier dysfunction in sepsis and elevated NETs may be a key biomarker identifying the Hyperimmune group. Our results establish significant associations between specific neutrophil functional phenotypes and disease severity and identify important functional parameters in sepsis pathophysiology that may provide a new approach to classify sepsis patients for specific therapeutic interventions.


Assuntos
Neutrófilos , Sepse , Humanos , Neutrófilos/metabolismo , Células Endoteliais , Proteômica , Biomarcadores/metabolismo , Fenótipo , Gravidade do Paciente
13.
Microvasc Res ; 89: 107-14, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23557880

RESUMO

OBJECTIVE: Leukocytes play a key role in the early response to tissue injury/infection resulting from physical, chemical or biological stimuli. This process involves the initiation of the leukocyte adhesion cascade mediated by a series of interactions between receptors and ligands on the endothelium and the leukocytes. Here, we characterize the adhesion profile of functionalized particles under physiological flow conditions in an idealized synthetic microvascular network (SMN) characterized by a bifurcation. We hypothesize that differences in the level of adhesion of functionalized particles in bifurcating SMNs are dependent on the ratio of adhesion molecules on the particles as well as geometric features of the in vitro networks. METHODS: Functionalized particles were prepared by coating their surfaces with different ratios of antibodies against ICAM-1 and E-selectin (aICAM-1:aE-selectin=100:0, 70:30, 50:50, 30:70, and 0:100). The adhesion of functionalized particles to 4h TNF-α activated human umbilical vein endothelial cells under shear flow (0.5, 2, and 4dyn/cm(2)) in bifurcating SMNs and in a parallel plate flow chamber was then quantified. RESULTS: The level of adhesion of 50:50 aICAM-1:aE-selectin particles was significantly higher compared to other particles in the bifurcating SMNs (~1.5-4 fold higher). However, in the parallel plate flow chamber 70:30 aICAM-1:aE-selectin particles exhibited a significantly higher level of adhesion (~1.5-2.5 fold higher). Furthermore, the adhesion of particles in junction regions was about 3-18 fold higher than that in straight sections of the SMNs. As expected, in straight sections of the SMNs and in the parallel plate flow chamber particle adhesion increased with decreasing shear. However, particle adhesion did not change significantly with decreasing shear at the junction regions of SMNs for all functionalized particles. CONCLUSION: Adhesion efficiency of functionalized particles is significantly affected by cell-adhesion molecule ratio density as well as geometric features of the vessels. Moreover, the differential adhesion patterns of particles between straight sections of bifurcating SMNs and parallel plate flow chamber, as well as straight sections and junction regions of bifurcating SMNs, indicates that adhesion profile of particles is highly dependent on the vascular geometry of the system used.


Assuntos
Endotélio Vascular/citologia , Microvasos , Anticorpos Monoclonais/química , Adesão Celular , Portadores de Fármacos , Selectina E/metabolismo , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/citologia , Ligantes , Microcirculação , Modelos Cardiovasculares , Poliestirenos/química , Resistência ao Cisalhamento , Estresse Mecânico
14.
Adv Exp Med Biol ; 765: 307-314, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22879049

RESUMO

Noninvasive injection of pro-angiogenic compounds such as vascular endothelial growth factor (VEGF) has shown promising results in regenerating cardiac microvasculature. However, these results have failed to translate into successful clinical trials in part due to the short half-life of VEGF in circulation. Increasing the dose of VEGF may increase its availability to the target tissue, but harmful side-effects remain a concern. Encapsulating and selectively targeting VEGF to the MI border zone may circumvent these problems. Anti-P-selectin conjugated immunoliposomes containing VEGF were developed to target the infarct border zone in a rat MI model. Targeted VEGF therapy significantly improves vascularization and cardiac function after an infarction.


Assuntos
Sistemas de Liberação de Medicamentos , Coração/efeitos dos fármacos , Lipossomos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Selectina-P/metabolismo , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Animais , Meia-Vida , Humanos , Masculino , Neovascularização Patológica/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
Shock ; 59(2): 224-231, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36377365

RESUMO

ABSTRACT: Sepsis is a major health issue and a leading cause of death in hospitals globally. The treatment of sepsis is largely supportive, and there are no therapeutics available that target the underlying pathophysiology of the disease. The development of therapeutics for the treatment of sepsis is hindered by the heterogeneous nature of the disease. The presence of multiple, distinct immune phenotypes ranging from hyperimmune to immunosuppressed can significantly impact the host response to infection. Recently, omics, biomarkers, cell surface protein expression, and immune cell profiles have been used to classify immune status of sepsis patients. However, there has been limited studies of immune cell function during sepsis and even fewer correlating omics and biomarker alterations to functional consequences. In this review, we will discuss how the heterogeneity of sepsis and associated immune cell phenotypes result from changes in the omic makeup of cells and its correlation with leukocyte dysfunction. We will also discuss how emerging techniques such as in silico modeling and machine learning can help in phenotyping sepsis patients leading to precision medicine.


Assuntos
Sepse , Humanos , Sepse/metabolismo , Leucócitos/metabolismo , Biomarcadores/metabolismo , Fenótipo , Simulação por Computador
16.
Front Cell Infect Microbiol ; 13: 1274842, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38259971

RESUMO

Sepsis is a global health concern accounting for more than 1 in 5 deaths worldwide. Sepsis is now defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis can develop from bacterial (gram negative or gram positive), fungal or viral (such as COVID) infections. However, therapeutics developed in animal models and traditional in vitro sepsis models have had little success in clinical trials, as these models have failed to fully replicate the underlying pathophysiology and heterogeneity of the disease. The current understanding is that the host response to sepsis is highly diverse among patients, and this heterogeneity impacts immune function and response to infection. Phenotyping immune function and classifying sepsis patients into specific endotypes is needed to develop a personalized treatment approach. Neutrophil-endothelium interactions play a critical role in sepsis progression, and increased neutrophil influx and endothelial barrier disruption have important roles in the early course of organ damage. Understanding the mechanism of neutrophil-endothelium interactions and how immune function impacts this interaction can help us better manage the disease and lead to the discovery of new diagnostic and prognosis tools for effective treatments. In this review, we will discuss the latest research exploring how in silico modeling of a synergistic combination of new organ-on-chip models incorporating human cells/tissue, omics analysis and clinical data from sepsis patients will allow us to identify relevant signaling pathways and characterize specific immune phenotypes in patients. Emerging technologies such as machine learning can then be leveraged to identify druggable therapeutic targets and relate them to immune phenotypes and underlying infectious agents. This synergistic approach can lead to the development of new therapeutics and the identification of FDA approved drugs that can be repurposed for the treatment of sepsis.


Assuntos
Neutrófilos , Sepse , Animais , Humanos , Comunicação Celular , Sepse/tratamento farmacológico , Simulação por Computador , Aprendizado de Máquina
17.
Biophys J ; 102(9): 2086-94, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22824272

RESUMO

Liposomal drugs are a useful alternative to conventional drugs and hold great promise for targeted delivery in the treatment of many diseases. Most of the liposomal drugs on the market or under clinical trials include cholesterol as a membrane stabilizing agent. Here, we used liposomal CA4P, an antivascular drug, to demonstrate that cholesterol content can actually modulate the release and cytotoxicity of liposomal drugs in a delicate and predictable manner. We found that both the rate of the CA4P release from the interior aqueous compartment of the liposomes to the bulk aqueous phase and the extent of the drug's cytotoxicity undergo a biphasic variation, as large as 50%, with liposomal cholesterol content at the theoretically predicted C(r), e.g., 22.0, 22.2, 25.0, 33.3, 40.0, and 50.0 mol % cholesterol for maximal superlattice formation. It appears that at C(r), CA4P can be released from the liposomes more readily than at non-C(r), probably due to the increased domain boundaries between superlattice and nonsuperlattice regions, which consequently results in increased cytotoxicity. The idea that the increased domain boundaries at C(r) would facilitate the escape of molecules from membranes was further supported by the data of dehydroergosterol transfer from liposomes to MßCD. These results together show that the functional importance of sterol superlattice formation in liposomes can be propagated to distal targeted cells and reveal a new, to our knowledge, mechanism for how sterol content and membrane lateral organization can control the release of entrapped or embedded molecules in membranes.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Preparações de Ação Retardada/química , Lipossomos/química , Estilbenos/administração & dosagem , Estilbenos/química , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/química , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular , Difusão , Composição de Medicamentos/métodos , Feminino , Humanos
18.
Vasc Biol ; 4(1): R15-R34, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35515704

RESUMO

During sepsis, defined as life-threatening organ dysfunction due to dysregulated host response to infection, systemic inflammation activates endothelial cells and initiates a multifaceted cascade of pro-inflammatory signaling events, resulting in increased permeability and excessive recruitment of leukocytes. Vascular endothelial cells share many common properties but have organ-specific phenotypes with unique structure and function. Thus, therapies directed against endothelial cell phenotypes are needed to address organ-specific endothelial cell dysfunction. Omics allow for the study of expressed genes, proteins and/or metabolites in biological systems and provide insight on temporal and spatial evolution of signals during normal and diseased conditions. Proteomics quantifies protein expression, identifies protein-protein interactions and can reveal mechanistic changes in endothelial cells that would not be possible to study via reductionist methods alone. In this review, we provide an overview of how sepsis pathophysiology impacts omics with a focus on proteomic analysis of mouse endothelial cells during sepsis/inflammation and its relationship with the more clinically relevant omics of human endothelial cells. We discuss how omics has been used to define septic endotype signatures in different populations with a focus on proteomic analysis in organ-specific microvascular endothelial cells during sepsis or septic-like inflammation. We believe that studies defining septic endotypes based on proteomic expression in endothelial cell phenotypes are urgently needed to complement omic profiling of whole blood and better define sepsis subphenotypes. Lastly, we provide a discussion of how in silico modeling can be used to leverage the large volume of omics data to map response pathways in sepsis.

19.
Microcirculation ; 18(5): 380-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21418388

RESUMO

OBJECTIVE: Particle adhesion in vivo is dependent on the microcirculation environment, which features unique anatomical (bifurcations, tortuosity, cross-sectional changes) and physiological (complex hemodynamics) characteristics. The mechanisms behind these complex phenomena are not well understood. In this study, we used a recently developed in vitro model of microvascular networks, called SMN, for characterizing particle adhesion patterns in the microcirculation. METHODS: SMNs were fabricated using soft-lithography processes followed by particle adhesion studies using avidin and biotin-conjugated microspheres. Particle adhesion patterns were subsequently analyzed using CFD-based modeling. RESULTS: Experimental and modeling studies highlighted the complex and heterogeneous fluid flow patterns encountered by particles in microvascular networks resulting in significantly higher propensity of adhesion (>1.5×) near bifurcations compared with the branches of the microvascular networks. CONCLUSION: Bifurcations are the focal points of particle adhesion in microvascular networks. Changing flow patterns and morphology near bifurcations are the primary factors controlling the preferential adhesion of functionalized particles in microvascular networks. SMNs provide an in vitro framework for understanding particle adhesion.


Assuntos
Microesferas , Microvasos/fisiologia , Modelos Cardiovasculares , Animais , Avidina/química , Biotina/química , Adesão Celular/fisiologia , Humanos , Microvasos/anatomia & histologia
20.
Microvasc Res ; 82(3): 210-20, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21763328

RESUMO

Cell-fluid and cell-cell interactions are critical components of many physiological and pathological conditions in the microvasculature. Similarly, particle-cell interactions play an important role in targeted delivery of therapeutics to tissue. Development of in vitro fluidic devices to mimic these microcirculatory processes has been a critical step forward in our understanding of the inflammatory process, developing of nano-particulate drug carriers, and developing realistic in vitro models of the microvasculature and its surrounding tissue. However, widely used parallel plate flow based devices and assays have a number of important limitations for studying the physiological conditions in vivo. In addition, these devices are resource hungry and time consuming for performing various assays. Recently developed, more realistic, microfluidic based devices have been able to overcome many of these limitations. In this review, an overview of the fluidic devices and their use in studying the effects of shear forces on cell-cell and cell-particle interactions is presented. In addition, use of mathematical models and computational fluid dynamics (CFD) based models for interpreting the complex flow patterns in the microvasculature is highlighted. Finally, the potential of 3D microfluidic devices and imaging for better representing in vivo conditions under which cell-cell and cell-particle interactions take place is discussed.


Assuntos
Adesão Celular , Comunicação Celular , Hemorreologia , Microcirculação , Técnicas Analíticas Microfluídicas/instrumentação , Microvasos/fisiologia , Animais , Simulação por Computador , Desenho de Equipamento , Humanos , Microvasos/citologia , Modelos Cardiovasculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA