Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 228, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38777955

RESUMEN

Diabetic cardiomyopathy (DCM) is a prevalent complication of type 2 diabetes (T2D). 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) is a glycolysis regulator. However, the potential effects of PFKFB3 in the DCM remain unclear. In comparison to db/m mice, PFKFB3 levels decreased in the hearts of db/db mice. Cardiac-specific PFKFB3 overexpression inhibited myocardial oxidative stress and cardiomyocyte apoptosis, suppressed mitochondrial fragmentation, and partly restored mitochondrial function in db/db mice. Moreover, PFKFB3 overexpression stimulated glycolysis. Interestingly, based on the inhibition of glycolysis, PFKFB3 overexpression still suppressed oxidative stress and apoptosis of cardiomyocytes in vitro, which indicated that PFKFB3 overexpression could alleviate DCM independent of glycolysis. Using mass spectrometry combined with co-immunoprecipitation, we identified optic atrophy 1 (OPA1) interacting with PFKFB3. In db/db mice, the knockdown of OPA1 receded the effects of PFKFB3 overexpression in alleviating cardiac remodeling and dysfunction. Mechanistically, PFKFB3 stabilized OPA1 expression by promoting E3 ligase NEDD4L-mediated atypical K6-linked polyubiquitination and thus prevented the degradation of OPA1 by the proteasomal pathway. Our study indicates that PFKFB3/OPA1 could be potential therapeutic targets for DCM.


Asunto(s)
Cardiomiopatías Diabéticas , GTP Fosfohidrolasas , Miocitos Cardíacos , Fosfofructoquinasa-2 , Ubiquitinación , Fosfofructoquinasa-2/metabolismo , Fosfofructoquinasa-2/genética , Animales , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/genética , Ratones , GTP Fosfohidrolasas/metabolismo , GTP Fosfohidrolasas/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Masculino , Estrés Oxidativo , Apoptosis/genética , Miocardio/metabolismo , Miocardio/patología , Ratones Endogámicos C57BL , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Glucólisis , Humanos , Estabilidad Proteica
2.
Cancer Sci ; 114(5): 1972-1985, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36692143

RESUMEN

The Brother of Regulator of Imprinted Sites (BORIS, gene symbol CTCFL) has previously been shown to promote colorectal cancer cell proliferation, inhibit cancer cell apoptosis, and resist chemotherapy. However, it is unknown whether Boris plays a role in the progression of in situ colorectal cancer. Here Boris knockout (KO) mice were constructed. The function loss of the cloned Boris mutation that was retained in KO mice was verified by testing its activities in colorectal cell lines compared with the Boris wild-type gene. Boris knockout reduced the incidence and severity of azoxymethane/dextran sulfate-sodium (AOM/DSS)-induced colon cancer. The importance of Boris is emphasized in the progression of in situ colorectal cancer. Boris knockout significantly promoted the phosphorylation of γH2AX and the DNA damage in colorectal cancer tissues and suppressed Wnt and MAPK pathways that are responsible for the callback of DNA damage repair. This indicates the strong inhibition of colorectal cancer in Boris KO mice. By considering that the DSS-promoted inflammation contributes to tumorigenesis, Boris KO mice were also studied in DSS-induced colitis. Our data showed that Boris knockout alleviated DSS-induced colitis and that Boris knockdown inhibited the NF-κB signaling pathway in RAW264.7 cells. Therefore Boris knockout eliminates colorectal cancer generation by inhibiting DNA damage repair in cancer cells and relieving inflammation in macrophages. Our findings demonstrate the importance of Boris in the development of in situ colorectal cancer and provide evidence for the feasibility of colorectal cancer therapy on Boris.


Asunto(s)
Colitis , Neoplasias Colorrectales , Animales , Masculino , Ratones , Azoximetano/toxicidad , Colitis/inducido químicamente , Colitis/genética , Colitis/complicaciones , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/tratamiento farmacológico , Sulfato de Dextran/toxicidad , Sulfato de Dextran/uso terapéutico , Modelos Animales de Enfermedad , Daño del ADN/genética , Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Chemistry ; 29(58): e202301879, 2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37706579

RESUMEN

The underlying biophysical principle governing the cytotoxicity of the oligomeric aggregates of ß-amyloid (Aß) peptides has long been an enigma. Here we show that the size of Aß40 oligomers can be actively controlled by incubating the peptides in reverse micelles. Our approach allowed for the first time a detailed comparison of the structures and dynamics of two Aß40 oligomers of different sizes, viz., 10 and 23 nm, by solid-state NMR. From the chemical shift data, we infer that the conformation and/or the chemical environments of the residues from K16 to K28 are different between the 10-nm and 23-nm oligomers. We find that the 10-nm oligomers are more cytotoxic, and the molecular motion of the sidechain of its charged residue K16 is more dynamic. Interestingly, the residue A21 exhibits unusually high structural rigidity. Our data raise an interesting possibility that the cytotoxicity of Aß40 oligomers could also be correlated to the motional dynamics of the sidechains.


Asunto(s)
Péptidos beta-Amiloides , Micelas , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/química , Espectroscopía de Resonancia Magnética , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/química , Amiloide/química
4.
Prostaglandins Other Lipid Mediat ; 167: 106740, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37119935

RESUMEN

Epoxyeicosatrienoic acids (EETs), which are synthesized from arachidonic acid by cytochrome P450 epoxygenases, function primarily as autocrine and paracrine effectors in the cardiovascular system. So far, most research has focused on the vasodilatory, anti-inflammatory, anti-apoptotic and mitogenic properties of EETs in the systemic circulation. However, whether EETs could suppress tissue factor (TF) expression and prevent thrombus formation remains unknown. Here we utilized in vivo and in vitro models to investigate the effects and underlying mechanisms of exogenously EETs on LPS induced TF expression and inferior vein cava ligation induced thrombosis. We observed that the thrombus formation rate and the size of the thrombus were greatly reduced in 11,12-EET treated mice,accompanied by decreased TF and inflammatory cytokines expression. Further in vitro studies showed that by enhancing p38 MAPK activation and subsequent tristetraprolin (TTP) phosphorylation, LPS strengthened the stability of TF mRNA and induced increased TF expression. However, by strengthening PI3K-dependent Akt phosphorylation, which acted as a negative regulator of p38-TTP signaling pathway,11,12-EET reduced LPS-induced TF expression in monocytes. In addition, 11,12-EET inhibited LPS-induced NF-κB nuclear translocation by activating the PI3K/Akt pathway. Further study indicated that the inhibitory effect of 11,12-EET on TF expression was mediated by antagonizing LPS-induced activation of thromboxane prostanoid receptor. In conclusion, our study demonstrated that 11,12-EET prevented thrombosis by reducing TF expression and targeting the CYP2J2 epoxygenase pathway may represent a novel approach to mitigate thrombosis related diseases.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Trombosis , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Lipopolisacáridos/farmacología , Tromboplastina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Transducción de Señal , Citocromo P-450 CYP2J2 , Ácido 8,11,14-Eicosatrienoico/metabolismo , Trombosis/tratamiento farmacológico , Estabilidad del ARN
5.
J Biol Chem ; 296: 100667, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33864813

RESUMEN

The epoxyeicosatrienoic acid (EET) exerts beneficial effects on insulin resistance and/or hypertension. EETs could be readily converted to less biological active diols by soluble epoxide hydrolase (sEH). However, whether sEH inhibition can ameliorate the comorbidities of insulin resistance and hypertension and the underlying mechanisms of this relationship are unclear. In this study, C57BL/6 mice were rendered hypertensive and insulin resistant through a high-fat and high-salt (HF-HS) diet. The sEH inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), was used to treat mice (1 mg/kg/day) for 8 weeks, followed by analysis of metabolic parameters. The expression of sEH and the sodium-glucose cotransporter 2 (SGLT2) was markedly upregulated in the kidneys of mice fed an HF-HS diet. We found that TPPU administration increased kidney EET levels, improved insulin resistance, and reduced hypertension. Furthermore, TPPU treatment prevented upregulation of SGLT2 and the associated increased urine volume and the excretion of urine glucose and urine sodium. Importantly, TPPU alleviated renal inflammation. In vitro, human renal proximal tubule epithelial cells (HK-2 cells) were used to further investigate the underlying mechanism. We observed that 14,15-EET or sEH knockdown or inhibition prevented the upregulation of SGLT2 upon treatment with palmitic acid or NaCl by inhibiting the inhibitory kappa B kinase α/ß/NF-κB signaling pathway. In conclusion, sEH inhibition by TPPU alleviated insulin resistance and hypertension induced by an HF-HS diet in mice. The increased urine excretion of glucose and sodium was mediated by decreased renal SGLT2 expression because of inactivation of the inhibitory kappa B kinase α/ß/NF-κB-induced inflammatory response.


Asunto(s)
Epóxido Hidrolasas/antagonistas & inhibidores , Regulación de la Expresión Génica , Hipertensión/prevención & control , Resistencia a la Insulina , Riñón/metabolismo , Enfermedades Metabólicas/prevención & control , Transportador 2 de Sodio-Glucosa/metabolismo , Animales , Regulación hacia Abajo , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/patología , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Transportador 2 de Sodio-Glucosa/genética
6.
Pharmacol Res ; 183: 106367, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35882293

RESUMEN

Diabetic nephropathy (DN) is the most common cause of end-stage renal disease, and few treatment options that prevent the progressive loss of renal function are available. Studies have shown that dietary fiber intake improves kidney diseases and metabolism-related diseases, most likely through short-chain fatty acids (SCFAs). The present study aimed to examine the protective effects of inulin-type fructans (ITFs) on DN through 16 S rRNA gene sequencing, gas chromatographymass spectrometry (GCMS) analysis and fecal microbiota transplantation (FMT). The results showed that ITFs supplementation protected against kidney damage in db/db mice and regulated the composition of the gut microbiota. Antibiotic treatment and FMT experiments further demonstrated a key role of the gut microbiota in mediating the beneficial effects of ITFs. The ITFs treatment-induced changes in the gut microbiota led to an enrichment of SCFA-producing bacteria, especially the genera Akkermansia and Candidatus Saccharimonas, which increased the fecal and serum acetate concentrations. Subsequently, acetate supplementation improved glomerular damage and renal fibrosis by attenuating mitochondrial dysfunction and reducing toxic glucose metabolite levels. In conclusion, ITFs play a renoprotective role by modulating the gut microbiota and increasing acetate production. Furthermore, acetate mediates renal protection by regulating glucose metabolism, decreasing glycotoxic product levels and improving mitochondrial function.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Microbioma Gastrointestinal , Animales , Bacterias/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/prevención & control , Ácidos Grasos Volátiles/metabolismo , Fructanos/farmacología , Fructanos/uso terapéutico , Inulina/metabolismo , Inulina/uso terapéutico , Ratones
7.
Am J Physiol Heart Circ Physiol ; 321(2): H353-H368, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34142887

RESUMEN

Arterial stiffness, a consequence of smoking, is an underlying risk factor of cardiovascular diseases. Epoxyeicosatrienoic acids (EETs), hydrolyzed by soluble epoxide hydrolase (sEH), have beneficial effects against vascular dysfunction. However, the role of sEH knockout in nicotine-induced arterial stiffness was not characterized. We hypothesized that sEH knockout could prevent nicotine-induced arterial stiffness. In the present study, Ephx2 (the gene encodes sEH enzyme) null (Ephx2-/-) mice and wild-type (WT) littermate mice were infused with or without nicotine and administered with or without nicotinamide [NAM, sirtuin-1 (SIRT1) inhibitor] simultaneously for 4 wk. Nicotine treatment increased sEH expression and activity in the aortas of WT mice. Nicotine infusion significantly induced vascular remodeling, arterial stiffness, and SIRT1 deactivation in WT mice, which was attenuated in Ephx2 knockout mice (Ephx2-/- mice) without NAM treatment. However, the arterial protective effects were gone in Ephx2-/- mice with NAM treatment. In vitro, 11,12-EET treatment attenuated nicotine-induced matrix metalloproteinase 2 (MMP2) upregulation via SIRT1-mediated yes-associated protein (YAP) deacetylation. In conclusion, sEH knockout attenuated nicotine-induced arterial stiffness and vascular remodeling via SIRT1-induced YAP deacetylation.NEW & NOTEWORTHY We presently show that sEH knockout repressed nicotine-induced arterial stiffness and extracellular matrix remodeling via SIRT1-induced YAP deacetylation, which highlights that sEH is a potential therapeutic target in smoking-induced arterial stiffness and vascular remodeling.


Asunto(s)
Aorta/efectos de los fármacos , Epóxido Hidrolasas/genética , Niacinamida/farmacología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Sirtuina 1/metabolismo , Rigidez Vascular/efectos de los fármacos , Complejo Vitamínico B/farmacología , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacología , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Aorta/metabolismo , Aorta/fisiopatología , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/genética , Ratones , Ratones Noqueados , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/efectos de los fármacos , Rigidez Vascular/genética , Vasodilatadores/farmacología , Proteínas Señalizadoras YAP
8.
Pharm Dev Technol ; 26(9): 943-952, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34372745

RESUMEN

Oral absorption of peptides/proteins is usually compromised by various gastrointestinal tract barriers. To improve delivery efficiency, chitosan-conjugated deoxycholic acid (CS-DCA) coupled with sodium alginate (ALG) was prepared to load insulin into pH-sensitive nanoparticles. The insulin-loaded chitosan-deoxycholic acid/alginate nanoparticles (CDA NPs) were characterized by size (143.3 ± 10.8 nm), zeta potential (19.5 ± 1.6 mV), entrapment efficiency (61.14 ± 1.67%), and insulin drug loading (3.36 ± 0.09%). The CDA NPs exhibited pH-triggered release characteristics in vitro and protected the wrapped insulin from gastric degradation. Stability of the CDA NPs in enzyme-containing simulated gastrointestinal fluids suggested that the NPs could partially protect the wrapped insulin from enzymatic degradation. Additionally, CS-DCA-modified NPs promoted the permeability of Caco-2 cells and enhanced intracellular absorption of FITC-labeled insulin by 9.4 and 1.2-folds, when compared to insulin solution and unmodified NPs, respectively. The positively charged NPs increased intestinal villi adhesion and enhanced insulin absorption in the intestines of diabetic rat models. Furthermore, the hypoglycemic test showed that CDA NPs prolonged insulin release in vivo and exerted a remarkable hypoglycemic effect on diabetic rats with an oral bioavailability of 15%. In conclusion, CDA NPs is a potential oral insulin delivery system.


Asunto(s)
Alginatos/administración & dosificación , Quitosano/administración & dosificación , Ácido Desoxicólico/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Insulina/administración & dosificación , Nanopartículas/administración & dosificación , Administración Oral , Alginatos/metabolismo , Animales , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Quitosano/metabolismo , Ácido Desoxicólico/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Concentración de Iones de Hidrógeno , Insulina/metabolismo , Masculino , Nanopartículas/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Biochemistry ; 59(15): 1482-1492, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32266809

RESUMEN

Aggregation of polypeptides and proteins is commonly associated with human and other vertebrate diseases. For example, amyloid plaques consisting of amyloid-ß proteins are frequently identified in Alzheimer's disease and islet amyloid formed by islet amyloid polypeptide (IAPP, amylin) can be found in most patients with type 2 diabetes (T2D). Although many fluorescent dyes have been developed to stain amyloid fibrils, very few examples have been designed for IAPP. In this study, a series of environmentally sensitive fluorescent probes using flavonoid as a scaffold design are rationally designed and synthesized. One of these probes, namely 3-HF-ene-4'-OMe, can bind to IAPP fibrils but not nonfibrillar IAPP by exhibiting a much stronger fluorescent enhancement at 535 nm. In addition, this probe shows better detection sensitivity to IAPP fibrils compared with that of conventionally used thioflavin-T. We demonstrate that 3-HF-ene-4'-OMe can be used to monitor the kinetics of IAPP fibril formation in vitro even in the presence an amyloid inhibitor. To test the specificity of the probe, we attempt to incubate this probe with amyloid fibrils formed from other amyloidogenic proteins. Interestingly, this probe shows different responses when mixed with these fibrils, suggesting the mode of binding of this probe on these fibrils could be different. Moreover, we show that this probe is not toxic to pancreatic mouse ß-cells. Further structural optimization based on the structure of 3-HF-ene-4'-OMe may yield a specific probe for imaging islet amyloid in the pancreas. That would improve our understanding of the relationship between islet amyloid and T2D.


Asunto(s)
Diseño de Fármacos , Flavonoides/química , Colorantes Fluorescentes/química , Polipéptido Amiloide de los Islotes Pancreáticos/química , Animales , Línea Celular Tumoral , Flavonoides/síntesis química , Colorantes Fluorescentes/síntesis química , Humanos , Ratones , Estructura Molecular , Imagen Óptica
10.
FASEB J ; 33(7): 8436-8452, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30995868

RESUMEN

Cardiac aging is characterized by myocardial hypertrophy, fibrosis, and diastolic dysfunction. Human kallikrein (hKLK1) protects against fibrosis in various pathogenic states. However, the effects of hKLK1 overexpression on cardiac aging-related fibrosis and the underlying mechanisms remain unknown. Moreover, the role of hKLK1 in regulating macrophage function leading to cardiac fibrosis has not been investigated. Thus, in this study, we determined the effects of hKLK1 on cardiac aging and explored the mechanisms through which hKLK1 regulated aging-related fibrosis. Echocardiographic measurements showed that aging caused significant alternations in cardiac morphology, hypertrophy, and fibrosis in rats, and hKLK1 overexpression protected against aging-induced cardiac dysfunction. Compared with wild-type hearts, the hKLK1 transgene decreased the expression of monocyte chemoattractant protein 1 and suppressed mitochondrial dysfunction and excess oxidative stress, leading to decreased recruitment and retention of alternatively activated (M2) macrophages and reduced secretion of profibrotic cytokines mediated by the TGF-ß1-Smad3 signaling pathway in hearts of aging rats. Furthermore, these cardioprotective effects of hKLK1 overexpression were associated with the Janus kinase-signal transducer and activator of transcription 3 signaling pathway. H2O2-induced senescence promoted the differentiation of RAW264.7 cells into M2-type cells induced by IL-4 treatment. Bradykinin treatment relieved the migratory capacity of macrophages induced by H2O2. Thus, hKLK1 overexpression reduced cardiac fibrosis and improved aging-related cardiac dysfunction through reduced shift of macrophages to M2 macrophages, indicating that hKLK1 may alleviate aging-related cardiac dysfunction.-Hu, D., Dong, R., Yang, Y., Chen, Z., Tang, Y., Fu, M., Wang, D. W., Xu, X., Tu, L. Human kallikrein overexpression alleviates cardiac aging by alternatively regulating macrophage polarization in aged rats.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/fisiología , Corazón/fisiología , Calicreínas/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiología , Animales , Línea Celular , Fibrosis/metabolismo , Humanos , Activación de Macrófagos/fisiología , Masculino , Ratones , Miocardio/metabolismo , Estrés Oxidativo/fisiología , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Transducción de Señal/fisiología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
11.
Biophys J ; 116(12): 2304-2313, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31155148

RESUMEN

Protein glycation, also known as nonenzymatic glycosylation, is a spontaneous post-translational modification that would change the structure and stability of proteins or hormone peptides. Recent studies have indicated that glycation plays a role in type 2 diabetes (T2D) and neurodegenerative diseases. Over the last two decades, many types of advanced glycation end products (AGEs), formed through the reactions of an amino group of proteins with reducing sugars, have been identified and detected in vivo. However, the effect of glycation on protein aggregation has not been fully investigated. In this study, we aim to elucidate the impact of protein glycation on islet amyloid polypeptide (IAPP, also known as amylin) aggregation, which was strongly associated with T2D. We chemically synthesized glycated IAPP (AGE-IAPP) to mimic the consequence of this hormone peptide in a hyperglycemia (high blood sugar) environment. Our data revealed that AGE-IAPP formed amyloid faster than normal IAPP, and higher-molecular-weight AGE-IAPP oligomers were also observed in the early stage of aggregation. Circular dichroism spectra also indicated that AGE-IAPP exhibited faster conformational changes from random coil to its ß-sheet fibrillar states. Moreover, AGE-IAPP can induce normal IAPP to expedite its aggregation process, and its fibrils can also act as templates to promote IAPP aggregation. AGE-IAPP, like normal IAPP, is capable of interacting with synthetic membranes and also exhibits cytotoxicity. Our studies demonstrated that glycation modification of IAPP promotes the amyloidogenic properties of IAPP, and it may play a role in accumulating additional amyloid during T2D progression.


Asunto(s)
Amiloide/química , Amiloide/metabolismo , Glioxal/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Glicosilación/efectos de los fármacos , Ratones , Peso Molecular , Agregado de Proteínas/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos
12.
Artículo en Inglés | MEDLINE | ID: mdl-30296488

RESUMEN

Macrophages in adipose tissue are associated with obesity-induced low-grade inflammation, which contributed to insulin resistance and the related metabolic diseases. Macrophages display phenotypic plasticity, and polarize under the condition of obesity. Epoxyeicosatrienoic Acids (EETs) are lipid mediators that are involved in the regulation of inflammatory cascades and glucose homeostasis. In this mini-review, we briefly summarize the macrophages recruitment, infiltration and polarization in obese mice, and also discuss the immune-metabolic regulatory role of EETs in this process (See Fig. 1).


Asunto(s)
Ácidos Araquidónicos/inmunología , Inflamación/inmunología , Resistencia a la Insulina/inmunología , Obesidad/inmunología , Adaptación Fisiológica , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Ácidos Araquidónicos/metabolismo , Dieta Alta en Grasa , Humanos , Inflamación/metabolismo , Inflamación/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Obesos , Obesidad/metabolismo
13.
J Am Chem Soc ; 139(46): 16748-16758, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29072444

RESUMEN

Transiently populated oligomers formed en route to amyloid fibrils may constitute the most toxic aggregates associated with many amyloid-associated diseases. Most nucleation theories used to describe amyloid aggregation predict low oligomer concentrations and do not take into account free energy costs that may be associated with structural rearrangements between the oligomer and fiber states. We have used isotope labeling and two-dimensional infrared spectroscopy to spectrally resolve an oligomeric intermediate during the aggregation of the human islet amyloid protein (hIAPP or amylin), the protein associated with type II diabetes. A structural rearrangement includes the F23G24A25I26L27 region of hIAPP, which starts from a random coil structure, evolves into ordered ß-sheet oligomers containing at least 5 strands, and then partially disorders in the fibril structure. The supercritical concentration is measured to be between 150 and 250 µM, which is the thermodynamic parameter that sets the free energy of the oligomers. A 3-state kinetic model fits the experimental data, but only if it includes a concentration independent free energy barrier >3 kcal/mol that represents the free energy cost of refolding the oligomeric intermediate into the structure of the amyloid fibril; i.e., "oligomer activation" is required. The barrier creates a transition state in the free energy landscape that slows fibril formation and creates a stable population of oligomers during the lag phase, even at concentrations below the supercritical concentration. Largely missing in current kinetic models is a link between structure and kinetics. Our experiments and modeling provide evidence that protein structural rearrangements during aggregation impact the populations and kinetics of toxic oligomeric species.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/química , Termodinámica , Diabetes Mellitus Tipo 2 , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/síntesis química , Cinética , Replegamiento Proteico , Factores de Tiempo
14.
Cell Physiol Biochem ; 42(2): 761-779, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28628912

RESUMEN

BACKGROUND/AIMS: Sepsis is a common disease that continues to increase in prevalence worldwide, and diabetes mellitus may make the situation worse. This study was designed to determine the role of Liver Kinase B1 (LKB1)/adenosine monophosphate-activated protein kinase (AMPK) signaling pathway in diabetic mice complicated with systemic endotoxemia. METHODS: The effects of LKB1/AMPK signaling pathway activation on endotoxemia were investigated in streptozotocin induced diabetic mice (STZ-mice) and db/db diabetic mice. Primary peritoneal macrophages and human umbilical vein endothelial cells (HUVECs) monolayers were simultaneously stimulated by both high glucose and LPS and used as a model to investigate the potential molecular mechanisms in vitro. RESULTS: After treatment with LPS, high glucose or both LPS and high glucose, phosphor-AMPK expression was decreased, and moreover, AMPK activation by metformin treatment alleviated the decrease in phosphor-AMPK expression in HUVECs and macrophages as well as in lung tissue. Furthermore, both LPS and high glucose co-treatment decreased LKB1 and phosphor-AMPK expression via enhanced oxidative stress response, and importantly, LKB1 overexpression mediated by adenovirus inhibited the decrease in phosphor-AMPK expression in macrophages and HUVECs. AMPK activation by metformin administration improved the survival of STZ-induced diabetic mice and db/db diabetic mice, which was associated with reduced lung endothelial hyperpermeability and systemic inflammatory response. Furthermore, the permeability of HUVECs monolayers induced by both high glucose and LPS stimulation was also alleviated by AMPK activation, which was partly via suppression of VE-cadherin phosphorylation. CONCLUSION: These data demonstrated that LKB1/AMPK signaling pathway activation improved the survival of diabetic mice complicated with endotoxemia. Thus, LKB1/AMPK signaling pathway may serve as a potentially useful therapeutic target for severe infection in diabetic patients.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Diabetes Mellitus Experimental/genética , Endotoxemia/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Progresión de la Enfermedad , Endotoxemia/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipoglucemiantes/administración & dosificación , Hígado/metabolismo , Hígado/patología , Metformina/administración & dosificación , Ratones , Ratones Endogámicos NOD/genética , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
J Biol Chem ; 290(51): 30475-85, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26483547

RESUMEN

Deposition of human islet amyloid polypeptide (hIAPP, also known as amylin) as islet amyloid is a characteristic feature of the pancreas in type 2 diabetes, contributing to increased ß-cell apoptosis and reduced ß-cell mass. Matrix metalloproteinase-9 (MMP-9) is active in islets and cleaves hIAPP. We investigated whether hIAPP fragments arising from MMP-9 cleavage retain the potential to aggregate and cause toxicity, and whether overexpressing MMP-9 in amyloid-prone islets reduces amyloid burden and the resulting ß-cell toxicity. Synthetic hIAPP was incubated with MMP-9 and the major hIAPP fragments observed by MS comprised residues 1-15, 1-25, 16-37, 16-25, and 26-37. The fragments 1-15, 1-25, and 26-37 did not form amyloid fibrils in vitro and they were not cytotoxic when incubated with ß cells. Mixtures of these fragments with full-length hIAPP did not modulate the kinetics of fibril formation by full-length hIAPP. In contrast, the 16-37 fragment formed fibrils more rapidly than full-length hIAPP but was less cytotoxic. Co-incubation of MMP-9 and fragment 16-37 ablated amyloidogenicity, suggesting that MMP-9 cleaves hIAPP 16-37 into non-amyloidogenic fragments. Consistent with MMP-9 cleavage resulting in largely non-amyloidogenic degradation products, adenoviral overexpression of MMP-9 in amyloid-prone islets reduced amyloid deposition and ß-cell apoptosis. These findings suggest that increasing islet MMP-9 activity might be a strategy to limit ß-cell loss in type 2 diabetes.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/enzimología , Polipéptido Amiloide de los Islotes Pancreáticos/toxicidad , Metaloproteinasa 9 de la Matriz/metabolismo , Péptidos/toxicidad , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Humanos , Células Secretoras de Insulina/patología , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Transgénicos
16.
Cell Physiol Biochem ; 38(5): 1928-38, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27160852

RESUMEN

UNLABELLED: Backgroud: Myocardial fibrosis results in myocardial remodelling and dysfunction. Celastrol, a traditional oriental medicine, has been suggested to have cardioprotective effects. However, its underlying mechanism is unknown. This study investigated the ability of celastrol to prevent cardiac fibrosis and dysfunction and explored the underlying mechanisms. METHODS: Animal and cell models of cardiac fibrosis were used in this study. Myocardial fibrosis was induced by transverse aortic constriction (TAC) in mice. Cardiac hypertrophy and fibrosis were evaluated based on histological and biochemical measurements. Cardiac function was evaluated by echocardiography. The levels of transforming growth factor beta 1 (TGF-ß1), extracellular signal regulated kinases 1/2 (ERK1/2) signalling were measured using Western blotting, while the expression of miR-21was analyzed by real-time qRT-PCR in vitro and in vivo. In vitro studies, cultured cardiac fibroblasts (CFs) were treated with TGF-ß1 and transfected with microRNA-21(miR21). RESULTS: Celastrol treatment reduced the increased collagen deposition and down-regulated α-smooth muscle actin (α-SMA), atrial natriuretic peptide (ANP), brain natriuretic peptides (BNP), beta-myosin heavy chain (ß-MHC), miR-21 and p-ERK/ERK. Cardiac dysfunction was significantly attenuated by celastrol treatment in the TAC mice model. Celastrol treatment reduced myocardial fibroblast viability and collagen content and down-regulated α-SMA in cultured CFs in vitro. Celastrol also inhibited the miR-21/ERK signalling pathway. Celastrol attenuated miR-21 up-regulation by TGF-ß1 and decreased elevated p-ERK/ERK levels in CFs transfected with miR-21. CONCLUSION: MiR-21/ERK signalling could be a potential therapeutic pathway for the prevention of myocardial fibrosis. Celastrol ameliorates myocardial fibrosis and cardiac dysfunction, these probably related to miR-21/ERK signaling pathways in vitro and in vivo.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , MicroARNs/metabolismo , Miocardio/patología , Triterpenos/farmacología , Actinas/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Cardiomegalia/patología , Supervivencia Celular/efectos de los fármacos , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Ventrículos Cardíacos/diagnóstico por imagen , Masculino , Ratones , MicroARNs/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Péptido Natriurético Encefálico/metabolismo , Triterpenos Pentacíclicos , Fosforilación/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Triterpenos/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos
17.
Proc Natl Acad Sci U S A ; 110(48): 19279-84, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24218607

RESUMEN

Islet amyloid polypeptide (IAPP) is responsible for amyloid formation in type 2 diabetes and contributes to the failure of islet cell transplants, however the mechanisms of IAPP-induced cytotoxicity are not known. Interactions with model anionic membranes are known to catalyze IAPP amyloid formation in vitro. Human IAPP damages anionic membranes, promoting vesicle leakage, but the features that control IAPP-membrane interactions and the connection with cellular toxicity are not clear. Kinetic studies with wild-type IAPP and IAPP mutants demonstrate that membrane leakage is induced by prefibrillar IAPP species and continues over the course of amyloid formation, correlating additional membrane disruption with fibril growth. Analyses of a set of designed mutants reveal that membrane leakage does not require the formation of ß-sheet or α-helical structures. A His-18 to Arg substitution enhances leakage, whereas replacement of all of the aromatic residues via a triple leucine mutant has no effect. Biophysical measurements in conjunction with cytotoxicity studies show that nonamyloidogenic rat IAPP is as effective as human IAPP at disrupting standard anionic model membranes under conditions where rat IAPP does not induce cellular toxicity. Similar results are obtained with more complex model membranes, including ternary systems that contain cholesterol and are capable of forming lipid rafts. A designed point mutant, I26P-IAPP; a designed double mutant, G24P, I26P-IAPP; a double N-methylated variant; and pramlintide, a US Food and Drug Administration-approved IAPP variant all induce membrane leakage, but are not cytotoxic, showing that there is no one-to-one relationship between disruption of model membranes and induction of cellular toxicity.


Asunto(s)
Amiloide/biosíntesis , Diabetes Mellitus Tipo 2/fisiopatología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Membranas Artificiales , Secuencia de Aminoácidos , Animales , Benzotiazoles , Biofisica , Humanos , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Oxazinas , Ratas , Especificidad de la Especie , Tiazoles , Xantenos
18.
BMC Complement Altern Med ; 16: 241, 2016 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-27457615

RESUMEN

BACKGROUND: We previously demonstrated oxymatrine, an alkaloid from the Chinese medicine radix Sophorae flavescentis, ameliorates hemodynamic disturbances and cardiac fibrosis; however, the underlying mechanisms are unclear. Here, we investigated the effect and mechanism of action of oxymatrine on aldosterone-induced cardiac fibroblast to myofibroblast differentiation in vitro. METHODS: Cardiac fibroblasts were isolated purified from neonatal Sprague Dawley rats. The optimal concentration of aldosterone to stimulate cardiac fibroblast proliferation was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cardiac fibroblasts were pretreated with 7.57 × 10(-4) mol/L or 3.78 × 10(-4) mol/L oxymatrine or without oxymatrine for 2 h, and then coincubated with 1 × 10(-8) mol/L aldosterone for 48 h. The MTT assay and Masson staining were used to detect the cardiac fibroblast proliferation and myofibroblast differentiation. The secretion of type I and III collagen was measured by commercial ELISA kits, and the hydroxyproline content was determined by the colorimetric assay. Western blotting assayed the Smad-2, Smad-3, and Smad-4 protein expression in cardiac fibroblasts. RESULTS: The present results confirmed that aldosterone induced cardiac fibroblast to myofibroblast proliferation and differentiation. The MTT assay and Masson staining indicated oxymatrine significantly inhibited aldosterone-induced cardiac fibroblast proliferation and myofibroblast differentiation. Oxymatrine significantly inhibited aldosterone-induced secretion of type I and III collagen, as indicated by commercial ELISA kits, and aldosterone-induced increase in hydroxyproline content, as indicated by a colorimetric assay. Western blotting revealed oxymatrine attenuated aldosterone-induced Smad-2, Smad-3, and Smad-4 expression in cardiac fibroblasts. CONCLUSION: Oxymatrine can inhibit cardiac fibroblast proliferation and differentiation into myofibroblasts via a mechanism linked to attenuation of the Smad signaling pathway.


Asunto(s)
Aldosterona/farmacología , Alcaloides/farmacología , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Sustancias Protectoras/farmacología , Quinolizinas/farmacología , Proteínas Smad/metabolismo , Alcaloides/química , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Miocardio/citología , Sustancias Protectoras/química , Quinolizinas/química , Ratas , Ratas Sprague-Dawley , Proteínas Smad/análisis , Proteínas Smad/genética
19.
Biochemistry ; 54(3): 666-76, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25531836

RESUMEN

The process of amyloid formation by the normally soluble hormone islet amyloid polypeptide (IAPP) contributes to ß-cell death in type 2 diabetes and in islet transplants. There are no clinically approved inhibitors of islet amyloidosis, and the mode of action of existing inhibitors is not well-understood. Resveratrol, a natural polyphenol, has been reported to inhibit amyloid formation by IAPP and by the Alzheimer's disease Aß peptide. The mechanism of action of this compound is not known, nor is its mode of interaction with IAPP. In this study, we use a series of IAPP variants to examine possible interactions between resveratrol and IAPP. Fluorescence assays, transmission electron microscopy, and mass spectrometry demonstrate that resveratrol is much less effective as an inhibitor of IAPP amyloid formation than the polyphenol (-)-epigallocatechin 3-gallate (EGCG) and, unlike EGCG, does not significantly disaggregate preformed IAPP amyloid fibrils. Resveratrol is also shown to interfere with thioflavin-T assays. His-18 mutants, a truncation mutant, mutants of each of the aromatic residues, and mutants of Arg-11 of IAPP were examined. Mutation of His to Gln or Leu weakens the ability of resveratrol to inhibit amyloid formation by IAPP, as do mutations of Arg-11, Phe-15, or Tyr-37 to Leu, and truncation to form the variant Ac 8-37-IAPP, which removes the first seven residues to eliminate Lys-1 and the N-terminal amino group. In contrast, replacement of Phe-23 with Leu has a smaller effect. The data highlight Phe-15, His-18, and Tyr-37 as being important for IAPP-resveratrol interactions and are consistent with a potential role of the N-terminus and Arg-11 in polypeptide-resveratrol interactions.


Asunto(s)
Aminoácidos Aromáticos/metabolismo , Amiloide/metabolismo , Histidina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Estilbenos/farmacología , Secuencia de Aminoácidos , Amiloide/ultraestructura , Arginina/metabolismo , Benzotiazoles , Catequina/análogos & derivados , Catequina/farmacología , Análisis Mutacional de ADN , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/ultraestructura , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura , Resveratrol , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad , Tiazoles/metabolismo
20.
Cell Physiol Biochem ; 37(4): 1592-606, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26517864

RESUMEN

BACKGROUND/AIMS: Bradykinin has been shown to exert a variety of protective effects against vascular injury, and to reduce the levels of several factors involved in the coagulation cascade. A key determinant of thrombin generation is tissue factor (TF). However, whether bradykinin can regulate TF expression remains to be investigated. METHODS: To study the effect of bradykinin on TF expression, we used Lipopolysaccharides (LPS) to induce TF expression in human umbilical vein endothelial cells and monocytes. Transcript levels were determined by RT-PCR, protein abundance by Western blotting. In the in vivo study, bradykinin and equal saline were intraperitoneally injected into mice for three days ahead of inferior cava vein ligation that we took to induce thrombus formation, after which bradykinin and saline were injected for another two days. Eventually, the mice were sacrificed and tissues were harvested for tests. RESULTS: Exogenous bradykinin markedly inhibited TF expression in mRNA and protein level induced by LPS in a dose-dependent manner. Moreover, the NO synthase antagonist L-NAME and PI3K inhibitor LY294002 dramatically abolished the inhibitory effects of bradykinin on tissue factor expression. PI3K/Akt signaling pathway activation induced by bradykinin administration reduced the activity of GSK-3ß and MAPK, and reduced NF-x03BA;B level in the nucleus, thereby inhibiting TF expression. Consistent with this, intraperitoneal injection of C57/BL6 mice with bradykinin also inhibited the thrombus formation induced by ligation of inferior vena cava. CONCLUSION: Bradykinin suppressed TF protein expression in human umbilical vein endothelial cells and monocytes in vitro; in line with this, it inhibits thrombus formation induced by ligation of inferior vena cava in vivo.


Asunto(s)
Bradiquinina/farmacología , Transducción de Señal/efectos de los fármacos , Tromboplastina/metabolismo , Trombosis de la Vena/patología , Animales , Cromonas/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Morfolinas/farmacología , FN-kappa B/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Receptor de Bradiquinina B2/metabolismo , Tromboplastina/antagonistas & inhibidores , Tromboplastina/genética , Trombosis de la Vena/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA