Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Ann Biomed Eng ; 52(5): 1270-1279, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38374519

RESUMO

Healthy aortic heart valves are essential to the regulation of unidirectional blood flow. Calcific aortic valve disease (CAVD) is an actively progressive disease that involves the disorganization of valve cells and accumulation of calcium deposits on the aortic valve leaflets. CAVD involves disruption of cell environment homeostasis that prior cell culture models have found difficult to portray and model. As it is still poorly understood how tissue stiffening associates with lesion formation, here, we implement a novel 3D culture platform to characterize the relationship between mechanical stress and tissue remodeling and analyze how the application of pro-osteogenic stimulation dysregulates the native ability of valve cells to organize its matrix. Through a temporal study of macroscopic remodeling, we determine that aortic valve interstitial neo-tissues undergo varying stiffness and mechanical stress, demonstrate greater myofibroblastic gene expression, and show greater remodeling activity in the outer surface of the neo-tissue in a banding pattern when cultured in osteogenic growth medium. In human aortic valve interstitial cells cultured in osteogenic growth medium, we observed an increase in stress but significant decreases in myofibroblastic gene expression with the addition of growth factors. In summary, we are able to see the interplay of biochemical and biomechanical stimuli in valvular remodeling by using our platform to model dynamic stiffening of valve interstitial neo-tissues under different biochemical conditions.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica/patologia , Calcinose , Humanos , Células Cultivadas , Estenose da Valva Aórtica/patologia , Osteogênese
2.
Cardiovasc Pathol ; 58: 107414, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35074515

RESUMO

BACKGROUND: Calcific aortic valve disease (CAVD), a major cause for surgical aortic valve replacement, currently lacks available pharmacological treatments. Cadherin-11 (Cad11), a promising therapeutic target, promotes aortic valve calcification in vivo, but direct Cad11 inhibition in clinical trials has been unsuccessful. Targeting of downstream Cad11 effectors instead may be clinically useful; however, the downstream effectors that mediate Cad11-induced aortic valve cellular pathogenesis have not been investigated. APPROACH AND RESULTS: Immunofluorescence of calcified human aortic valves revealed that GTP-Rac1 is highly upregulated in calcified leaflets and is 2.15 times more co-localized with Cad11 in calcified valves than GTP-RhoA. Using dominant negative mutants in porcine aortic valve interstitial cells (PAVICs), we show that Cad11 predominantly regulates Runx2 nuclear localization via Rac1. Rac1-GEF inhibition via NSC23766 effectively reduces calcification in ex vivo porcine aortic valve leaflets treated with osteogenic media by 2.8-fold and also prevents Cad11-induced cell migration, compaction, and calcification in PAVICs. GTP-Rac1 and Trio, a known Cad11 binding partner and Rac1-GEF, are significantly upregulated in Nfatc1Cre; R26-Cad11Tg/Tg (Cad11 OX) mice that conditionally overexpress Cad11 in the heart valves by 3.1-fold and 6.3-fold, respectively. Finally, we found that the Trio-specific Rac1-GEF inhibitor, ITX3, effectively prevents Cad11-induced calcification and Runx2 induction in osteogenic conditions. CONCLUSION: Here we show that Cad11 induces many cellular pathogenic processes via Rac1 and that Rac1 inhibition effectively prevents many Cad11-induced aortic disease phenotypes. These findings highlight the therapeutic potential of blocking Rac1-GEFs in CAVD.


Assuntos
Estenose da Valva Aórtica , Calcinose , Animais , Valva Aórtica/patologia , Estenose da Valva Aórtica/patologia , Caderinas , Calcinose/etiologia , Células Cultivadas , Camundongos , Suínos
3.
Biomaterials ; 269: 120669, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33482604

RESUMO

OBJECTIVE: Calcific aortic valve disease (CAVD) is an actively regulated degenerative disease process. Clinical lesions exhibit marked 3D complexity not represented in current in vitro systems. We here present a unique mechanically stressed 3D culture system that recapitulates valve interstitial cell (VIC) induced matrix calcification through myofibroblastic activation and osteoblastic differentiation. We test the hypothesis that valve endothelial (VEC) - interstitial collaborative interactions modulate the risk and complexity of calcific pathogenesis within mechanically stressed and pro-inflammatory environments. APPROACH AND RESULTS: Porcine aortic valve endothelial and interstitial cells (VEC and VIC) were seeded in a mechanically constrained collagen hydrogels alone or in co-culture configurations. Raised 3D VIC-filled lesions formed within 7 days when cultured in osteogenic media (OGM), and surprisingly exacerbated by endothelial coculture. We identified a spatially coordinated pro-endochondral vs. pro-osteogenic signaling program within the lesion. VEC underwent Endothelial-to-Mesenchymal Transformation (EndMT) and populated the lesion center. The spatial complexity of molecular and cellular signatures of this 3D in vitro CAVD system were consistent with human diseased aortic valve histology. SNAI1 was highly expressed in the VEC and subendothelial direct VIC corroborates with human CAVD lesions. Spatial distribution of Sox9 vs. Runx2 expression within the developed lesions (Sox9 peri-lesion vs. Runx2 predominantly within lesions) mirrored their expression in heavily calcified human aortic valves. Finally, we demonstrate the applicability of this platform for screening potential pharmacologic therapies through blocking the canonical NFκB pathway via BAY 11-7082. CONCLUSIONS: Our results establish that VEC actively induce VIC pathological remodeling and calcification via EndMT and paracrine signaling. This mechanically constrained culture platform enables the interrogation of accelerated cell-mediated matrix remodeling behavior underpinned by this cellular feedback circuit. The high fidelity of this complex 3D model system to human CAVD mechanisms supports its use to test mechanisms of intercellular communication in valves and their pharmacological control.


Assuntos
Estenose da Valva Aórtica , Calcinose , Animais , Valva Aórtica , Diferenciação Celular , Células Cultivadas , Humanos , Suínos
4.
PLoS One ; 15(4): e0228973, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32298267

RESUMO

Electrically conductive composite ultrafiltration membranes composed of carbon nanotubes have exhibited efficient fouling inhibition in wastewater treatment applications. In the current study, poly(vinyl-alcohol)-carbon nanotube membranes were applied to fed batch crossflow electroultrafiltration of dilute (0.1 g/L of each species) single and binary protein solutions of α-lactalbumin and hen egg-white lysozyme at pH 7.4, 4 mM ionic strength, and 1 psi. Electroultrafiltration using the poly(vinyl-alcohol)-carbon nanotube composite membranes yielded temporary enhancements in sieving for single protein filtration and in selectivity for binary protein separation compared to ultrafiltration using the unmodified PS-35 membranes. Assessment of membrane fouling based on permeate flux, zeta potential measurements, and scanning electron microscopy visualization of the conditioned membranes indicated significant resulting protein adsorption and aggregation which limited the duration of improvement during electroultrafiltration with an applied cathodic potential of -4.6 V (vs. Ag/AgCl). These results imply that appropriate optimization of electroultrafiltration using carbon nanotube-deposited polymeric membranes may provide substantial short-term improvements in binary protein separations.


Assuntos
Condutividade Elétrica , Proteínas de Membrana/análise , Membranas Artificiais , Nanotubos de Carbono/química , Cloreto de Polivinila/química , Ultrafiltração , Adsorção , Animais , Galinhas , Eletrodos , Concentração de Íons de Hidrogênio , Eletricidade Estática
5.
Cardiovasc Pathol ; 46: 107194, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31982687

RESUMO

BACKGROUND: Celecoxib, a selective cyclooxygenase-2 inhibitor, was recently associated with increased incidence of aortic stenosis and found to produce a valvular calcification risk in vitro. Several cyclooxygenase-2 independent celecoxib derivatives have been developed and identified as possible therapies for inflammatory diseases due to their cadherin-11 inhibitory functions. Potential cardiovascular toxicities associated with these cyclooxygenase-2 independent celecoxib derivatives have not yet been investigated. Furthermore, the mechanism by which celecoxib produces valvular toxicity is not known. METHODS AND RESULTS: Celecoxib treatment produces a 2.8-fold increase in calcification in ex vivo porcine aortic valve leaflets and a more than 2-fold increase in calcification in porcine aortic valve interstitial cells cultured in osteogenic media. Its cyclooxygenase-2 independent derivative, 2,5-dimethylcelecoxib, produces a similar 2.5-fold increase in calcification in ex vivo leaflets and a 13-fold increase in porcine aortic valve interstitial cells cultured in osteogenic media. We elucidate that this offtarget effect depends on the presence of either of the two media components: dexamethasone, a synthetic glucocorticoid used for osteogenic induction, or cortisol, a natural glucocorticoid present at basal levels in the fetal bovine serum. In the absence of glucocorticoids, these inhibitors effectively reduce calcification. By adding glucocorticoids or hydrocortisone to a serum substitute lacking endogenous glucocorticoids, we show that dimethylcelecoxib conditionally induces a 3.5-fold increase in aortic valve calcification and osteogenic expression. Treatment with the Mitogen-activated protein kinase kinase inhibitor, U0126, rescues the offtarget effect, suggesting that celecoxib and dimethylcelecoxib conditionally augment Mitogen-activated protein kinase kinase/extracellular-signal-regulated kinase activity in the presence of glucocorticoids. CONCLUSION: Here we identify glucocorticoids as a possible source of the increased valvular calcification risk associated with celecoxib and its cyclooxygenase-2 independent derivatives. In the absence of glucocorticoids, these inhibitors effectively reduce calcification. Furthermore, the offtarget effects are not due to the drug's intrinsic properties as dual cyclooxygenase-2 and cadherin-11 inhibitors. These findings inform future design and development of celecoxib derivatives for potential clinical therapy.


Assuntos
Valva Aórtica/efeitos dos fármacos , Calcinose/induzido quimicamente , Celecoxib/toxicidade , Inibidores de Ciclo-Oxigenase 2/toxicidade , Dexametasona/toxicidade , Glucocorticoides/toxicidade , Doenças das Valvas Cardíacas/induzido quimicamente , Hidrocortisona/toxicidade , Osteogênese/efeitos dos fármacos , Pirazóis/toxicidade , Sulfonamidas/toxicidade , Animais , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Caderinas/genética , Caderinas/metabolismo , Calcinose/genética , Calcinose/metabolismo , Calcinose/patologia , Celecoxib/análogos & derivados , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/metabolismo , Doenças das Valvas Cardíacas/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Sus scrofa , Técnicas de Cultura de Tecidos
6.
Arterioscler Thromb Vasc Biol ; 40(3): 638-655, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31893948

RESUMO

OBJECTIVE: Although often studied independently, little is known about how aortic valve endothelial cells and valve interstitial cells interact collaborate to maintain tissue homeostasis or drive valve calcific pathogenesis. Inflammatory signaling is a recognized initiator of valve calcification, but the cell-type-specific downstream mechanisms have not been elucidated. In this study, we test how inflammatory signaling via NFκB (nuclear factor κ-light-chain enhancer of activated B cells) activity coordinates unique and shared mechanisms of valve endothelial cells and valve interstitial cells differentiation during calcific progression. Approach and Results: Activated NFκB was present throughout the calcific aortic valve disease (CAVD) process in both endothelial and interstitial cell populations in an established mouse model of hypercholesterolemia-induced CAVD and in human CAVD. NFκB activity induces endothelial to mesenchymal transformation in 3-dimensional cultured aortic valve endothelial cells and subsequent osteogenic calcification of transformed cells. Similarly, 3-dimensional cultured valve interstitial cells calcified via NFκB-mediated osteogenic differentiation. NFκB-mediated endothelial to mesenchymal transformation was directly demonstrated in vivo during CAVD via genetic lineage tracking. Genetic deletion of NFκB in either whole valves or valve endothelium only was sufficient to prevent valve-specific molecular and cellular mechanisms of CAVD in vivo despite the persistence of a CAVD inducing environment. CONCLUSIONS: Our results identify NFκB signaling as an essential molecular regulator for both valve endothelial and interstitial participation in CAVD pathogenesis. Direct demonstration of valve endothelial cell endothelial to mesenchymal transformation transmigration in vivo during CAVD highlights a new cellular population for further investigation in CAVD morbidity. The efficacy of valve-specific NFκB modulation in inhibiting hypercholesterolemic CAVD suggests potential benefits of multicell type integrated investigation for biological therapeutic development and evaluation for CAVD.


Assuntos
Valva Aórtica/metabolismo , Calcinose/metabolismo , Diferenciação Celular , Células Endoteliais/metabolismo , Doenças das Valvas Cardíacas/metabolismo , NF-kappa B/metabolismo , Osteogênese , Animais , Valva Aórtica/patologia , Calcinose/etiologia , Calcinose/patologia , Células Cultivadas , Microambiente Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Doenças das Valvas Cardíacas/etiologia , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/patologia , Humanos , Hipercolesterolemia/complicações , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/genética , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
7.
Curr Opin Biomed Eng ; 5: 82-89, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30276357

RESUMO

Monocytes and macrophages are critical components of the myeloid niche of the innate immune system. In addition to traditional roles as phagocytes, this subsection of innate immunity has been implicated in its ability to regulate tissue homeostasis and inflammation across diverse physiological systems. Recent emergence of discriminatory features within the monocyte/macrophage niche within the last 5 years has helped to clarify specific function(s) of the subpopulations of these cells. It is becoming increasingly aware that these cells are likely implicated in valve development and disease. This review seeks to use current literature and opinions to show the diverse roles and potential contributions of this niche throughout valvulogenic processes, adult homeostatic function, valve disease mechanisms, and tissue engineering approaches.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA