Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(36): e2202577119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36037361

RESUMO

Calcific aortic valve disease (CAVD) is common in people over the age of 65. Progressive valvular calcification is a characteristic of CAVD and due to chronic inflammation in aortic valve interstitial cells (AVICs) resulting in CAVD progression. IL-38 is a naturally occurring anti-inflammatory cytokine; here, we report lower levels of endogenous IL-38 in AVICs isolated from patients' CAVD valves compared to AVICs from non-CAVD valves. Recombinant IL-38 suppressed spontaneous inflammatory activity and calcium deposition in cultured AVICs. In mice, knockdown of IL-38 enhanced the production of inflammatory mediators in murine AVICs exposed to the proinflammatory stimulant matrilin-2. We also observed that in cultured AVICs matrilin-2 stimulation activated the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome with procaspase-1 cleavage into active caspase-1. The addition of IL-38 to matrilin-2-treated AVICs suppressed caspase-1 activation and reduced the expression of intercellular adhesion molecule-1, vascular cell adhesion molecule-1, runt-related transcription factor 2, and alkaline phosphatase. Aged IL-38-deficient mice fed a high-fat diet exhibited aortic valve lesions compared to aged wild-type mice fed the same diet. The interleukin-1 receptor 9 (IL-1R9) is the putative receptor mediating the anti-inflammatory properties of IL-38; we observed that IL-1R9-deficient mice exhibited spontaneous aortic valve thickening and greater calcium deposition in AVICs compared to wild-type mice. These data demonstrate that IL-38 suppresses spontaneous and stimulated osteogenic activity in aortic valve via inhibition of the NLRP3 inflammasome and caspase-1. The findings of this study suggest that IL-38 has therapeutic potential for prevention of CAVD progression.


Assuntos
Estenose da Valva Aórtica , Calcinose , Interleucinas , Animais , Anti-Inflamatórios/farmacologia , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Estenose da Valva Aórtica/tratamento farmacológico , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/metabolismo , Calcinose/tratamento farmacológico , Cálcio/metabolismo , Caspases/metabolismo , Células Cultivadas , Humanos , Inflamassomos/metabolismo , Interleucina-1 , Interleucinas/genética , Interleucinas/metabolismo , Interleucinas/farmacologia , Proteínas Matrilinas/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Osteogênese , Receptores de Interleucina-9/genética , Proteínas Recombinantes/farmacologia
2.
J Surg Res ; 286: 118-126, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36822134

RESUMO

INTRODUCTION: Calcific aortic valve disease (CAVD) is a slowly progressive fibro-calcific valve leaflet disorder. The underlying pathophysiology is complex and not yet well understood. Complement is known to play a role in the pathogenesis of CAVD by upregulating Runx2 to induce profibrogenic change in human aortic valve interstitial cells (AVICs). Furthermore, H-K-ATPase has independently been shown to induce tissue calcification. Therefore, we hypothesized that complement cross talks with H-K-ATPase to upregulate Runx2 in human AVICs. MATERIALS AND METHODS: Human AVICs were isolated from normal and calcified aortic valves. Cells were treated with a variation of complement, H-K-ATPase, or ERK1/2 inhibitors. H-K-ATPase and its association with complement in AVICs were investigated by reverse transcriptase-polymerase chain reaction, immunofluorescence, and Western blot. RESULTS: Calcified human AVICs expressed significantly higher H-K-ATPase level than normal human AVICs. Presence of complement C3 with H-K-ATPase is found in AVICs after complement treatment. Complement induced both H-K-ATPase and Runx2 expression in AVICs, which was associated with increased phosphorylation of ERK1/2 and its downstream molecule p-70 S6. Pharmacological inhibition of either H-K-ATPase or Erk1/2 abolished complement-induced Runx2 expression. CONCLUSIONS: These findings indicate that complement cross talks with H-K-ATPase to upregulate Runx2 in human AVICs by activation of ERK1/2 signaling pathways. The study revealed the potential role of H-K-ATPase in the pathogenesis of CAVD and therapeutically targeting either complement system or H-K-ATPase may limit the development of CAVD.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica , Humanos , Estenose da Valva Aórtica/metabolismo , Transdução de Sinais , Adenosina Trifosfatases/metabolismo , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo
3.
Mol Med ; 28(1): 5, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35062861

RESUMO

BACKGROUND: Calcific aortic valve disease (CAVD) is the most prevalent heart valve disorder in the elderly. Valvular fibrocalcification is a characteristic pathological change. In diseased valves, monocyte accumulation is evident, and aortic valve interstitial cells (AVICs) display greater fibrogenic and osteogenic activities. However, the impact of activated monocytes on valular fibrocalcification remains unclear. We tested the hypothesis that pro-inflammatory mediators from activated monocytes elevate AVIC fibrogenic and osteogenic activities. METHODS AND RESULTS: Picro-sirius red staining and Alizarin red staining revealed collagen and calcium depositions in cultured human AVICs exposed to conditioned media derived from Pam3CSK4-stimulated monocytes (Pam3 CM). Pam3 CM up-regulated alkaline phosphatase (ALP), an osteogenic biomarker, and extracellular matrix proteins collagen I and matrix metalloproteinase-2 (MMP-2). ELISA analysis identified high levels of RANTES and TNF-α in Pam3 CM. Neutralizing RANTES in the Pam3 CM reduced its effect on collagen I and MMP-2 production in AVICs while neutralizing TNF-α attenuated the effect on AVIC ALP production. In addition, Pam3 CM induced NF-κB and JNK activation. While JNK mediated the effect of Pam3 CM on collagen I and MMP-2 production, NF-κB was critical for the effect of Pam3 CM on ALP production in AVICs. CONCLUSIONS: This study demonstrates that activated monocytes elevate the fibrogenic and osteogenic activities in human AVICs through a paracrine mechanism. TNF-α and RANTES mediate the pro-fibrogenic effect of activated monocytes on AVICs through activation of JNK, and TNF-α also activates NF-κB to elevate AVIC osteogenic activity. The results suggest that infiltrated monocytes elevate AVIC fibrocalcific activity to promote CAVD progression.


Assuntos
Estenose da Valva Aórtica/etiologia , Estenose da Valva Aórtica/metabolismo , Valva Aórtica/patologia , Calcinose/etiologia , Calcinose/metabolismo , Suscetibilidade a Doenças , Mediadores da Inflamação/metabolismo , Monócitos/metabolismo , Valva Aórtica/metabolismo , Biomarcadores , Células Cultivadas , Colágeno/metabolismo , Meios de Cultivo Condicionados , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos
4.
Inflamm Res ; 71(5-6): 681-694, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35411432

RESUMO

OBJECTIVE: Inflammatory infiltration in aortic valves promotes calcific aortic valve disease (CAVD) progression. While soluble extracellular matrix (ECM) proteins induce inflammatory responses in aortic valve interstitial cells (AVICs), the impact of monocytes on AVIC inflammatory responses is unknown. We tested the hypothesis that monocytes enhance AVIC inflammatory responses to soluble ECM protein in this study. METHODS: Human AVICs isolated from normal aortic valves were cocultured with monocytes and stimulated with soluble ECM protein (matrilin-2). ICAM-1 and IL-6 productions were assessed. YAP and NF-κB phosphorylation were analyzed. Recombinant CD18, neutralizing antibodies against ß2-integrin or ICAM-1, and inhibitor of YAP or NF-κB were applied. RESULTS: AVIC expression of ICAM-1 and IL-6 was markedly enhanced by the presence of monocytes, although matrilin-2 did not affect monocyte production of ICAM-1 or IL-6. Matrilin-2 up-regulated the expression of monocyte ß2-integrin and AVIC ICAM-1, leading to monocyte-AVIC adhesion. Neutralizing ß2-integrin or ICAM-1 in coculture suppressed monocyte adhesion to AVICs and the expression of ICAM-1 and IL-6. Recombinant CD18 enhanced the matrilin-2-induced ICAM-1 and IL-6 expression in AVIC monoculture. Further, stimulation of coculture with matrilin-2 induced greater YAP and NF-κB phosphorylation. Inhibiting either YAP or NF-κB markedly suppressed the inflammatory response to matrilin-2 in coculture. CONCLUSION: Monocyte ß2-integrin interacts with AVIC ICAM-1 to augment AVIC inflammatory responses to soluble matrilin-2 through enhancing the activation of YAP and NF-κB signaling pathways. Infiltrated monocytes may promote valvular inflammation through cell-cell interaction with AVICs to enhance their sensitivity to damage-associated molecular patterns.


Assuntos
Valva Aórtica , Monócitos , Valva Aórtica/metabolismo , Antígenos CD18/metabolismo , Células Cultivadas , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/metabolismo , Proteínas Matrilinas/metabolismo , Monócitos/metabolismo , NF-kappa B/metabolismo
5.
Dig Dis Sci ; 67(10): 4732-4741, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35001242

RESUMO

BACKGROUND: Observational studies demonstrate a protective effect of statins on the development and progression of esophageal adenocarcinoma. The role of statins in the prevention of reflux-induced esophageal changes remains unknown. AIMS: Using a mixed gastroduodenal reflux mouse model, we hypothesized that oral administration of simvastatin would attenuate reflux-induced mucosal changes of the distal esophagus. METHODS: Human Barrett's (CPB) and esophageal adenocarcinoma (FLO1 and OE19) cells were treated with simvastatin. Cell proliferation and apoptosis were evaluated using the MTS proliferation and annexin V apoptosis assays, respectively. A reflux mouse model was generated by performing a side-to-side anastomosis between the gastroesophageal junction and first portion of the duodenum (duodeno-gastroesophageal anastomosis, DGEA). DGEA mice were fed a standard or simvastatin-containing diet following surgery. Mice were euthanized 6 weeks post-operatively. RESULTS: Simvastatin significantly decreased proliferation and increased apoptosis in all cell lines. Compared to control animals, mice undergoing DGEA who were fed a standard diet demonstrated a fourfold increase in mucosal thickness and significant increase in proliferating cells (p < 0.0001). DGEA mice fed a simvastatin-containing diet had an attenuated response to reflux, with a significant reduction in mucosal hyperplasia and proliferation (p < 0.0001). DGEA mice fed a simvastatin-containing diet demonstrated significant upregulation of procaspase-3 (p = 0.009) and cleaved caspase-3 (p = 0.034) in the distal esophagus. CONCLUSIONS: We demonstrate for the first time a reduction in reflux-induced histologic changes of the distal esophagus following oral administration of simvastatin in vivo. These findings identify simvastatin as a potential preventative agent to inhibit the development and progression of reflux-induced esophageal injury.


Assuntos
Adenocarcinoma , Esôfago de Barrett , Neoplasias Esofágicas , Esofagite Péptica , Refluxo Gastroesofágico , Inibidores de Hidroximetilglutaril-CoA Redutases , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Anexina A5 , Esôfago de Barrett/tratamento farmacológico , Esôfago de Barrett/patologia , Caspase 3 , Modelos Animais de Doenças , Neoplasias Esofágicas/patologia , Refluxo Gastroesofágico/tratamento farmacológico , Refluxo Gastroesofágico/patologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Camundongos , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico
6.
Proc Natl Acad Sci U S A ; 116(12): 5514-5522, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30819901

RESUMO

Interleukin-37 (IL-37), a member of the IL-1 family of cytokines, is a fundamental suppressor of innate and acquired immunities. Here, we used an integrative approach that combines biophysical, biochemical, and biological studies to elucidate the unique characteristics of IL-37. Our studies reveal that single amino acid mutations at the IL-37 dimer interface that result in the stable formation of IL-37 monomers also remain monomeric at high micromolar concentrations and that these monomeric IL-37 forms comprise higher antiinflammatory activities than native IL-37 on multiple cell types. We find that, because native IL-37 forms dimers with nanomolar affinity, higher IL-37 only weakly suppresses downstream markers of inflammation whereas lower concentrations are more effective. We further show that IL-37 is a heparin binding protein that modulates this self-association and that the IL-37 dimers must block the activity of the IL-37 monomer. Specifically, native IL-37 at 2.5 nM reduces lipopolysaccharide (LPS)-induced vascular cell adhesion molecule (VCAM) protein levels by ∼50%, whereas the monomeric D73K mutant reduced VCAM by 90% at the same concentration. Compared with other members of the IL-1 family, both the N and the C termini of IL-37 are extended, and we show they are disordered in the context of the free protein. Furthermore, the presence of, at least, one of these extended termini is required for IL-37 suppressive activity. Based on these structural and biological studies, we present a model of IL-37 interactions that accounts for its mechanism in suppressing innate inflammation.


Assuntos
Tolerância Imunológica , Imunidade Inata , Interleucina-1/metabolismo , Linhagem Celular , Cristalografia por Raios X , Humanos , Tolerância Imunológica/imunologia , Tolerância Imunológica/fisiologia , Interleucina-1/genética , Interleucina-1/fisiologia , Espectroscopia de Ressonância Magnética , Multimerização Proteica
7.
J Surg Res ; 259: 562-568, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33261858

RESUMO

BACKGROUND: Gastroesophageal reflux and Barrett's esophagus are significant risk factors for the development of esophageal adenocarcinoma. Group IIa secretory phospholipase A2 (sPLA2) catalyzes the production of various proinflammatory metabolites and plays a critical role in promoting reflux-induced inflammatory changes within the distal esophagus. We hypothesized that inhibition of sPLA2 in human Barrett's cells would attenuate adhesion molecule expression via decreased activation of nuclear factor kappa B (NF-κB) and decrease cell proliferation, possibly mitigating the invasive potential of Barrett's esophagus. MATERIALS AND METHODS: Normal human esophageal epithelial cells (HET1A) and Barrett's cells (CPB) were assayed for baseline sPLA2 expression. CPB cells were treated with a specific inhibitor of sPLA2 followed by tumor necrosis factor-α. Protein expression was evaluated using immunoblotting. Cell proliferation was assessed using an MTS cell proliferation assay kit. Statistical analysis was performed using the Student's t-test or analysis of variance, where appropriate. RESULTS: CPB cells demonstrated higher baseline sPLA2 expression than HET1A cells (P = 0.0005). Treatment with 30 µM sPLA2 inhibitor significantly attenuated intercellular adhesion molecule-1 (P = 0.004) and vascular cell adhesion molecule-1 (P < 0.0001) expression as well as decreased NF-κB activation (P = 0.002). sPLA2 inhibition decreased cell proliferation in a dose-dependent manner (P < 0.001 for 15, 20, and 30 µM doses). CONCLUSIONS: sPLA2 inhibition in human Barrett's cells decreases cellular adhesive properties and NF-κB activation as well as decreases cell proliferation, signifying downregulation of the inflammatory response and possible attenuation of cellular malignant potential. These findings identify sPLA2 inhibition as a potential chemopreventive target for premalignant lesions of the esophagus.


Assuntos
Esôfago de Barrett/patologia , Esôfago/patologia , Fosfolipases A2 do Grupo II/antagonistas & inibidores , Ácidos Pentanoicos/farmacologia , Adenocarcinoma/patologia , Adenocarcinoma/prevenção & controle , Esôfago de Barrett/tratamento farmacológico , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/prevenção & controle , Esôfago/citologia , Fosfolipases A2 do Grupo II/metabolismo , Humanos , Ácidos Pentanoicos/uso terapêutico
8.
J Surg Res ; 260: 436-447, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33272595

RESUMO

BACKGROUND: Esophageal adenocarcinoma (EAC) is a lethal malignancy with poor prognosis. Pharmacologic inhibitors of inflammation, such as statins, have been shown to decrease the risk of development and progression of esophageal cancer, but the mechanism of this protection is unclear. The objective of this study was to elucidate the effect of statins on toll-like receptor 4-mediated-proliferation of human EAC cells and identify the mechanism responsible for these observed effects. METHODS: Human EAC cells (OE33 and FLO1) were treated with simvastatin or atorvastatin for increasing doses and time periods. Toll-like receptor 4 (TLR4) expression was assessed. Cells were pretreated with statin followed by lipopolysaccharide (LPS). Cell proliferation and expression of signaling proteins were evaluated. FLO1 cells were injected into the flank of nude mice. Mice received intraperitoneal injections of simvastatin, atorvastatin, or control solution and tumor volume was measured. RESULTS: OE33 and FLO1 cells demonstrated decreased TLR4 expression after treatment with simvastatin or atorvastatin for 8 h (P < 0.05). LPS increased proliferation, whereas pretreatment with statin abolished this response (P < 0.05). Statins decreased expression and activation of LPS-induced signaling proteins, including MyD88, TRAF6, Akt, and NF-κB (P < 0.05). Mice receiving daily statin injections demonstrated smaller tumors than control mice (P < 0.001 at day 33). CONCLUSIONS: Treatment of EAC cells with simvastatin or atorvastatin decreases TLR4-mediated proliferation and in vivo tumor growth. Decreased TLR4 expression and subsequent reduction in MyD88-dependent signaling could be a mechanism by which statins act to reduce tumor growth rates.


Assuntos
Adenocarcinoma/tratamento farmacológico , Biomarcadores Tumorais/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Neoplasias Esofágicas/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Receptor 4 Toll-Like/antagonistas & inibidores , Carga Tumoral/efeitos dos fármacos , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Atorvastatina/metabolismo , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Biomarcadores Tumorais/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Relação Dose-Resposta a Droga , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Nus , Fator 88 de Diferenciação Mieloide/metabolismo , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/metabolismo , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Receptor 4 Toll-Like/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Dig Dis Sci ; 66(3): 784-795, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32277371

RESUMO

BACKGROUND: Receptor tyrosine kinases of the epidermal growth factor receptor (EGFR) family such as human epidermal receptor-2 (HER2) are involved in the development and progression of esophageal adenocarcinoma (EAC). Prior studies have demonstrated that group IIa secretory phospholipase A2 (sPLA2 IIa) can function as a ligand for the EGFR family of receptors and lead to an increase in receptor signaling. AIMS: We hypothesized that sPLA2 IIa inhibition downregulates the expression of EGFR and HER-2 in EAC and through this mechanism decreases proliferation in EAC. METHODS: Normal human esophageal epithelium, Barrett's esophagus (BE), and EAC tissue samples were assayed for baseline expression of EGFR, HER-2, and sPLA2 IIa. sPLA2 IIa was attenuated via inhibitor or lentiviral knockdown in esophageal cell lines, and cells were assayed for EGFR and HER2 expression as well as proliferation. FLO1 EAC cells were injected into the flank of nude mice. After randomization, mice received daily group IIA sPLA2 inhibitor or a control solution, and tumor volume was measured with calipers. RESULTS: sPLA2 IIa, EGFR, and HER2 expression increased across the spectrum of normal esophageal epithelium to EAC. sPLA2 IIa inhibition and knockdown decreased the expression of HER-2 and EGFR and proliferation. Mice treated with sPLA2 IIa inhibitor had smaller tumors than controls. CONCLUSIONS: sPLA2 IIa inhibition decreases EGFR and HER2 expression and lowers proliferation of human EAC. The discovery of sPLA2 IIa inhibition's ability to attenuate growth factor receptor signaling underscores the exciting potential of sPLA2 IIa inhibitors as therapeutics in the treatment of EAC.


Assuntos
Adenocarcinoma/tratamento farmacológico , Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Neoplasias Esofágicas/tratamento farmacológico , Fosfolipases A2 do Grupo II/antagonistas & inibidores , Adenocarcinoma/enzimologia , Animais , Esôfago de Barrett/tratamento farmacológico , Esôfago de Barrett/enzimologia , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Mucosa Esofágica/enzimologia , Neoplasias Esofágicas/enzimologia , Humanos , Camundongos , Estudos Prospectivos , Receptor ErbB-2/metabolismo , Bancos de Tecidos
10.
Proc Natl Acad Sci U S A ; 114(7): 1631-1636, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28137840

RESUMO

Calcific aortic valve disease is a chronic inflammatory process, and aortic valve interstitial cells (AVICs) from diseased aortic valves express greater levels of osteogenic factors in response to proinflammatory stimulation. Here, we report that lower cellular levels of IL-37 in AVICs of diseased human aortic valves likely account for augmented expression of bone morphogenetic protein-2 (BMP-2) and alkaline phosphatase (ALP) following stimulation of Toll-like receptor (TLR) 2 or 4. Treatment of diseased AVICs with recombinant human IL-37 suppresses the levels of BMP-2 and ALP as well as calcium deposit formation. In mice, aortic valve thickening is observed when exposed to a TLR4 agonist or a high fat diet for a prolonged period; however, mice expressing human IL-37 exhibit significantly lower BMP-2 levels and less aortic valve thickening when subjected to the same regimens. A high fat diet in mice results in oxidized low-density lipoprotein (oxLDL) deposition in aortic valve leaflets. Moreover, the osteogenic responses in human AVICs induced by oxLDL are suppressed by recombinant IL-37. Mechanistically, reduced osteogenic responses to oxLDL in human AVICs are associated with the ability of IL-37 to inhibit NF-κB and ERK1/2. These findings suggest that augmented expression of osteogenic factors in AVICs of diseased aortic valves from humans is at least partly due to a relative IL-37 deficiency. Because recombinant IL-37 suppresses the osteogenic responses in human AVICs and alleviates aortic valve lesions in mice exposed to high fat diet or a proinflammatory stimulus, IL-37 has therapeutic potential for progressive calcific aortic valve disease.


Assuntos
Estenose da Valva Aórtica/prevenção & controle , Valva Aórtica/efeitos dos fármacos , Calcinose/prevenção & controle , Interleucinas/farmacologia , Osteogênese/efeitos dos fármacos , Idoso , Fosfatase Alcalina/metabolismo , Animais , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/metabolismo , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Calcinose/genética , Calcinose/metabolismo , Células Cultivadas , Feminino , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Proteínas Recombinantes/farmacologia
11.
J Stroke Cerebrovasc Dis ; 29(11): 105286, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33066914

RESUMO

BACKGROUND: Stroke is one of the leading causes of death and disability worldwide. Scalp acupuncture and exercise therapy have been proven as two effective methods for the treatment of stroke. However, their combined action and mechanisms have not been fully elucidated. The present study aimed to investigate the protective effect of scalp acupuncture combined with exercise therapy on neurons in rats with ischemic brain injury. METHODS: 100 rats were randomly divided into 5 groups including sham group, model group, acupuncture group, rehabilitation group, and experimental group (scalp acupuncture combined with exercise therapy). Middle cerebral artery occlusion (MCAO) model in rats was established according to Longa modified suture method to mimic ischemic stroke. The modified Bedexer's neurological function score was used to evaluate the neurological deficits of rats and the brain infarct volume was measured using 2, 3, 5-triphenyl tetrazolium chloride monohydrate (TTC) staining. Moreover, the apoptosis in the hippocampus was detected by western blotting and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The pro-inflammatory cytokines such as interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α), reactive oxygen species (ROS) and superoxide dismutase (SOD) were determined by corresponding kits. Immunohistochemistry or immunofluorescence was performed to detect the expression of brain-derived neurotrophic factor (BDNF), S100ß and glial fibrillary acidic protein (GFAP) in the hippocampi of rats. RESULTS: The neurological deficit score, the expression levels of apoptotic factors such as cleaved caspase-3 and Bax, and the TUNEL-positive cell rate of the experimental group were significantly lower than those of the acupuncture group and the rehabilitation group. However, apoptosis inhibitor Bcl-2 showed downregulated expression in the MCAO model rats but this trend was reverted by single and combinatorial treatments. In addition, the contents of TNF-α, IL-1ß and ROS in the acupuncture group and the rehabilitation group were significantly lower than those in the model group, but higher than the experimental group. While the opposite results were obtained in SOD activity. Furthermore, compared with the model group, the ratios of BDNF, S100ß, and GFAP-positive cells in the acupuncture, rehabilitation and experimental groups were significantly increased, and the highest ratios were recorded in the experimental group. CONCLUSIONS: This study demonstrated that scalp acupuncture combined with exercise therapy effectively counteracts ischemic brain injury via the downregulation of pro-inflammatory mediators and ROS, the increased production of the antioxidant enzyme SOD, neurotrophic factor BDNF and astrocyte activities.


Assuntos
Pontos de Acupuntura , Terapia por Acupuntura , Apoptose , Encéfalo/patologia , Terapia por Exercício , Infarto da Artéria Cerebral Média/prevenção & controle , Couro Cabeludo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Comportamento Animal , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/psicologia , Mediadores da Inflamação/metabolismo , Masculino , Necrose , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Superóxido Dismutase/metabolismo
12.
Int J Mol Sci ; 21(4)2020 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-32074942

RESUMO

BACKGROUND: Calcific aortic valve disease (CAVD) is a chronic inflammatory disease that manifests as progressive valvular fibrosis and calcification. An inflammatory milieu in valvular tissue promotes fibrosis and calcification. Aortic valve interstitial cell (AVIC) proliferation and the over-production of the extracellular matrix (ECM) proteins contribute to valvular thickening. However, the mechanism underlying elevated AVIC fibrogenic activity remains unclear. Recently, we observed that AVICs from diseased aortic valves express higher levels of neurotrophin 3 (NT3) and that NT3 exerts pro-osteogenic and pro-fibrogenic effects on human AVICs. HYPOTHESIS: Pro-inflammatory stimuli upregulate NT3 production in AVICs to promote fibrogenic activity in human aortic valves. METHODS AND RESULTS: AVICs were isolated from normal human aortic valves and were treated with lipopolysaccharide (LPS, 0.20 µg/mL). LPS induced TLR4-dependent NT3 production. This effect of LPS was abolished by inhibition of the Akt and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) pathways. The stimulation of TLR4 in human AVICs with LPS resulted in a greater proliferation rate and an upregulated production of matrix metallopeptidases-9 (MMP-9) and collagen III, as well as augmented collagen deposition. Recombinant NT3 promoted AVIC proliferation in a tropomyosin receptor kinase (Trk)-dependent fashion. The neutralization of NT3 or the inhibition of Trk suppressed LPS-induced AVIC fibrogenic activity. CONCLUSIONS: The stimulation of TLR4 in human AVICs upregulates NT3 expression and promotes cell proliferation and collagen deposition. The NT3-Trk cascade plays a critical role in the TLR4-mediated elevation of fibrogenic activity in human AVICs. Upregulated NT3 production by endogenous TLR4 activators may contribute to aortic valve fibrosis associated with CAVD progression.


Assuntos
Cardiopatias Congênitas/patologia , Doenças das Valvas Cardíacas/patologia , Neurotrofina 3/metabolismo , Receptor 4 Toll-Like/metabolismo , Valva Aórtica/citologia , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Doença da Válvula Aórtica Bicúspide , Carbazóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Feminino , Cardiopatias Congênitas/metabolismo , Doenças das Valvas Cardíacas/metabolismo , Humanos , Alcaloides Indólicos/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor trkA/antagonistas & inibidores , Receptor trkA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Regulação para Cima/efeitos dos fármacos
13.
Mol Cell Biochem ; 456(1-2): 145-156, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30684134

RESUMO

Secretory phospholipase A2 IIa (sPLA2 IIa) catalyzes the production of multiple inflammatory mediators that influence the development of lung and other cancers. The most potent of these carcinogenic mediators is prostaglandin E2 (PGE2). We hypothesize that sPLA2 IIa inhibition modulates the production of PGE2, and sPLA2 IIa inhibition exerts its antineoplastic effects via downregulation of PGE2 production. We aim to evaluate these relationships via analysis of PGE2-mediated growth regulation pathways. A549 and H1650 lung adenocarcinoma cells were assayed for PGE2 production in the presence of sPLA2 IIa inhibitor. A549 and H1650 cells were treated with PGE2 and immune blotting was performed to assess ICAM-1 expression and STAT-3 activity. PGE2-induced ICAM-1 expression was measured via immunofluorescence. A549 and H1650 cells were treated with PGE2 in the presence of STAT3 inhibitor and assayed for ICAM-1 expression. A549 cells were treated with PGE2 in the presence ICAM-1 blocking antibody and assayed for invasion. PGE2 stimulation significantly increased the invasiveness and proliferation of lung adenocarcinoma (invasion p < 0.05, proliferation p < 0.05 A549 cells, p < 0.005 H1650 cells). sPLA2 IIa inhibition reduced PGE2 secretion (p < 0.05). PGE2 stimulation significantly upregulated the expression of cell adhesion molecule ICAM-1 and the phosphorylation of anti-apoptotic transcription factor STAT3 (p < 0.05). STAT3 inhibition attenuated ICAM-1 expression demonstrating the dependence of ICAM-1 on the STAT3 pathway (p < 0.05). ICAM-1 blockade attenuated the pro-invasive effects of PGE2 (p < 0.05). sPLA2 IIa inhibition attenuates the potent effects of PGE2-induced invasiveness. This is mediated by decreasing pro-inflammatory and invasion-promoting ICAM-1via the STAT-3 pathway. These data further describe how sPLA2 IIa inhibition mechanistically exerts its anticancer effects and support its use as an antineoplastic agent.


Assuntos
Adenocarcinoma de Pulmão/enzimologia , Dinoprostona/metabolismo , Neoplasias Pulmonares/enzimologia , Proteínas de Neoplasias/metabolismo , Fosfolipases A2 Secretórias/metabolismo , Células A549 , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Fator de Transcrição STAT3/metabolismo
14.
J Surg Res ; 235: 44-51, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30691826

RESUMO

BACKGROUND: Calcific aortic valve disease (CAVD) is an age-related and slowly progressive valvular disorder. Overexpression of matrix metalloproteinase 12 (MMP-12) has been found in atherosclerosis, stiffed vascular tissue, and calcified aortic valves. We hypothesized that MMP-12 may induce the pro-osteogenic responses in human aortic valve interstitial cells (AVICs). METHODS: Human AVICs were isolated from normal and calcified aortic valves. Cells were treated with MMP-12. The pro-osteogenic marker Runt-related transcription factor 2 (RUNX-2), bone morphogenetic protein 2 (BMP-2), and alkaline phosphatase (ALP), as well as MMP-12-associated signaling molecules, were analyzed. RESULTS: Human calcified aortic valves expressed significantly higher MMP-12 than normal human aortic valves. MMP-12-induced the expression of RUNX-2, BMP-2, ALP, and calcium deposit formation. Suppression of MMP-12 by its inhibitor decreased the expression of RUNX-2, BMP-2, and ALP. MMP-12-induced osteogenic responses were associated with higher levels of phosphorylation of p38 mitogen-activated protein kinases (MAPK), low density lipoprotein-related protein 6 (LRP-6), and ß-catenin signaling molecules. Calcified aortic valves exhibited markedly higher levels of LRP-6 and ß-catenin levels. Inhibition of either p38 MAPK or LRP-6 attenuated MMP-12-induced expression of RUNX-2, BMP-2, and ALP. Suppression of p38 MAPK abrogated MMP-12-induced activation of LRP-6 and ß-catenin signaling pathways. CONCLUSIONS: MMP-12 induces pro-osteogenic responses in AVICs by activation of p38 MAPK-mediated LRP-6 and ß-catenin signaling pathways. The study revealed that the potential role of MMP-12 in the pathogenesis of CAVD and therapeutically targeting MMP-12 may suppress the development of CAVD.


Assuntos
Valva Aórtica/citologia , Metaloproteinase 12 da Matriz/fisiologia , Osteogênese/fisiologia , Idoso , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Estenose da Valva Aórtica/etiologia , Estenose da Valva Aórtica/metabolismo , Proteína Morfogenética Óssea 2/fisiologia , Calcinose/etiologia , Calcinose/metabolismo , Células Cultivadas , Feminino , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Pessoa de Meia-Idade , Transdução de Sinais , beta Catenina/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
15.
J Surg Res ; 233: 124-131, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30502238

RESUMO

BACKGROUND: Paraplegia remains a significant complication of thoracoabdominal aortic intervention. We previously reported that diazoxide (DZ), enhances the neuroprotective efficacy of erythropoietin (EPO). We hypothesized that DZ and EPO combined treatment attenuates spinal cord ischemic injury through upregulation of nerve growth factor (NGF). METHODS: DZ (pretreatment) was given to adult male C57/BL6 mice by oral gavage and EPO (before surgery) was intraperitoneally injected 32 h after administration of DZ. Spinal cords were harvested 0, 2, 4, and 6 h after injection of EPO. NGF expression was analyzed by western blot. After determining the optimal time, NGF expression was compared between DZ (pretreatment) + EPO (before surgery), DZ + PBS, PBS + EPO, and PBS + PBS (ischemic control). Four groups were studied to compare the motor function after ischemia: DZ + EPO (n = 11), ischemic control (n = 9), DZ + EPO + tropomyosin receptor kinase A receptor inhibitor (n = 9), and sham (without cross-clamp, n = 4). Spinal cord ischemia was induced by a 4-min thoracic aortic cross-clamp. Functional scoring (Basso Mouse Score) was done at 12-h intervals until 48 h, and spinal cords were harvested for evaluation of NGF expression and histological changes. RESULTS: NGF expression was significantly upregulated 4 h after administration of EPO. At 4 h after injection of EPO, NGF expression in the DZ + EPO group was significantly higher than that in the other groups. DZ + EPO significantly preserved motor function compared with all other groups. At 48 h after reperfusion, the level of NGF expression in the DZ + EPO group, was significantly higher than in all other groups. CONCLUSIONS: DZ + EPO attenuates spinal cord ischemic injury through upregulation of NGF. Better understanding of this mechanism may serve to further prevent ischemic complications for aortic intervention.


Assuntos
Diazóxido/administração & dosagem , Eritropoetina/administração & dosagem , Fator de Crescimento Neural/metabolismo , Isquemia do Cordão Espinal/prevenção & controle , Animais , Aneurisma da Aorta Torácica/cirurgia , Diazóxido/farmacocinética , Modelos Animais de Doenças , Sinergismo Farmacológico , Eritropoetina/farmacocinética , Humanos , Masculino , Camundongos , Paraplegia/etiologia , Paraplegia/prevenção & controle , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Isquemia do Cordão Espinal/etiologia , Isquemia do Cordão Espinal/patologia , Regulação para Cima/efeitos dos fármacos , Procedimentos Cirúrgicos Vasculares/efeitos adversos
16.
Int J Mol Sci ; 21(1)2019 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-31861929

RESUMO

Aortic valve interstitial cells (AVICs) play a major role in valvular calcification associated with calcific aortic valve disease (CAVD). Although AVICs from diseased valves display a pro-osteogenic phenotype, the underlying mechanism causing this remains unclear. MicroRNA-204 (miR-204) is a negative regulator of osteoblast differentiation. We sought to analyze miR-204 expression in diseased human aortic valves and determine the role of this miR in AVIC osteogenic activity associated with CAVD pathobiology. In situ hybridization and PCR analysis revealed miR-204 deficiency in diseased valves and in AVICs from diseased valves. MiR-204 mimic suppressed alkaline phosphatase (ALP) expression and calcium deposition in AVICs from diseased valves. MiR-204 antagomir enhanced ALP expression in AVICs from normal valves through induction of Runx2 and Osx, and expression of miR-204 antagomir in mouse aortic valves promoted calcium deposition through up-regulation of Runx2 and Osx. Further, miR-204 mimic suppressed the osteogenic responses to TGF-ß1 in AVICs of normal valves. In conclusion, miR-204 deficiency contributes to the mechanism underlying elevated osteogenic activity in diseased aortic valves, and miR-204 is capable of reversing the pro-osteogenic phenotype of AVICs of diseased valves and suppressing AVIC osteogenic response to stimulation. Exogenous miR-204 may have therapeutic potential for inhibiting valvular calcification associated with CAVD progression.


Assuntos
Estenose da Valva Aórtica/genética , Valva Aórtica/patologia , Calcinose/genética , Regulação para Baixo , MicroRNAs/genética , Osteogênese , Idoso , Animais , Valva Aórtica/citologia , Valva Aórtica/metabolismo , Estenose da Valva Aórtica/patologia , Calcinose/patologia , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
17.
J Mol Cell Cardiol ; 119: 40-50, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29660306

RESUMO

Aging is usually accompanied with overt structural and functional changes as well as suppressed autophagy in the heart although the precise regulatory mechanisms are somewhat unknown. Here we evaluated the role of the innate proinflammatory mediator toll-like receptor 4 (TLR4) in cardiac aging and the underlying mechanism with a focus on autophagy. Cardiac geometry and function were monitored in young or old wild-type (WT) and TLR4 knockout (TLR4-/-) mice using echocardiography, IonOptix® edge-detection and fura-2 techniques. Levels of autophagy and mitophagy, nuclear receptor corepressor 1 (NCoR1) and histone deacetylase I (HDAC1) were examined using western blot. Transmission electronic microscopy (TEM) was employed to monitor myocardial ultrastructure. Our results revealed that TLR4 ablation alleviated advanced aging (24 months)-induced changes in myocardial remodeling (increased heart weight, chamber size, cardiomyocyte cross-sectional area), contractile function and intracellular Ca2+ handling as well as autophagy and mitophagy [Beclin-1, Atg5, LC3B, PTEN-induced putative kinase 1 (PINK1), Parkin and p62]. Aging downregulated levels of NCoR1 and HDAC1 as well as their interaction, the effects were significantly attenuated or negated by TLR4 ablation. Advanced aging disturbed myocardial ultrastructure as evidenced by loss of myofilament alignment and swollen mitochondria, which was obliterated by TLR4 ablation. Moreover, aging suppressed autophagy (GFP-LC3B puncta) in neonatal mouse cardiomyocytes, the effect of which was negated by the TLR4 inhibitor CLI-095. Inhibition of HDCA1 using apicidin cancelled off CLI095-induced beneficial response of GFP-LC3B puncta against aging. Our data collectively indicate a role for TLR4-mediated autophagy in cardiac remodeling and contractile dysfunction in aging through a HDAC1-NCoR1-dependent mechanism.


Assuntos
Envelhecimento/genética , Histona Desacetilase 1/genética , Correpressor 1 de Receptor Nuclear/genética , Receptor 4 Toll-Like/genética , Envelhecimento/patologia , Animais , Animais Recém-Nascidos , Autofagia/genética , Histona Desacetilase 1/antagonistas & inibidores , Humanos , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Remodelação Ventricular/genética , Remodelação Ventricular/fisiologia
18.
J Biol Chem ; 292(21): 8657-8666, 2017 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-28377507

RESUMO

Calcific aortic valve disease (CAVD) is common in the elderly population, but pharmacological interventions for managing valvular calcification are unavailable. Transforming growth factor ß1 (TGF-ß1) and bone morphogenetic protein 2 (BMP-2) induce pro-osteogenic activation of human aortic valve interstitial cells (AVICs) that play an important role in valvular calcification. However, the molecular mechanism underlying pro-osteogenic activation in AVICs is incompletely understood. Here, we investigated an epigenetic regulatory mechanism in human AVIC pro-osteogenic activation induced by TGF-ß1 and BMP-2. Microarray and real-time PCR analyses revealed that microRNA (miR)-486 up-regulation and miR-204 down-regulation were characteristic changes in TGF-ß1- and BMP-2-stimulated normal AVICs and in AVICs from calcified valves. Both TGF-ß1 and BMP-2 down-regulated miR-204 through Smad pathways. Interestingly, an miR-486 antagomir diminished the effect of TGF-ß1 and BMP-2 on miR-204 levels and calcium deposit formation. Furthermore, the miR-486 antagomir increased the expression of Smurf2, a Smad inhibitor, in the presence or absence of TGF-ß1 or BMP-2 stimulation, whereas a miR-486 mimic reduced Smurf2 expression. Smurf2 knockdown augmented TGF-ß1- or BMP-2-induced miR-204 down-regulation and resulted in increased expression of the osteoblastic biomarkers Osx and Runx2. In summary, we found that TGF-ß1 and BMP-2 up-regulate miR-486 and down-regulate miR-204 in human AVICs to promote pro-osteogenic activity and that miR-486 inhibits Smurf2 expression to augment the miR-204 down-regulation. We conclude that the miR-486-Smurf2-Smad loop plays an important role in regulating AVIC pro-osteogenic activation in response to TGF-ß1 or BMP-2. Targeting this regulatory loop may have therapeutic potential for suppressing aortic valve calcification.


Assuntos
Valva Aórtica/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Epigênese Genética , Doenças das Valvas Cardíacas/metabolismo , Osteogênese , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Calcificação Vascular/metabolismo , Idoso , Valva Aórtica/patologia , Células Cultivadas , Regulação para Baixo , Feminino , Doenças das Valvas Cardíacas/patologia , Humanos , Masculino , MicroRNAs/biossíntese , Pessoa de Meia-Idade , Osteoblastos/metabolismo , Osteoblastos/patologia , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima , Calcificação Vascular/patologia
19.
J Surg Res ; 230: 101-109, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30100024

RESUMO

BACKGROUND: Calcific aortic stenosis is a chronic inflammatory disease. Proinflammatory stimulation via toll-like receptor 4 (TLR4) causes the aortic valve interstitial cell (AVIC) to undergo phenotypic change. The AVIC first assumes an inflammatory phenotype characterized by the production of inflammatory mediators such as intercellular adhesion molecule-1 (ICAM-1), interleukin-8 (IL-8), and monocyte chemoattractant protein-1 (MCP-1). This change has been linked with an osteogenic phenotypic response. Statins have recently been shown to have anti-inflammatory properties. We therefore hypothesized that statins may have an anti-inflammatory effect on human AVICs by downregulation of TLR4-stimulated inflammatory responses. Our purposes were (1) to determine the effect of simvastatin on TLR4-induced expression of inflammatory mediators in human AVICs and (2) to determine the mechanism(s) through which simvastatin exert this effect. MATERIALS AND METHODS: Human AVICs were isolated from the explanted hearts of four patients undergoing cardiac transplantation. Cells were treated with simvastatin (50 µM) for 1 h before stimulation with TLR4 agonist lipopolysaccharide (LPS, 0.2 µg/mL). Immunoblotting (IB) was used to analyze cell lysates for ICAM-1 expression, and enzyme-linked immunosorbent assay was used to detect IL-8 and MCP-1 in cell culture media. Likewise, lysates were analyzed for TLR4 and nuclear factor-kappa B activation (IB). After simvastatin treatment, lysates were analyzed for TLR4 levels (IB). Statistics were by analysis of variance (P < 0.05). RESULTS: Simvastatin reduced TLR4-induced ICAM-1, IL-8, and MCP-1 expression in AVICs. Simvastatin down-regulated TLR4 levels and suppressed TLR4-induced phosphorylation of nuclear factor-kappa B. CONCLUSIONS: These data demonstrate the potential of a medical therapy (simvastatin) to impact the pathogenesis of aortic stenosis.


Assuntos
Estenose da Valva Aórtica/tratamento farmacológico , Valva Aórtica/patologia , Calcinose/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Sinvastatina/farmacologia , Receptor 4 Toll-Like/imunologia , Adulto , Valva Aórtica/citologia , Valva Aórtica/imunologia , Estenose da Valva Aórtica/imunologia , Estenose da Valva Aórtica/patologia , Calcinose/imunologia , Calcinose/patologia , Cardiomiopatia Dilatada/cirurgia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Transplante de Coração , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Masculino , Pessoa de Meia-Idade , Miofibroblastos , Cultura Primária de Células , Sinvastatina/uso terapêutico
20.
Arterioscler Thromb Vasc Biol ; 37(7): 1339-1351, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28546218

RESUMO

OBJECTIVE: Extracellular matrix proteinases are implicated in the pathogenesis of calcific aortic valve disease. The ADAMTS5 (a disintegrin and metalloproteinase with thrombospondin motifs 5) enzyme is secreted, matrix-associated metalloendopeptidase, capable of degrading extracellular matrix proteins, particularly matrilin 2. We sought to determine the role of the ADAMTS5/matrilin 2 axis in mediating the phenotype transition of valvular interstitial cells (VICs) associated with calcific aortic valve disease. APPROACH AND RESULTS: Levels of ADAMTS5, matrilin 2, and α-SMA (α-smooth muscle actin) were evaluated in calcified and normal human aortic valve tissues and VICs. Calcified aortic valves have reduced levels of ADAMTS5 and higher levels of matrilin 2 and α-SMA. Treatment of normal VICs with soluble matrilin 2 caused an increase in α-SMA level through Toll-like receptors 2 and 4, which was accompanied by upregulation of runt-related transcription factor 2 and alkaline phosphatase. In addition, ADAMTS5 knockdown in normal VICs enhanced the effect of matrilin 2. Matrilin 2 activated nuclear factor (NF) κB and NF of activated T cells complex 1 and induced the interaction of these 2 NFs. Inhibition of either NF-κB or NF of activated T cells complex 1 suppressed matrilin 2's effect on VIC phenotype change. Knockdown of α-SMA reduced and overexpression of α-SMA enhanced the expression of pro-osteogenic factors and calcium deposit formation in human VICs. CONCLUSIONS: Matrilin 2 induces myofibroblastic transition and elevates pro-osteogenic activity in human VICs via activation of NF-κB and NF of activated T cells complex 1. Myofibroblastic transition in human VICs is an important mechanism of elevating the pro-osteogenic activity. Matrilin 2 accumulation associated with relative ADAMTS5 deficiency may contribute to the mechanism underlying calcific aortic valve disease progression.


Assuntos
Proteína ADAMTS5/deficiência , Estenose da Valva Aórtica/enzimologia , Valva Aórtica/enzimologia , Valva Aórtica/patologia , Calcinose/enzimologia , Transdiferenciação Celular , Miofibroblastos/enzimologia , Osteogênese , Proteína ADAMTS5/genética , Actinas/genética , Actinas/metabolismo , Adulto , Idoso , Fosfatase Alcalina/metabolismo , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/patologia , Calcinose/genética , Calcinose/patologia , Estudos de Casos e Controles , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , Masculino , Proteínas Matrilinas/metabolismo , Pessoa de Meia-Idade , Miofibroblastos/patologia , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Fenótipo , Interferência de RNA , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA