Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Physiol Rep ; 10(16): e15425, 2022 08.
Article En | MEDLINE | ID: mdl-35986504

eNOS-deficient mice were previously shown to develop hypertension and metabolic alterations associated with insulin resistance either in standard dietary conditions (eNOS-/- homozygotes) or upon high-fat diet (HFD) (eNOS+/- heterozygotes). In the latter heterozygote model, the present study investigated the pancreatic morphological changes underlying the abnormal glycometabolic phenotype. C57BL6 wild type (WT) and eNOS+/- mice were fed with either chow or HFD for 16 weeks. After being longitudinally monitored for their metabolic state after 8 and 16 weeks of diet, mice were euthanized and fragments of pancreas were processed for histological, immuno-histochemical and ultrastructural analyses. HFD-fed WT and eNOS+/- mice developed progressive glucose intolerance and insulin resistance. Differently from WT animals, eNOS+/- mice showed a blunted insulin response to a glucose load, regardless of the diet regimen. Such dysregulation of insulin secretion was associated with pancreatic ß-cell hyperplasia, as shown by larger islet fractional area and ß-cell mass, and higher number of extra-islet ß-cell clusters than in chow-fed WT animals. In addition, only in the pancreas of HFD-fed eNOS+/- mice, there was ultrastructural evidence of a number of hybrid acinar-ß-cells, simultaneously containing zymogen and insulin granules, suggesting the occurrence of a direct exocrine-endocrine transdifferentiation process, plausibly triggered by metabolic stress associated to deficient endothelial NO production. As suggested by confocal immunofluorescence analysis of pancreatic histological sections, inhibition of Notch-1 signaling, likely due to a reduced NO availability, is proposed as a novel mechanism that could favor both ß-cell hyperplasia and acinar-ß-cell transdifferentiation in eNOS-deficient mice with impaired insulin response to a glucose load.


Insulin Resistance , Insulin-Secreting Cells , Animals , Blood Glucose/metabolism , Cell Transdifferentiation , Diet, High-Fat/adverse effects , Glucose/metabolism , Hyperplasia/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Antioxidants (Basel) ; 10(1)2020 Dec 28.
Article En | MEDLINE | ID: mdl-33379141

In this paper we review the mechanisms of the antitumor effects of Hypericum perforatum L. (St. John's wort, SJW) and its main active component hyperforin (HPF). SJW extract is commonly employed as antidepressant due to its ability to inhibit monoamine neurotransmitters re-uptake. Moreover, further biological properties make this vegetal extract very suitable for both prevention and treatment of several diseases, including cancer. Regular use of SJW reduces colorectal cancer risk in humans and prevents genotoxic effects of carcinogens in animal models. In established cancer, SJW and HPF can still exert therapeutic effects by their ability to downregulate inflammatory mediators and inhibit pro-survival kinases, angiogenic factors and extracellular matrix proteases, thereby counteracting tumor growth and spread. Remarkably, the mechanisms of action of SJW and HPF include their ability to decrease ROS production and restore pH imbalance in tumor cells. The SJW component HPF, due to its high lipophilicity and mild acidity, accumulates in membranes and acts as a protonophore that hinders inner mitochondrial membrane hyperpolarization, inhibiting mitochondrial ROS generation and consequently tumor cell proliferation. At the plasma membrane level, HPF prevents cytosol alkalization and extracellular acidification by allowing protons to re-enter the cells. These effects can revert or at least attenuate cancer cell phenotype, contributing to hamper proliferation, neo-angiogenesis and metastatic dissemination. Furthermore, several studies report that in tumor cells SJW and HPF, mainly at high concentrations, induce the mitochondrial apoptosis pathway, likely by collapsing the mitochondrial membrane potential. Based on these mechanisms, we highlight the SJW/HPF remarkable potentiality in cancer prevention and treatment.

3.
Int J Mol Sci ; 21(21)2020 Oct 30.
Article En | MEDLINE | ID: mdl-33143088

Diabetes mellitus is a very common chronic disease with progressively increasing prevalence. Besides the well-known autoimmune and inflammatory pathogenesis of type 1 diabetes, in many people, metabolic changes and inappropriate lifestyle favor a subtle chronic inflammatory state that contributes to development of insulin resistance and progressive loss of ß-cell function and mass, eventually resulting in metabolic syndrome or overt type 2 diabetes. In this paper, we review the anti-inflammatory effects of the extract of Hypericum perforatum L. (St. John's wort, SJW) and its main active ingredients firstly in representative pathological situations on inflammatory basis and then in pancreatic ß cells and in obese or diabetic animal models. The simultaneous and long-lasting inhibition of signal transducer and activator of transcription (STAT)-1, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinases (MAPKs)/c-jun N-terminal kinase (JNK) signaling pathways involved in pro-inflammatory cytokine-induced ß-cell dysfunction/death and insulin resistance make SJW particularly suitable for both preventive and therapeutic use in metabolic diseases. Hindrance of inflammatory cytokine signaling is likely dependent on the hyperforin content of SJW extract, but recent data reveal that hypericin can also exert relevant protective effects, mediated by activation of the cyclic adenosine monophosphate (cAMP)/protein kinase cAMP-dependent (PKA)/adenosine monophosphate activated protein kinase (AMPK) pathway, against high-fat-diet-induced metabolic abnormalities. Actually, the mechanisms of action of the two main components of SJW appear complementary, strengthening the efficacy of the plant extract. Careful quantitative analysis of SJW components and suitable dosage, with monitoring of possible drug-drug interaction in a context of remarkable tolerability, are easily achievable pre-requisites for forthcoming clinical applications.


Diabetes Mellitus, Type 2/prevention & control , Hypericum/chemistry , Inflammation/drug therapy , Phloroglucinol/analogs & derivatives , Plant Extracts/pharmacology , Terpenes/pharmacology , Animals , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Humans , Phloroglucinol/pharmacology , Phytotherapy
5.
Diabetes ; 69(3): 279-290, 2020 03.
Article En | MEDLINE | ID: mdl-32079704

Paraphrasing the Swiss physician and father of toxicology Paracelsus (1493-1541) on chemical agents used as therapeutics, "the dose makes the poison," it is now realized that this aptly applies to the calorigenic nutrients. The case here is the pancreatic islet ß-cell presented with excessive levels of nutrients such as glucose, lipids, and amino acids. The short-term effects these nutrients exert on the ß-cell are enhanced insulin biosynthesis and secretion and changes in glucose sensitivity. However, chronic fuel surfeit triggers additional compensatory and adaptive mechanisms by ß-cells to cope with the increased insulin demand or to protect itself. When these mechanisms fail, toxicity due to the nutrient surplus ensues, leading to ß-cell dysfunction, dedifferentiation, and apoptosis. The terms glucotoxicity, lipotoxicity, and glucolipotoxicity have been widely used, but there is some confusion as to what they mean precisely and which is most appropriate for a given situation. Here we address the gluco-, lipo-, and glucolipo-toxicities in ß-cells by assessing the evidence both for and against each of them. We also discuss potential mechanisms and defend the view that many of the identified "toxic" effects of nutrient excess, which may also include amino acids, are in fact beneficial adaptive processes. In addition, candidate fuel-excess detoxification pathways are evaluated. Finally, we propose that a more general term should be used for the in vivo situation of overweight-associated type 2 diabetes reflecting both the adaptive and toxic processes to mixed calorigenic nutrients excess: "nutrient-induced metabolic stress" or, in brief, "nutri-stress."


Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Glucose , Humans , Insulin , Nutrients , Stress, Physiological
6.
J Pharm Pharmacol ; 71(1): 93-103, 2019 Jan.
Article En | MEDLINE | ID: mdl-28990659

OBJECTIVES: St John's wort extract (SJW) and its component hyperforin (HPF) were shown to potently inhibit cytokine-induced STAT-1 and NF-κB activation in pancreatic ß cells and protect them against injury. This study aimed at exploring the time course of STAT-1 inhibition afforded by these natural compounds in the ß-cell line INS-1E. METHODS: INS-1E cells were pre-incubated with SJW extract (2-5 µg/ml) or HPF (0.5-2 µm) and then exposed to a cytokine mixture. In some experiments, these compounds were added after or removed before cytokine exposure. STAT-1 activation was assessed by electrophoretic mobility shift assay, apoptosis by caspase-3 activity assay, mRNA gene expression by RT-qPCR. KEY FINDINGS: Pre-incubation with SJW/HPF for 1-2 h exerted a remarkable STAT-1 downregulation, which was maintained upon removal of the compounds before early or delayed cytokine addition. When the protective compounds were added after cell exposure to cytokines, between 15 and 90 min, STAT-1 inhibition also occurred at a progressively decreasing extent. Upon 24-h incubation, SJW and HPF counteracted cytokine-induced ß-cell dysfunction, apoptosis and target gene expression. CONCLUSIONS: SJW and HPF confer to ß cells a state of 'cytokine resistance', which can be elicited both before and after cytokine exposure and safeguards these cells from deleterious cytokine effects.


Hypericum/chemistry , Insulin-Secreting Cells/drug effects , Phloroglucinol/analogs & derivatives , Plant Extracts/pharmacology , Terpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytokines/metabolism , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/metabolism , Phloroglucinol/administration & dosage , Phloroglucinol/isolation & purification , Phloroglucinol/pharmacology , Plant Extracts/administration & dosage , Rats , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , Terpenes/administration & dosage , Terpenes/isolation & purification , Time Factors
7.
Nanomedicine ; 14(7): 2191-2203, 2018 10.
Article En | MEDLINE | ID: mdl-30016718

To improve the efficiency of pancreatic islet transplantation, we performed in-vitro and in-vivo experiments with isolated human pancreatic islets coated by multi-layer nano-encapsulation using differently charged polymers [chitosan and poly(sodium styrene sulfonate)] to obtain up to 9 layers. The islet coating (thickness: 104.2 ±â€¯4.2 nm) was uniform, with ≥ 90% cell viability and well preserved beta- and alpha-cell ultrastructure. Nano-encapsulated islets maintained physiological glucose-stimulated insulin secretion by both static incubation and perifusion studies. Notably, palmitate- or cytokine-induced toxicity was significantly reduced in nano-coated islets. Xenotransplantation of nano-encapsulated islets under the kidney capsule of streptozotocin-induced C57Bl/6J diabetic mice allowed long term normal or near normal glycemia, associated with minimal infiltration of immune cell into the grafts, well preserved islet morphology and signs of re-vascularization. In summary, the multi-layer nano-encapsulation approach described in the present study provides a promising tool to effectively protect human islets both in-vitro andin-vivo conditions.


Coated Materials, Biocompatible/chemistry , Diabetes Mellitus, Experimental/prevention & control , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Nanostructures/administration & dosage , Animals , Blood Glucose/analysis , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Nanostructures/chemistry , Transplantation, Heterologous
8.
Int J Biochem Cell Biol ; 81(Pt A): 92-104, 2016 12.
Article En | MEDLINE | ID: mdl-27780755

The extract of the herbaceous plant St. John's wort (SJW) and its phloroglucinol component hyperforin (HPF) were previously shown to inhibit cytokine-induced STAT-1 and NF-κB activation and prevent damage in pancreatic ß cells. To further clarify the mechanisms underlying their protective effects, we evaluated the phosphorylation state of various factors of cytokine signaling pathways and the expression of target genes involved in ß-cell function, inflammatory response and apoptosis induction. In the INS-1E ß-cell line, exposed to a cytokine mixture with/without SJW extract (2-5µg/ml) or HPF (1-5µM), protein phosphorylation was assessed by western blotting and expression of target genes by real-time quantitative PCR. SJW and HPF markedly inhibited, in a dose-dependent manner (from 60 to 100%), cytokine-induced activating phosphorylations of STAT-1, NF-κB p65 subunit and IKK (NF-κB inhibitory subunit IκBα kinase). MAPK and Akt pathways were also modulated by the vegetal compounds through hindrance of p38 MAPK, ERK1/2, JNK and Akt phosphorylations, each reduced by at least 65% up to 100% at the higher dose. Consistently, SJW and HPF a) abolished cytokine-induced mRNA expression of pro-inflammatory genes; b) avoided down-regulation of relevant ß-cell functional/differentiation genes; c) corrected cytokine-driven imbalance between pro- and anti-apoptotic factors, by fully preventing up-regulation of pro-apoptotic genes and preserving expression or function of anti-apoptotic Bcl-2 family members; d) protected INS-1E cells against cytokine-induced apoptosis. In conclusion, SJW extract and HPF exert their protective effects through simultaneous inhibition of multiple phosphorylation steps along various cytokine signaling pathways and consequent restriction of inflammatory and apoptotic gene expression. Thus, they have a promising therapeutic potential for the prevention or limitation of immune-mediated ß-cell dysfunction and damage leading to type 1 diabetes.


Apoptosis/genetics , Cytokines/metabolism , Hypericum/chemistry , Insulin-Secreting Cells/drug effects , Phloroglucinol/analogs & derivatives , Plant Extracts/pharmacology , Signal Transduction/drug effects , Terpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , DNA/metabolism , Enzyme Activation/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phloroglucinol/pharmacology , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , STAT1 Transcription Factor/metabolism , Transcriptional Activation/drug effects
9.
J Biomed Nanotechnol ; 11(4): 730-8, 2015 Apr.
Article En | MEDLINE | ID: mdl-26310079

Limited tools are available for the non-invasive monitoring of transplanted islets. In this study, we have compared the widely used superparamagnetic iron oxide nanoparticle ferumoxide (Endorem) and multiwalled carbon nanotubes (MWCNTs) for islet cell labeling and tracking. INS-1 E cells and human pancreatic islets isolated from 12 non-diabetic cadaveric organ donors (age: 62 ±16 yr, BMI: 24.6 ± 3.3 kg/m2) were incubated with 50 µg/ml Endorem or 15 µg/ml MWCNTs and studied after 7 or 14 days to assess beta cell morphology, ultrastructure, function, cell survival and in-vitro and in-vivo magnetic resonance imaging (MRI). Light and electron (EM) microscopy showed the well-maintained morphology and ultrastructure of both INS-1 E and human islets during the incubation. EM also revealed the presence of Endorem and MWCNTs within the beta but not the alpha cells. The compounds did not affect beta cell function and viability, and in-vitro MRI showed that labeled INS-1 E cells and human islets could be imaged. Finally, MWCNT labeled human islets were successfully transplanted into the subcutis of rats localized in the desired site via magnetic field and tracked by MRI. These data suggest that MWCNTs can be an alternative labeling compound to be used with human islets for experimental and transplantation studies.


Islets of Langerhans Transplantation/methods , Islets of Langerhans/drug effects , Nanotechnology/methods , Nanotubes, Carbon/chemistry , Aged , Animals , Cell Survival , Cells, Cultured , Contrast Media/chemistry , Dextrans/chemistry , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Microscopy, Electron , Microscopy, Fluorescence , Middle Aged , Rats
10.
PLoS One ; 9(8): e104156, 2014.
Article En | MEDLINE | ID: mdl-25093405

Abnormalities in eNOS gene, possibly interacting with high fat diet (HFD), affect peripheral vascular function and glucose metabolism. The relative role of eNOS gene, HFD and metabolic derangement on coronary function has not been fully elucidated. We test whether eNOS gene deficiency per se or in association with HFD modulates coronary function through mechanisms involving molecular pathways related to insulin signaling. Wild type (WT), eNOS-/- and eNOS+/- mice were studied. WT and eNOS+/- mice were fed with either standard or HF diet for 16 weeks and compared with standard diet fed eNOS-/-. Glucose and insulin tolerance tests were performed during the last week of diet. Coronary resistance (CR) was measured at baseline and during infusions of acetylcholine (Ach) or sodium-nitroprusside (SNP) to evaluate endothelium-dependent or independent vasodilation, in the Langendorff isolated hearts. Cardiac expression of Akt and ERK genes as evaluation of two major insulin-regulated signaling pathways involved in the control of vascular tone were assessed by western blot. HFD-fed mice developed an overt diabetic state. Conversely, chow-fed genetically modified mice (in particular eNOS-/-) showed a metabolic pattern characterized by normoglycemia and hyperinsulinemia with a limited degree of insulin resistance. CR was significantly higher in animals with eNOS gene deletions than in WT, independently of diet. Percent decrease in CR, during Ach infusion, was significantly lower in both eNOS-/- and eNOS+/- mice than in WT, independently of diet. SNP reduced CR in all groups except eNOS-/-. The cardiac ERK1-2/Akt ratio, increased in animals with eNOS gene deletions compared with WT, independently of diet. These results suggest that the eNOS genetic deficiency, associated or not with HFD, has a relevant effect on coronary vascular function, possibly mediated by increase in blood insulin levels and unbalance in insulin-dependent signaling in coronary vessels, consistent with a shift towards a vasoconstrictive pattern.


Coronary Vessels/physiopathology , Diet, High-Fat , Gene Deletion , Hyperinsulinism/physiopathology , Insulin/metabolism , Myocardium/enzymology , Nitric Oxide Synthase Type III/genetics , Signal Transduction , Animals , Blood Glucose/metabolism , Body Weight , Coronary Vessels/metabolism , Coronary Vessels/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glucose Tolerance Test , Hyperinsulinism/blood , Hyperinsulinism/complications , In Vitro Techniques , Insulin/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Myocardium/pathology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Resistance
11.
Eur J Pharmacol ; 729: 37-44, 2014 Apr 15.
Article En | MEDLINE | ID: mdl-24530416

We previously showed that the innovative radical scavenger bis(1-hydroxy-2,2,6,6-tetramethyl-4-piperidinyl)-decandioate (IAC) improves metabolic dysfunctions in a diabetic mouse model. Here, we compared the in vivo effects of IAC with those of the anti-diabetic drugs pioglitazone (PIO) and exendin-4 (EX-4). Diabetes was induced in C57Bl/6J mice by streptozotocin and nicotinamide administration. Paralleled by healthy controls, diabetic animals (D) were randomly assigned to four groups and treated daily for 7 consecutive weeks: D+saline, ip; D+IAC 30mg/kgb.w., ip; D+PIO 10mg/kgb.w. per os; and D+EX-4, 50µg/kgb.w., ip. Our results show that IAC reduced basal hyperglycemia and improved glucose tolerance better than PIO or EX-4. Interestingly, in the heart of diabetic mice, IAC treatment normalized the increased levels of GSSG/GSH ratio and thiobarbituric acid reactive substances, indexes of oxidative stress and damage, while PIO and EX-4 were less effective. As supported by immunohistochemical data, IAC markedly prevented diabetic islet ß-cell reduced density, differently from PIO and EX-4 that had only a moderate effect. Interestingly, in diabetic animals, IAC treatment enhanced the activity of pancreatic-duodenal homeobox 1 (PDX-1), an oxidative stress-sensitive transcription factor essential for maintenance of ß-cell function, as evaluated by quantification of its nuclear immunostaining, whereas PIO or EX-4 treatments did not. Altogether, these observations support the improvement of the general redox balance and ß-cell function induced by IAC treatment in streptozotocin-nicotinamide diabetic mice. Furthermore, in this model, the correction of diabetic alterations was better obtained by treatment with the radical scavenger IAC than with pioglitazone or exendin-4.


Diabetes Mellitus, Experimental/drug therapy , Free Radical Scavengers/therapeutic use , Hypoglycemic Agents/therapeutic use , Peptides/therapeutic use , Thiazolidinediones/therapeutic use , Venoms/therapeutic use , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Exenatide , Free Radical Scavengers/pharmacology , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Niacinamide/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptides/pharmacology , Pioglitazone , Random Allocation , Streptozocin/toxicity , Thiazolidinediones/pharmacology , Venoms/pharmacology
12.
J Nat Prod ; 77(3): 543-9, 2014 Mar 28.
Article En | MEDLINE | ID: mdl-24417609

Garcinol (1), a polyisoprenylated benzophenone occurring in Garcinia species, has been reported to exert anti-inflammatory activity in LPS-stimulated macrophages, through inhibition of NF-κB and/or JAK/STAT-1 activation. In order to provide deeper insight into its effects on the cytokine signaling pathway and to clarify the underlying molecular mechanisms, 1 was isolated from the fruits of Garcinia cambogia along with two other polyisoprenylated benzophenones, guttiferones K (2) and guttiferone M (3), differing from each other in their isoprenyl moieties and their positions on the benzophenone core. The affinities of 1-3 for the STAT-1 protein have been evaluated by surface plasmon resonance and molecular docking studies and resulted in KD values in the micromolar range. Consistent with the observed high affinity toward the STAT-1 protein, garcinol and guttiferones K and M were able to modulate cytokine signaling in different cultured cell lines, mainly by inhibiting STAT-1 nuclear transfer and DNA binding, as assessed by an electrophorectic mobility shift assay.


Benzophenones/isolation & purification , Benzophenones/pharmacology , Garcinia cambogia/chemistry , Macrophages/drug effects , STAT1 Transcription Factor/drug effects , Terpenes/chemistry , Terpenes/pharmacology , Benzophenones/chemistry , Blotting, Northern , Female , Fruit/chemistry , Humans , Lipopolysaccharides/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Molecular Conformation , Molecular Structure , NF-kappa B/antagonists & inhibitors , NF-kappa B/drug effects , Signal Transduction/drug effects , Sri Lanka , Terpenes/isolation & purification
13.
Diabetologia ; 57(2): 362-5, 2014 Feb.
Article En | MEDLINE | ID: mdl-24233056

AIMS/HYPOTHESIS: Previous work has demonstrated that beta cell amount (whether measured as beta cell mass, beta cell volume or insulin-positive area) is decreased in type 2 diabetes; however, recent findings suggest that mechanisms other than death may contribute to beta cell failure in this disease. To better characterise beta cell mass and function in type 2 diabetes, we performed morphological, ultra-structural and functional studies using histological samples and isolated islets. METHODS: Pancreases from ten non-diabetic (ND) and ten matched type 2 diabetic organ donors were studied by insulin, glucagon and chromogranin A immunocytochemistry and electron microscopy (EM). Glucose-stimulated insulin secretion was assessed using isolated islets and studies were performed using independent ND islet preparations after 24 h exposure to 22.2 mmol/l glucose. RESULTS: Immunocytochemistry showed that the fractional islet insulin-positive area was lower in type 2 diabetic islets (54.9 ± 6.3% vs 72.1 ± 8.7%, p < 0.01), whereas glucagon (23.3 ± 5.4% vs 20.2 ± 5.3%) and chromogranin A (86.4 ± 6.1% vs 89.0 ± 5.5%) staining was similar between the two groups. EM showed that the proportion of beta cells in type 2 diabetic islets was only marginally decreased; marked beta cell degranulation was found in diabetic beta cells; these findings were all reproduced after exposing isolated ND islets to high glucose. Glucose-stimulated insulin secretion was 40­50% lower from type 2 diabetic islets (p < 0.01), which again was mimicked by culturing non-diabetic islets in high glucose. CONCLUSIONS/INTERPRETATION: These results suggest that, at least in subgroups of type 2 diabetic patients, the loss of beta cells as assessed so far might be overestimated, possibly due to changes in beta cell phenotype other than death, also contributing to beta cell failure in type 2 diabetes.


Chromogranin A/metabolism , Diabetes Mellitus, Type 2/pathology , Glucagon/metabolism , Insulin-Secreting Cells/pathology , Insulin/metabolism , Pancreas/pathology , Aged , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Immunohistochemistry , Male , Microscopy, Electron
14.
Acta Diabetol ; 51(1): 113-21, 2014 Feb.
Article En | MEDLINE | ID: mdl-24121871

The extract of Hypericum perforatum (St. John's wort, SJW) and its component hyperforin (HPF) were previously shown to inhibit cytokine-induced activation of signal transducer and activator of transcription-1 and nuclear factor κB and prevent apoptosis in a cultured ß-cell line. Objective of this study was to assess the protection exerted by SJW and HPF on isolated rat and human islets exposed to cytokines in vitro. Functional, ultrastructural, biomolecular and cell death evaluation studies were performed. In both rat and human islets, SJW and HPF counteracted cytokine-induced functional impairment and down-regulated mRNA expression of pro-inflammatory target genes, such as iNOS, CXCL9, CXCL10, COX2. Cytokine-induced NO production from cultured islets, evaluated by nitrites measurement in the medium, was significantly reduced in the presence of the vegetal compounds. Noteworthy, the increase in apoptosis and necrosis following 48-h exposure to cytokines was fully prevented by SJW and partially by HPF. Ultrastructural morphometric analysis in human islets exposed to cytokines for 20 h showed that SJW or HPF avoided early ß-cell damage (e.g., mitochondrial alterations and loss of insulin granules). In conclusion, SJW compounds protect rat and human islets against cytokine effects by counteracting key mechanisms of cytokine-mediated ß-cell injury and represent promising pharmacological tools for prevention or limitation of ß-cell dysfunction and loss in type 1 diabetes.


Cytokines/toxicity , Cytoprotection/drug effects , Hypericum , Islets of Langerhans/drug effects , Phloroglucinol/analogs & derivatives , Plant Extracts/pharmacology , Terpenes/pharmacology , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Glucose/pharmacology , Humans , Hypericum/chemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Male , Phloroglucinol/pharmacology , Rats , Rats, Sprague-Dawley
15.
J Med Chem ; 56(11): 4718-28, 2013 Jun 13.
Article En | MEDLINE | ID: mdl-23662847

ATP-sensitive potassium (KATP) channels play a prominent role in controlling cardiovascular function. In this paper, a novel series of 4-(1-oxo-2-cyclopentenyl)-1,4-benzothiazine derivatives modified at the C-2, and C-6 positions were synthesized as openers of vascular KATP channels. Most of the tested compounds evoked vasorelaxing effects on rat aortic rings and membrane hyperpolarization in human vascular smooth muscle cells, with potency similar or superior to that of the reference levcromakalim (LCRK). The selective KATP blocker glibenclamide antagonized the above vascular effects, confirming that KATP channels are closely involved in the mechanism of action. The experimental results confirmed the 1,4-benzothiazine nucleus as an optimal scaffold for activators of vascular KATP channels; moreover, the high level of potency exhibited by the 6-acetyl substituted benzothiazine 8, along with the lack of any significant interference with insulin secretion from pancreatic ß-cells, paves the way to further develop a new series of potent activators of vascular KATP channels.


Cyclopentanes/chemical synthesis , KATP Channels/metabolism , Thiazines/chemical synthesis , Vasodilator Agents/chemical synthesis , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Cyclopentanes/chemistry , Cyclopentanes/pharmacology , Humans , Hydrogen Bonding , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Ion Channel Gating , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Rats , Rats, Wistar , Structure-Activity Relationship , Thiazines/chemistry , Thiazines/pharmacology , Vasodilation/drug effects , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacology
16.
Biochim Biophys Acta ; 1833(8): 1904-13, 2013 Aug.
Article En | MEDLINE | ID: mdl-23545415

PRIMA-1 is a chemical compound identified as a growth suppressor of tumor cells expressing mutant p53. We previously found that in the MDA-MB-231 cell line expressing high level of the mutant p53-R280K protein, PRIMA-1 induced p53 ubiquitination and degradation associated to cell death. In this study, we investigated the ability of PRIMA-1 to induce autophagy in cancer cells. In MDA-MB-231 and HCT116 cells, expressing mutant or wild type p53, respectively, autophagy occurred following exposure to PRIMA-1, as shown by acridine orange staining, anti-LC3 immunofluorescence and immunoblots, as well as by electron microscopy. Autophagy was triggered also in the derivative cell lines knocked-down for p53, although to a different extent than in the parental cells expressing mutant or wild type p53. In particular, while wild type p53 limited PRIMA-1 induced autophagy, mutant p53 conversely promoted autophagy, thus sustaining cell viability following PRIMA-1 treatment. Therefore, the autophagic potential of PRIMA-1, besides being cell context dependent, could be modulated in a different way by the presence of wild type or mutant p53. Furthermore, since both cell lines lacking p53 were more sensitive to the cytotoxic effect of PRIMA-1 than the parental ones, our findings suggest that a deregulated autophagy may favor cell death induced by this drug.


Autophagy/drug effects , Membrane Proteins/pharmacology , Mutation , Neoplasms/genetics , Nerve Tissue Proteins/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Autophagy/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , HCT116 Cells , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
18.
Acta Diabetol ; 49 Suppl 1: S247-52, 2012 Dec.
Article En | MEDLINE | ID: mdl-23184237

We performed an ultrastructural morphometric analysis of insulin secretory granules in pancreatic beta cells from control and type 2 diabetic multiorgan donors. The volume density of insulin granules significantly (p < 0.05) reduced in beta cells from type 2 diabetic patients with respect to non-diabetic subjects, and this reduction was mainly attributable to a decrease in mature granules. On the contrary, no significant difference was observed in the volume density of docked granules between controls and type 2 diabetic patients. In addition, there was a significant positive correlation between the density volume of total insulin granules and stimulated insulin secretion in non-diabetic islets. In conclusion, we detected significant changes in the intracellular distribution of insulin secretory granules within the beta cell that might be related with the alterations in insulin secretion observed in type 2 diabetes patients.


Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Secretory Vesicles/ultrastructure , Aged , Female , Humans , Insulin Secretion , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Male , Microscopy, Electron , Middle Aged , Secretory Vesicles/metabolism
19.
Front Physiol ; 3: 160, 2012.
Article En | MEDLINE | ID: mdl-22675305

Myocardial dysfunction and coronary macro/microvascular alterations are the hallmarks of diabetic cardiomyopathy and are ascribed to increased oxidative stress and altered nitric oxide synthase (NOS) activity. We hypothesize that pre-treatment by cobalt-protoporphyrin IX (CoPP) ameliorates both myocardial function and coronary circulation in streptozotocin (STZ)-induced diabetic rats. Isolated hearts from diabetic rats in Langendorff configuration displayed lower left ventricular function and higher coronary resistance (CR) compared to hearts from control animals. CoPP treatment of diabetic animals (0.3 mg/100 g body weight i.p., once a week for 3 weeks) significantly increased all the contractile/relaxation indexes (p < 0.01), while decreasing CR (p < 0.01). CoPP enhanced HO-1 protein levels and reduced oxidative stress in diabetic animals, as indicated by the significant (p < 0.05) decrease in heart % GSSG, [Formula: see text] and malondialdehyde (MDA) levels. CoPP increased adiponectin levels and phosphorylation of AKT and AMPK and reversed the eNOS/iNOS expression imbalance observed in the untreated diabetic heart. Furthermore, after CoPP treatment, a rise in malonyl-CoA as well as a decrease in acetyl-CoA was observed in diabetic hearts. In this experimental model of diabetic cardiomyopathy, CoPP treatment improved both cardiac function and coronary flow by blunting oxidative stress, restoring eNOS/iNOS expression balance and increasing HO-1 levels, thereby favoring improvement in both endothelial function and insulin sensitivity.

20.
PLoS One ; 7(5): e36188, 2012.
Article En | MEDLINE | ID: mdl-22563482

We have investigated the in vitro effects of increased levels of glucose and free fatty acids on autophagy activation in pancreatic beta cells. INS-1E cells and isolated rat and human pancreatic islets were incubated for various times (from 2 to 24 h) at different concentrations of glucose and/or palmitic acid. Then, cell survival was evaluated and autophagy activation was explored by using various biochemical and morphological techniques. In INS-1E cells as well as in rat and human islets, 0.5 and 1.0 mM palmitate markedly increased autophagic vacuole formation, whereas high glucose was ineffective alone and caused little additional change when combined with palmitate. Furthermore, LC3-II immunofluorescence co-localized with that of cathepsin D, a lysosomal marker, showing that the autophagic flux was not hampered in PA-treated cells. These effects were maintained up to 18-24 h incubation and were associated with a significant decline of cell survival correlated with both palmitate concentration and incubation time. Ultrastructural analysis showed that autophagy activation, as evidenced by the occurrence of many autophagic vacuoles in the cytoplasm of beta cells, was associated with a diffuse and remarkable swelling of the endoplasmic reticulum. Our results indicate that among the metabolic alterations typically associated with type 2 diabetes, high free fatty acids levels could play a role in the activation of autophagy in beta cells, through a mechanism that might involve the induction of endoplasmic reticulum stress.


Autophagy/drug effects , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Palmitates/pharmacology , Animals , Blotting, Western , Cathepsin D/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Glucose/pharmacology , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , Insulinoma/metabolism , Insulinoma/pathology , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
...