Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Pharmacol Res ; 204: 107207, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38734193

RESUMEN

In recent years several experimental observations demonstrated that the gut microbiome plays a role in regulating positively or negatively metabolic homeostasis. Indole-3-propionic acid (IPA), a Tryptophan catabolic product mainly produced by C. Sporogenes, has been recently shown to exert either favorable or unfavorable effects in the context of metabolic and cardiovascular diseases. We performed a study to delineate clinical and multiomics characteristics of human subjects characterized by low and high IPA levels. Subjects with low IPA blood levels showed insulin resistance, overweight, low-grade inflammation, and features of metabolic syndrome compared to those with high IPA. Metabolomics analysis revealed that IPA was negatively correlated with leucine, isoleucine, and valine metabolism. Transcriptomics analysis in colon tissue revealed the enrichment of several signaling, regulatory, and metabolic processes. Metagenomics revealed several OTU of ruminococcus, alistipes, blautia, butyrivibrio and akkermansia were significantly enriched in highIPA group while in lowIPA group Escherichia-Shigella, megasphera, and Desulfovibrio genus were more abundant. Next, we tested the hypothesis that treatment with IPA in a mouse model may recapitulate the observations of human subjects, at least in part. We found that a short treatment with IPA (4 days at 20/mg/kg) improved glucose tolerance and Akt phosphorylation in the skeletal muscle level, while regulating blood BCAA levels and gene expression in colon tissue, all consistent with results observed in human subjects stratified for IPA levels. Our results suggest that treatment with IPA may be considered a potential strategy to improve insulin resistance in subjects with dysbiosis.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Masculino , Animales , Femenino , Persona de Mediana Edad , Resistencia a la Insulina , Indoles , Ratones Endogámicos C57BL , Metabolómica , Ratones , Adulto , Síndrome Metabólico/sangre , Síndrome Metabólico/metabolismo , Síndrome Metabólico/microbiología , Comorbilidad , Músculo Esquelético/metabolismo , Músculo Esquelético/microbiología , Multiómica
2.
Acta Diabetol ; 60(11): 1441-1448, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37401947

RESUMEN

Hyperglycemia strongly affects endothelial function and activation, which in turn increases the risk of atherosclerotic cardiovascular disease. Among pharmacotherapies aimed at lowering blood glucose levels, glucagon-like peptide 1 receptor agonists (GLP-1RA) represent a class of drugs involved in the improvement of the endothelium damage and the progression of cardiovascular diseases. They show antihypertensive and antiatherosclerotic actions due at least in part to direct favorable actions on the coronary vascular endothelium, such as oxidative stress reduction and nitric oxide increase. However, cumulative peripheral indirect actions could also contribute to the antiatherosclerotic functions of GLP-1/GLP-1R agonists, including metabolism and gut microbiome regulation. Therefore, further research is necessary to clarify the specific role of this drug class in the management of cardiovascular disease and to identify specific cellular targets involved in the protective signal transduction. In the present review, we provide an overview of the effects of GLP-1RAs treatment on cardiovascular disease with particular attention on potential molecular mechanisms involving endothelium function on formation and progression of atherosclerotic plaque.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Humanos , Hipoglucemiantes/uso terapéutico , Endotelio Vascular/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas
3.
Front Endocrinol (Lausanne) ; 14: 1297847, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38313841

RESUMEN

Introduction: Type 1 diabetes mellitus (T1DM) development involves a complex interplay of genetic, environmental, and immunological factors. By modulating the activity of proteases and receptors, the protein tissue inhibitor of metalloproteinase 3 (TIMP3) plays a role in limiting the expression and function of pro-inflammatory cytokines, which have been implicated in the advancement of T1DM. This study was aimed at examining the effect of TIMP3 overexpression in myeloid cells on the development of T1DM. Methods and results: Twelve weeks after multiple low doses of streptozotocin (MLDS) treatment, diabetic mice overexpressing TIMP3 specifically in myeloid cells under the CD68 promoter (MacT3 mice) showed improved insulin secretion, islet morphology and vascularization, antioxidant defense system, and regulatory factors of mitochondrial biosynthesis and function. To get mechanistic insights into the origin of this protection, the severity of insulitis and inflammatory parameters were evaluated in pancreatic tissues 11 days after MLSD treatment, showing significantly reduced insulitis and levels of the pro-inflammatory cytokine tumor necrosis factor-α, interleukin -1ß, and interferon -γ in MacT3 mice. Discussion: The results indicate that TIMP3 is involved in maintaining islet architecture and functions, at least in part, through modulation of pro-inflammatory cytokine production associated with insulitis and may represent a novel therapeutic strategy for T1DM.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Inhibidor Tisular de Metaloproteinasa-3 , Animales , Ratones , Linaje de la Célula , Citocinas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Interferón gamma , Hormonas Pancreáticas , Estreptozocina , Inhibidor Tisular de Metaloproteinasa-3/genética
4.
Mol Metab ; 59: 101454, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35150905

RESUMEN

OBJECTIVE: Metabolic syndrome, obesity, and steatosis are characterized by a range of dysregulations including defects in ubiquitin ligase tagging proteins for degradation. The identification of novel hepatic genes associated with fatty liver disease and metabolic dysregulation may be relevant to unravelling new mechanisms involved in liver disease progression METHODS: Through integrative analysis of liver transcriptomic and metabolomic obtained from obese subjects with steatosis, we identified itchy E ubiquitin protein ligase (ITCH) as a gene downregulated in human hepatic tissue in relation to steatosis grade. Wild-type or ITCH knockout mouse models of non-alcoholic fatty liver disease (NAFLD) and obesity-related hepatocellular carcinoma were analyzed to dissect the causal role of ITCH in steatosis RESULTS: We show that ITCH regulation of branched-chain amino acids (BCAAs) degradation enzymes is impaired in obese women with grade 3 compared with grade 0 steatosis, and that ITCH acts as a gatekeeper whose loss results in elevation of circulating BCAAs associated with hepatic steatosis. When ITCH expression was specifically restored in the liver of ITCH knockout mice, ACADSB mRNA and protein are restored, and BCAA levels are normalized both in liver and plasma CONCLUSIONS: Our data support a novel functional role for ITCH in the hepatic regulation of BCAA metabolism and suggest that targeting ITCH in a liver-specific manner might help delay the progression of metabolic hepatic diseases and insulin resistance.


Asunto(s)
Aminoácidos de Cadena Ramificada , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Obesidad , Ubiquitina-Proteína Ligasas , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones Noqueados , Obesidad/complicaciones , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
5.
Diagnostics (Basel) ; 11(11)2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34829465

RESUMEN

BACKGROUND: The aim of this study was to evaluate how the high sensitivity C-reactive protein (hs-CRP) values influence the risk of carotid plaque instability in association with other cardiovascular risk factors. METHODS: One hundred and fifty-six carotid plaques from both symptomatic and asymptomatic patients requiring surgical carotid endarterectomy were retrospectively collected. According to the modified American Heart Association, atherosclerosis plaques have been histologically distinguished into unstable and stable. The following anamnestic and hematochemical data were also considered: age, gender, hypertension, diabetes mellitus, smoking habit, therapy, low-density lipoprotein (LDL)-C, kidney failure and hs-CRP. RESULTS: The results of our study clearly show that high levels of hs-CRP significantly increase the carotid plaque instability in dyslipidemic patients. Specifically, a 67% increase of the risk of carotid plaque instability was observed in patients with high LDL-C. Therefore, the highest risk was observed in male dyslipidemic patients 2333 (95% CI 0.73-7.48) and in aged female patients 2713 (95% CI 0.14-53.27). DISCUSSION: These data strongly suggest a biological relationship between the hs-CRP values and the alteration of lipidic metabolism mostly in male patients affected by carotid atherosclerosis. The measurement of hs-CRP might be useful as a potential screening tool in the prevention of atheroscletotic disease.

6.
Br J Pharmacol ; 178(24): 4891-4906, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34411281

RESUMEN

BACKGROUND AND PURPOSE: Amyotrophic lateral sclerosis (ALS) patients exhibit dysfunctional energy metabolism and weight loss, which is negatively correlated with survival, together with neuroinflammation. However, the possible contribution of neuroinflammation to deregulations of feeding behaviour in ALS has not been studied in detail. We here investigated if microglial KCa 3.1 is linked to hypothalamic neuroinflammation and affects feeding behaviours in ALS mouse models. EXPERIMENTAL APPROACH: hSOD1G93A and TDP43A315T mice were treated daily with 120 mg·kg-1 of TRAM-34 or vehicle by intraperitoneal injection from the presymptomatic until the disease onset phase. Body weight and food intake were measured weekly. The later by weighing food provided minus that left in the cage. RT-PCR and immunofluorescence analysis were used to characterize microglia phenotype and the main populations of melanocortin neurons in the hypothalamus of hSOD1G93A and age-matched non-tg mice. The cannabinoid-opioid interactions in feeding behaviour of hSOD1G93A mice were studied using an inverse agonist and an antagonist of the cannabinoid receptor CB1 (rimonabant) and µ-opioid receptors (naloxone), respectively. KEY RESULTS: We found that treatment of hSOD1G93A mice with the KCa 3.1 inhibitor TRAM-34 (i), attenuates the pro-inflammatory phenotype of hypothalamic microglia, (ii) increases food intake and promotes weight gain, (iii) increases the number of healthy pro-opiomelanocortin (POMC) neurons and (iv), changes the expression of cannabinoid receptors involved in energy homeostasis. CONCLUSION AND IMPLICATIONS: Using ALS mouse models, we describe defects in the hypothalamic melanocortin system that affect appetite control. These results reveal a new regulatory role for KCa 3.1 to counteract weight loss in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Conducta Alimentaria , Canales de Potasio Calcio-Activados/metabolismo , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Modelos Animales de Enfermedad , Metabolismo Energético , Homeostasis , Melanocortinas , Ratones , Ratones Transgénicos , Microglía/citología , Pirazoles/farmacología , Receptores de Cannabinoides , Médula Espinal/metabolismo , Superóxido Dismutasa-1/metabolismo , Aumento de Peso
7.
Acta Diabetol ; 58(12): 1587-1594, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34181080

RESUMEN

Diabetic kidney disease, one of the most severe complications associated with diabetes, is characterized by albuminuria, glomerulosclerosis and progressive loss of renal function. Loss of TIMP3, an Extracellular matrix-bound protein, is a hallmark of diabetic nephropathy in human and mouse models, suggesting its pivotal role in renal diseases associated to diabetes. There is currently no specific therapy for diabetic nephropathy, and the ability to restore high TIMP3 activity specifically in the kidney may represent a potential therapeutic strategy for the amelioration of renal injury under conditions in which its reduction is directly related to the disease. Increasing evidence shows that diabetic nephropathy is also regulated by epigenetic mechanisms, including noncoding RNA. This review recapitulates the pathological, diagnostic and therapeutic potential roles of TIMP3 and the noncoding RNA (microRNA, long noncoding RNA) related to its expression, in the progression of diabetic nephropathy.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , MicroARNs , Albuminuria , Animales , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/genética , Humanos , Riñón , Ratones , Pronóstico , Inhibidor Tisular de Metaloproteinasa-3
8.
J Clin Endocrinol Metab ; 106(11): e4350-e4359, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34192323

RESUMEN

CONTEXT: Type 2 diabetes (T2D) shows a high mortality rate, partly mediated by atherosclerotic plaque instability. Discovering novel biomarkers may help identify high-risk patients who would benefit from more aggressive and specific managements. We recently described a serum resistin and multicytokine inflammatory pathway (REMAP), including resistin, interleukin (IL)-1ß, IL-6, IL-8, and TNF-α, that is associated with cardiovascular disease. OBJECTIVE: We investigated whether REMAP is associated with and improves the prediction of mortality in T2D. METHODS: A REMAP score was investigated in 3 cohorts comprising 1528 patients with T2D (409 incident deaths) and in 59 patients who underwent carotid endarterectomy (CEA; 24 deaths). Plaques were classified as unstable/stable according to the modified American Heart Association atherosclerosis classification. RESULTS: REMAP was associated with all-cause mortality in each cohort and in all 1528 individuals (fully adjusted hazard ratio [HR] for 1 SD increase = 1.34, P < .001). In CEA patients, REMAP was associated with mortality (HR = 1.64, P = .04) and a modest change was observed when plaque stability was taken into account (HR = 1.58; P = .07). REMAP improved discrimination and reclassification measures of both Estimation of Mortality Risk in Type 2 Diabetic Patients and Risk Equations for Complications of Type 2 Diabetes, well-established prediction models of mortality in T2D (P < .05-< .001). CONCLUSION: REMAP is independently associated with and improves predict all-cause mortality in T2D; it can therefore be used to identify high-risk individuals to be targeted with more aggressive management. Whether REMAP can also identify patients who are more responsive to IL-6 and IL-1ß monoclonal antibodies that reduce cardiovascular burden and total mortality is an intriguing possibility to be tested.


Asunto(s)
Citocinas/sangre , Complicaciones de la Diabetes/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/mortalidad , Inflamación/sangre , Resistina/sangre , Anciano , Aterosclerosis/sangre , Aterosclerosis/complicaciones , Aterosclerosis/terapia , Biomarcadores/sangre , Estudios de Cohortes , Complicaciones de la Diabetes/terapia , Diabetes Mellitus Tipo 2/terapia , Femenino , Humanos , Inflamación/complicaciones , Interleucinas/sangre , Masculino , Persona de Mediana Edad , Placa Aterosclerótica/sangre , Placa Aterosclerótica/etiología , Placa Aterosclerótica/patología , Estudios Prospectivos , Factores de Riesgo , Factor de Necrosis Tumoral alfa/sangre
9.
Int J Mol Sci ; 22(10)2021 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-34068941

RESUMEN

Diabetic nephropathy (DN) is the most frequent cause of end-stage renal disease. Tubulointerstitial accumulation of lysine 63 (K63)-ubiquitinated (Ub) proteins is involved in the progression of DN fibrosis and correlates with urinary miR-27b-3p downregulation. We explored the renoprotective effect of an inhibitor of K63-Ub (NSC697923), alone or in combination with the ACE-inhibitor ramipril, in vitro and in vivo. Proximal tubular epithelial cells and diabetic DBA/2J mice were treated with NSC697923 and/or ramipril. K63-Ub protein accumulation along with α-SMA, collagen I and III, FSP-1, vimentin, p16INK4A expression, SA-α Gal staining, Sirius Red, and PAS staining were measured. Finally, we measured the urinary albumin to creatinine ratio (uACR), and urinary miR-27b-3p expression in mice. NSC697923, both alone and in association with ramipril, in vitro and in vivo inhibited hyperglycemia-induced epithelial to mesenchymal transition by significantly reducing K63-Ub proteins, α-SMA, collagen I, vimentin, FSP-1 expression, and collagen III along with tubulointerstitial and glomerular fibrosis. Treated mice also showed recovery of urinary miR-27b-3p and restored expression of p16INK4A. Moreover, NSC697923 in combination with ramipril demonstrated a trend in the reduction of uACR. In conclusion, we suggest that selective inhibition of K63-Ub, when combined with the conventional treatment with ACE inhibitors, might represent a novel treatment strategy to prevent the progression of fibrosis and proteinuria in diabetic nephropathy and we propose miR-27b-3p as a biomarker of treatment efficacy.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/prevención & control , Fibrosis/prevención & control , Lisina/química , Nitrofuranos/farmacología , Ramipril/farmacología , Sulfonas/farmacología , Ubiquitinación , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Quimioterapia Combinada , Femenino , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Ratones , Ratones Endogámicos DBA
10.
Clin Transl Med ; 11(2): e305, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33634991

RESUMEN

BACKGROUND: Diabetic nephropathy (DN), one of the major complications of diabetes, is characterized by albuminuria, glomerulosclerosis, and progressive loss of renal function. Loss of TIMP3, an Extracellular Matrix bound protein affecting both inflammation and fibrosis, is a hallmark of DN in human subjects and mouse models. METHODS: This study was designed to provide evidences that the modulation of the system involving TIMP3 and its target A Disintegrin And Metalloproteinase 17 (ADAM17), may rescue kidney pathology in diabetic mice. Mice with cell-targeted overexpression of TIMP3 in myeloid cells (MacT3), podocyte-specific ADAM17 knockout mice (∆PodA17), and DBA/2J mice, were rendered diabetic at 8 weeks of age with a low-dose streptozotocin protocol. DBA/2J mice were administered new peptides based on the human TIMP3 N-terminal domain, specifically conjugated with G3C12, a carrier peptide highly selective and efficient for transport to the kidney. Twelve weeks after Streptozotocin injections, 24-hour albuminuria was determined by ELISA, kidney morphometry was analyzed by periodic acid-shift staining, and Real Time-PCR and western blot analysis were performed on mRNA and protein extracted from kidney cortex. RESULTS: Our results showed that both genetic modifications and peptides treatment positively affect renal function and structure in diabetic mice, as indicated by a significant and consistent decline in albuminuria along with reduction in glomerular lesions, as indicated by reduced mesangial expansion and glomerular hypertrophy, decreased deposition of extracellular matrix in the mesangium, diminished protein expression of the NADPH oxidases 4 (NOX4), and the improvement of podocyte structural markers such as WT1, nephrin, and podocin. Moreover, the positive effects were exerted through a mechanism independent from glycemic control. CONCLUSIONS: In diabetic mice the targeting of TIMP3 system improved kidney structure and function, representing a valid approach to develop new avenues to treat this severe complication of diabetes.


Asunto(s)
Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/prevención & control , Expresión Génica/genética , Riñón/efectos de los fármacos , Inhibidor Tisular de Metaloproteinasa-3/genética , Animales , Antibióticos Antineoplásicos/administración & dosificación , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Estreptozocina/administración & dosificación
11.
Cell Death Dis ; 11(3): 165, 2020 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127514

RESUMEN

Since online publication of this article, the authors noticed that there was a basic citation error in PubMed citation data. Specifically, the name of the author "Piergiorgio La Rosa" is cited as "Rosa P" in the PubMed citation, when it should be "La Rosa P", "La Rosa" being the surname and "Piergiorgio" the name of the author.

12.
Cell Death Dis ; 11(1): 51, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974344

RESUMEN

Decreased expression of mitochondrial frataxin (FXN) causes Friedreich's ataxia (FRDA), a neurodegenerative disease with type 2 diabetes (T2D) as severe comorbidity. Brown adipose tissue (BAT) is a mitochondria-enriched and anti-diabetic tissue that turns excess energy into heat to maintain metabolic homeostasis. Here we report that the FXN knock-in/knock-out (KIKO) mouse shows hyperlipidemia, reduced energy expenditure and insulin sensitivity, and elevated plasma leptin, recapitulating T2D-like signatures. FXN deficiency leads to disrupted mitochondrial ultrastructure and oxygen consumption as well as lipid accumulation in BAT. Transcriptomic data highlights cold intolerance in association with iron-mediated cell death (ferroptosis). Impaired PKA-mediated lipolysis and expression of genes controlling mitochondrial metabolism, lipid catabolism and adipogenesis were observed in BAT of KIKO mice as well as in FXN-deficient T37i brown and primary adipocytes. Significant susceptibility to ferroptosis was observed in adipocyte precursors that showed increased lipid peroxidation and decreased glutathione peroxidase 4. Collectively our data point to BAT dysfunction in FRDA and suggest BAT as promising therapeutic target to overcome T2D in FRDA.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Ataxia de Friedreich/metabolismo , Proteínas de Unión a Hierro/metabolismo , Metabolismo de los Lípidos , Mitocondrias/metabolismo , Termogénesis/genética , Adipocitos/metabolismo , Tejido Adiposo Pardo/ultraestructura , Animales , Frío , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ferroptosis/genética , Ataxia de Friedreich/genética , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Resistencia a la Insulina/genética , Proteínas de Unión a Hierro/genética , Leptina/sangre , Lipólisis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Estrés Oxidativo/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , RNA-Seq , Frataxina
13.
Int J Biochem Cell Biol ; 118: 105659, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31765819

RESUMEN

The O subfamily of forkhead (FoxO) 1 is a crucial regulator of cell metabolism in several tissues, including the heart, where it is involved in cardiac regulation of glucose and lipid metabolic pathways, and endothelium, controlling the levels of some relevant biomarkers in atherosclerotic process. Despite the growing understanding of FoxO1 biology, the metabolic consequences of FoxO1 modifications and its implication in CVD, atherosclerosis and T2DM are still not incompletely described. In this review we discuss how FoxO1 affects cardiovascular pathophysiology and which of its effects should be restrained or enhanced to preserve endothelial and heart functions.


Asunto(s)
Aterosclerosis/genética , Enfermedades Cardiovasculares/genética , Diabetes Mellitus Tipo 2/genética , Proteína Forkhead Box O1/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Proteína Forkhead Box O1/metabolismo , Glucosa/genética , Glucosa/metabolismo , Corazón/fisiopatología , Humanos , Metabolismo de los Lípidos/genética , Miocardio/metabolismo , Transducción de Señal/genética
14.
Acta Diabetol ; 56(12): 1265-1274, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31292722

RESUMEN

AIM: Obesity and low-grade inflammation are associated with an increased risk of hepatocellular carcinoma (HCC), a leading cause of cancer-related death worldwide. The tissue inhibitor of metalloproteinase (TIMP) 3, an endogenous inhibitor of protease activity that represents a key mediator of inflammation, is reduced in inflammatory metabolic disorders and cancer. In contrast, Timp3-deficient mice (Timp3-/-) are highly resistant to developing HCC in response to a diethylnitrosamine (DEN); therefore, we aimed to elucidate the biological role of genetic loss of Timp3 in obesity-related hepatocarcinogenesis. METHODS: Fourteen-day-old male wild-type (wt) and Timp3-/- mice were injected with 25 mg/kg DEN or an equal volume of saline. After 4 weeks, mice were randomized into two dietary groups and fed either normal or high-fat diet and allowed to grow until 32 weeks of age. Liver histological features were analyzed, and differentially expressed genes in the liver were quantified. RESULTS: In Timp3-/- mice fed with the obesogenic diet, despite the increase in liver steatosis and inflammation, both the number of tumors and the total tumor size are significantly reduced 30 weeks post-DEN injection, compared to control mice. Moreover, Timp3 deletion in hepatocarcinogenesis during obesity is associated with a reduction in FoxM1 transcriptional activity through H19/miR-675/p53 pathway. CONCLUSIONS: This study suggests that Timp3 ablation leads to cell cycle perturbation, at least in part by repressing FoxM1 transcriptional activity through H19/miR-675/p53 pathway.


Asunto(s)
Carcinoma Hepatocelular/patología , Dieta Alta en Grasa/efectos adversos , Neoplasias Hepáticas/patología , Obesidad/etiología , Inhibidor Tisular de Metaloproteinasa-3/genética , Animales , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Dietilnitrosamina , Progresión de la Enfermedad , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/patología , Proteína Forkhead Box M1/metabolismo , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/patología , Transducción de Señal/genética
15.
Nanomedicine ; 17: 276-286, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30708054

RESUMEN

Gold nanoparticles functionalized with 3-mercapto-1-propansulfonate (AuNPs-3MPS) have been prepared and loaded with Methotrexate (MTX), an immunosuppressive agent used in the systemic treatment of moderate-severe inflammatory diseases. The effects of the AuNPs-3MPS@MTX topically administered in vitro on skin model and in vivo on imiquimod-induced psoriasis-like mice model, have been studied. Clinical response, epidermal thickness, cell proliferation rate and inflammation were tested. AuNPs-3MPS@MTX treated mice showed a decreasing of scaling and erythema score, reduction of epidermal thickness, parakeratosis and hyperkeratosis, compared to AuNPs-3MPS treated mice. Immunohistochemistry analysis staining displayed that Ki67, K6 CD3 and CD8 stainings were reduced in AuNPs-3MPS@MTX treated mice. Blood evaluation showed no differences in blood count and in ALT and AST levels before and after AuNPs-3MPS or AuNPs-3MPS@MTX treatment. Topical AuNPs-3MPS@MTX treatment is able to induce a reduction of keratinocytes hyperproliferation, epidermal thickness and also inflammatory infiltrate in vivo on imiquimod-induced psoriasis like mice model.


Asunto(s)
Portadores de Fármacos/química , Oro/química , Inmunosupresores/administración & dosificación , Metotrexato/administración & dosificación , Psoriasis/tratamiento farmacológico , Compuestos de Sulfhidrilo/química , Administración Tópica , Animales , Modelos Animales de Enfermedad , Inmunosupresores/uso terapéutico , Nanopartículas del Metal/química , Metotrexato/uso terapéutico , Ratones , Ratones Endogámicos C57BL
16.
Acta Diabetol ; 56(3): 273-280, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30259114

RESUMEN

AIMS: Inflammation plays a role in the development and progression of type 2 diabetes macroangiopathy. Interleukin 33 (IL-33) drives production of Th2-associated cytokines. The soluble form of suppression of tumorigenicity 2 (sST2) acting as a decoy receptor blocks IL-33 and tones down Th2 inflammatory response. We investigated the role of sST2 as a predictor of CV and all-cause mortality in a cohort of patients affected by established atherosclerotic disease. METHODS: 399 patients with atherosclerotic disease from the Tor Vergata Atherosclerosis Registry performed follow-up every year by phone interview. The primary endpoint was cardiovascular death and the secondary endpoint was death for any other disease. RESULTS: sST2 plasma levels were significantly increased from normal glucose-tolerant patients to patients with history of type 2 diabetes (p < 0.00001). Levels of sST2 were significantly correlated with fasting plasma glucose (R = 0.16, p = 0.002), HbA1c (R = 0.17, p = 0.002), and HOMA (R = 0.16, p = 0.004). Dividing patients in tertiles of sST2 levels, those belonging to the highest tertile showed an increased rate of all-cause and cardiovascular mortality, (all-cause mortality p = 0.045 and CVD mortality p = 0.02). A multivariate Cox analysis revealed that sST2 increased the risk in cardiovascular mortality per SD by hazard ratio 1.050 (95% CI 1.006-1.097, p = 0.025) after adjustment for age and hs-CRP while it did not significantly change the risk for all-cause mortality. CONCLUSIONS: High circulating level of sST2 is associated to increased CVD mortality and markers of metabolic dysfunction in subjects with atherosclerotic disease.


Asunto(s)
Biomarcadores/sangre , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/mortalidad , Angiopatías Diabéticas/diagnóstico , Angiopatías Diabéticas/mortalidad , Trastornos del Metabolismo de la Glucosa/diagnóstico , Proteína 1 Similar al Receptor de Interleucina-1/sangre , Anciano , Anciano de 80 o más Años , Aterosclerosis/sangre , Aterosclerosis/diagnóstico , Aterosclerosis/etiología , Aterosclerosis/mortalidad , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Enfermedades Cardiovasculares/sangre , Causas de Muerte , Estudios de Cohortes , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/mortalidad , Angiopatías Diabéticas/sangre , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Trastornos del Metabolismo de la Glucosa/sangre , Trastornos del Metabolismo de la Glucosa/etiología , Trastornos del Metabolismo de la Glucosa/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo
17.
Atherosclerosis ; 277: 179-185, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29958653

RESUMEN

BACKGROUND AND AIMS: We aimed to identify novel biomarkers for cardiovascular mortality through a non-targeted metabolomics approach in patients with established atherosclerotic disease from the Tor Vergata Atherosclerosis Registry (TVAR). METHODS: We compared the serum baseline metabolome of 19 patients with atherosclerosis suffering from cardiovascular death during follow-up with the baseline serum metabolome of 20 control patients matched for age, gender, body mass index (BMI) and atherosclerotic disease status, who survived during the observation period. RESULTS: Three metabolites were significantly different in the cardiovascular mortality (CVM) group compared to controls: 2-hydroxycaproate, gluconate and sorbitol. 2-hydroxycaproate (otherwise known as alpha hydroxy caproate) was also significantly correlated with time to death. The metabolites performed better when combined together rather than singularly on the identification of CVM status. CONCLUSIONS: Our analysis led to identify few metabolites potentially amenable of translation into the clinical practice as biomarkers for specific metabolic changes in the cardiovascular system in patients with established atherosclerotic disease.


Asunto(s)
Aterosclerosis/sangre , Aterosclerosis/mortalidad , Caproatos/sangre , Hidroxiácidos/sangre , Anciano , Anciano de 80 o más Años , Aterosclerosis/diagnóstico , Biomarcadores/sangre , Estudios de Casos y Controles , Causas de Muerte , Femenino , Humanos , Italia/epidemiología , Masculino , Metabolómica/métodos , Valor Predictivo de las Pruebas , Pronóstico , Sistema de Registros , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo
18.
PLoS One ; 13(5): e0195912, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29742122

RESUMEN

A large body of evidence suggests that persistent dietary fat overload causes mitochondrial dysfunction and systemic metabolic gridlock. Mitochondrial and lipid metabolism in skeletal muscle (SkM) are severely affected upon persistent high fat diet (HFD) leading to premature tissue aging. Here, we designed weekly cycles of fasting (called as time-controlled fasting, TCF) and showed that they were effective in limiting mitochondrial damage and metabolic disturbances induced by HFD. Specifically, TCF was able to prevent the decline of adipose triglyceride lipase (Atgl), maintain efficient mitochondrial respiration in SkM as well as improve blood glucose and lipid profile. Atgl was found to be the mediator of such preventive effects as its downregulation or up-regulation in C2C12 myotubes triggers mitochondrial alteration or protects against the deleterious effects of high fat levels respectively. In conclusion, TCF could represent an effective strategy to limit mitochondrial impairment and metabolic inflexibility that are typically induced by modern western diets or during aging.


Asunto(s)
Envejecimiento/metabolismo , Grasas de la Dieta/efectos adversos , Ayuno/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Animales , Línea Celular , Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Lipasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
19.
Acta Diabetol ; 55(2): 121-129, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29134286

RESUMEN

AIMS: The tissue inhibitor of metalloproteinase TIMP3 is a stromal protein that restrains the activity of both protease and receptor in the extracellular matrix and has been found to be down-regulated in diabetic nephropathy (DN), the leading cause of end-stage renal disease in developed countries. METHODS: In order to gain deeper insights on the association of loss of TIMP3 and DN, we performed differential proteomic analysis of kidney and blood metabolic profiling of wild-type and Timp3-knockout mice before and after streptozotocin (STZ) treatment, widely used to induce insulin deficiency and hyperglycemia. RESULTS: Kidney proteomic data and blood metabolic profiles suggest significant alterations of peroxisomal and mitochondrial fatty acids ß-oxidation in Timp3-knockout mice compared to wild-type mice under basal condition. These alterations were exacerbated in response to STZ treatment. CONCLUSIONS: Proteomic and metabolomic approaches showed that loss of TIMP3 alone or in combination with STZ treatment results in significant alterations of kidney lipid metabolism and peripheral acylcarnitine levels, supporting the idea that loss of TIMP3 may generate a phenotype more prone to DN.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Fallo Renal Crónico/metabolismo , Metabolómica , Proteómica , Inhibidor Tisular de Metaloproteinasa-3/genética , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/inducido químicamente , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Riñón/metabolismo , Fallo Renal Crónico/inducido químicamente , Fallo Renal Crónico/genética , Fallo Renal Crónico/patología , Metabolismo de los Lípidos , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoma/análisis , Proteoma/metabolismo , Estreptozocina
20.
Sci Rep ; 7(1): 6747, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28751722

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) encompasses a broad spectrum of conditions, ranging from non-progressive bland steatosis to hepatocarcinoma. Tissue inhibitor of metalloproteinase 3 (Timp3) has a role in the pathogenesis of fatty liver disease associated with obesity and is silenced during metabolic disorders and liver cancer. We generated an hepatocyte-specific TIMP3 'gain-of-function' mouse model under the control of the Albumin promoter (AlbT3) and investigated its effects during high-fat diet (HFD). After 16 weeks of HFD, TIMP3 overexpression significantly improved glucose metabolism, hepatic fatty acid oxidation and cholesterol homeostasis. In AlbT3 mice CYP7A1, MDR3 and MRP2 gene expressions were observed, consistent with higher bile acid synthesis and export. Next, to evaluate the role of A Disintegrin and Metalloproteinase 17 (ADAM17), a crucial target of TIMP3, in these processes, we created mice deficient in Adam17 specifically in hepatocyte (A17LKO) or in myeloid lineage (A17MKO), founding that only A17LKO showed improvement in liver steatosis induced by HFD. Moreover, both, AlbT3 and A17LKO significantly reduced diethylnitrosamine-initiated, HFD-promoted hepatic tumorigenesis assessed by tumor multiplicity and total tumor area. Taken together, these data indicate that hepatic TIMP3 can slow progression of NAFLD, and tumorigenesis, at least in part, through the regulation of ADAM17 activity.


Asunto(s)
Proteína ADAM17/genética , Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Inhibidor Tisular de Metaloproteinasa-3/genética , Proteína ADAM17/deficiencia , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Albúminas/genética , Albúminas/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinógenos/toxicidad , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/prevención & control , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Dieta Alta en Grasa/efectos adversos , Dietilnitrosamina/toxicidad , Modelos Animales de Enfermedad , Mutación con Ganancia de Función , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/prevención & control , Masculino , Ratones , Ratones Transgénicos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Regiones Promotoras Genéticas , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-3/agonistas , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA