Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mutagenesis ; 31(5): 511-29, 2016 09.
Article in English | MEDLINE | ID: mdl-27056944

ABSTRACT

The FP7 Sanowork project was aimed to minimise occupational hazard and exposure to engineered nanomaterials (ENM) through the surface modification in order to prevent possible health effects. In this frame, a number of nanoparticles (NP) have been selected, among which zirconium (ZrO2) and titanium (TiO2) dioxide. In this study, we tested ZrO2 NP and TiO2 NP either in their pristine (uncoated) form, or modified with citrate and/or silica on their surface. As benchmark material, Aeroxide® P25 was used. We assessed cytotoxicity, genotoxicity and induction of morphological neoplastic transformation of NP by using a panel of in vitro assays in an established mammalian cell line of murine origin (Balb/3T3). Cell viability was evaluated by means of colony-forming efficiency assay (CFE). Genotoxicity was investigated by cytokinesis-block micronucleus cytome assay (CBMN cyt) and comet assay, and by the use of the restriction enzymes EndoIII and Fpg, oxidatively damaged DNA was detected; finally, the morphological neoplastic transformation of NP was assayed in vitro by cell transformation assay (CTA). Our results show that the surface remediation has not been effective in modifying cyto- and genotoxic properties of the nanomaterials tested; indeed, in the case of remediation of zirconia and titania with citrate, there is a tendency to emphasise the toxic effects. The use of a panel of assays, such as those we have employed, allowing the evaluation of multiple endpoints, including cell transformation, seems particularly advisable especially in the case of long-term exposure effects in the same cell type.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , DNA Damage , Metal Nanoparticles/toxicity , Mutagenicity Tests , Titanium/toxicity , Zirconium/toxicity , Animals , Cell Line , Cell Survival , DNA/drug effects , Metal Nanoparticles/chemistry , Mice , Oxidative Stress , Titanium/pharmacology , Zirconium/pharmacology
2.
J Biol Chem ; 286(16): 14007-18, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21335552

ABSTRACT

Glutamate is the major excitatory neurotransmitter of the central nervous system (CNS) and may induce cytotoxicity through persistent activation of glutamate receptors and oxidative stress. Its extracellular concentration is maintained at physiological concentrations by high affinity glutamate transporters of the solute carrier 1 family (SLC1). Glutamate is also present in islet of Langerhans where it is secreted by the α-cells and acts as a signaling molecule to modulate hormone secretion. Whether glutamate plays a role in islet cell viability is presently unknown. We demonstrate that chronic exposure to glutamate exerts a cytotoxic effect in clonal ß-cell lines and human islet ß-cells but not in α-cells. In human islets, glutamate-induced ß-cell cytotoxicity was associated with increased oxidative stress and led to apoptosis and autophagy. We also provide evidence that the key regulator of extracellular islet glutamate concentration is the glial glutamate transporter 1 (GLT1). GLT1 localizes to the plasma membrane of ß-cells, modulates hormone secretion, and prevents glutamate-induced cytotoxicity as shown by the fact that its down-regulation induced ß-cell death, whereas GLT1 up-regulation promoted ß-cell survival. In conclusion, the present study identifies GLT1 as a new player in glutamate homeostasis and signaling in the islet of Langerhans and demonstrates that ß-cells critically depend on its activity to control extracellular glutamate levels and cellular integrity.


Subject(s)
Excitatory Amino Acid Transporter 2/biosynthesis , Gene Expression Regulation , Glutamate Plasma Membrane Transport Proteins/biosynthesis , Insulin-Secreting Cells/cytology , Animals , Apoptosis , Autophagy , Cell Survival , Excitatory Amino Acid Transporter 2/physiology , Glutamate Plasma Membrane Transport Proteins/physiology , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Homeostasis , Humans , Islets of Langerhans/cytology , Mice , Models, Biological , Oxidative Stress
3.
Proc Natl Acad Sci U S A ; 106(33): 13992-7, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19666551

ABSTRACT

beta-Cell dysfunction is an important factor in the development of hyperglycemia of type-2 diabetes mellitus, and pancreatic islet amyloidosis (IA) has been postulated to be one of the main contributors to impaired insulin secretion. The aim of this study was to evaluate the correlation of IA with metabolic parameters and its effect on islets of Langerhans remodeling and relative endocrine-cell volume in baboons. We sequenced the amylin peptide, determined the fibrillogenic propensities, and evaluated pancreatic histology, clinical and biochemical characteristics, and endocrine cell proliferation and apoptosis in 150 baboons with different metabolic status. Amylin sequence in the baboon was 92% similar to humans and showed superimposable fibrillogenic propensities. IA severity correlated with fasting plasma glucose (FPG) (r = 0.662, P < 0.001) and HbA1c (r = 0.726, P < 0.001), as well as with free fatty acid, glucagon values, decreased homeostasis model assessment (HOMA) insulin resistance, and HOMA-B. IA severity was associated with a decreased relative beta-cell volume, and increased relative alpha-cell volume and hyperglucagonemia. These results strongly support the concept that IA and beta-cell apoptosis in concert with alpha-cell proliferation and hypertrophy are key determinants of islets of Langerhans "dysfunctional remodeling" and hyperglycemia in the baboon, a nonhuman primate model of type-2 diabetes mellitus. The most important determinants of IA were age and FPG (R(2) = 0.519, P < 0.0001), and different FPG levels were sensitive and specific to predict IA severity. Finally, a predictive model for islet amyloid severity was generated with age and FPG as required variables.


Subject(s)
Amyloidosis/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/pathology , Amyloid/metabolism , Animals , Apoptosis , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Fatty Acids/metabolism , Female , Insulin Resistance , Islet Amyloid Polypeptide , Male , Papio
4.
J Immunol ; 183(2): 993-1004, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19561093

ABSTRACT

Human clinical trials in type 1 diabetes (T1D) patients using mesenchymal stem cells (MSC) are presently underway without prior validation in a mouse model for the disease. In response to this void, we characterized bone marrow-derived murine MSC for their ability to modulate immune responses in the context of T1D, as represented in NOD mice. In comparison to NOD mice, BALB/c-MSC mice were found to express higher levels of the negative costimulatory molecule PD-L1 and to promote a shift toward Th2-like responses in treated NOD mice. In addition, transfer of MSC from resistant strains (i.e., nonobese resistant mice or BALB/c), but not from NOD mice, delayed the onset of diabetes when administered to prediabetic NOD mice. The number of BALB/c-MSC trafficking to the pancreatic lymph nodes of NOD mice was higher than in NOD mice provided autologous NOD-MSC. Administration of BALB/c-MSC temporarily resulted in reversal of hyperglycemia in 90% of NOD mice (p = 0.002). Transfer of autologous NOD-MSC imparted no such therapeutic benefit. We also noted soft tissue and visceral tumors in NOD-MSC-treated mice, which were uniquely observed in this setting (i.e., no tumors were present with BALB/c- or nonobese resistant mice-MSC transfer). The importance of this observation remains to be explored in humans, as inbred mice such as NOD may be more susceptible to tumor formation. These data provide important preclinical data supporting the basis for further development of allogeneic MSC-based therapies for T1D and, potentially, for other autoimmune disorders.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Animals , Bone Marrow Cells , Cell Movement , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/immunology , Hyperglycemia/therapy , Immunologic Factors/immunology , Mesenchymal Stem Cell Transplantation/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Neoplasms/etiology , Treatment Outcome
5.
Cell Transplant ; 17(12): 1323-36, 2008.
Article in English | MEDLINE | ID: mdl-19364070

ABSTRACT

To learn more about nonimmune-mediated islet graft failure, we transplanted different preparations (preps) of isolated human islets under the kidney capsule of streptozotocin (STZ)-diabetic nude mice. One month after the implantation of 1,000 or 2,000 islets, grafts were harvested for morphological, immunohistochemical, and ultrastructural analysis. Only a single islet prep cured the diabetes out of all the recipients, while the remaining preps showed only partial function after the implantation of 2,000 islets. Transplanted mice showed high circulating proinsulin levels but, with the exclusion of those bearing curative grafts, relatively low mature insulin levels. Engrafted beta-cells showed positive carboxypeptidase E (CPE) and prohormone convertase 1 (PC1) staining, while prohormone convertase 2 (PC2) was undetectable. In contrast, PC2 was abundantly expressed by engrafted alpha-cells. Moreover, engrafted beta-cells did not show evidence of replication, and preapoptotic beta-cells, with intra- and extracellular amyloid deposition, were detected with electron microscopy. Cell cycle inhibitors p16(INK4), p21(WAF1), and p27(Kip1) were abundantly expressed in the islet grafts and showed a predominant nuclear localization. In conclusion, diabetic nude mice transplanted with human islets showed disproportionate hyperproinsulinemia and graft evidence of beta-cell restricted PC2 depletion, amyloid deposition and beta-cell death, and lack of beta-cell replication with nuclear translocation of p27(Kip1) and p21(WAF1) that together may contribute to delayed graft failure.


Subject(s)
Cell Cycle/physiology , Diabetes Mellitus, Experimental/surgery , Hyperinsulinism/etiology , Islets of Langerhans Transplantation/adverse effects , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Proinsulin/metabolism , Proprotein Convertase 2/deficiency , Transplantation, Heterologous/adverse effects , Animals , Autopsy , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/pathology , Male , Mice , Mice, Nude , Pancreas/pathology , Proinsulin/blood , Reference Values , Treatment Failure
6.
Endocr Pathol ; 19(2): 104-11, 2008.
Article in English | MEDLINE | ID: mdl-18568298

ABSTRACT

c-kit (CD117) is a tyrosine kinase receptor involved in the proliferation, differentiation, and secretory functions of various cells. In experimental animal models, c-kit has been detected in the pars intermedia of the normal pituitary gland and in alpha-melanocyte-stimulating-hormone-positive adenomas and it has been suggested that it plays a role in regulating adrenocorticotropic hormone (ACTH) secretion. To the best of our knowledge, the expression of c-kit in normal human pituitary cells and in pituitary adenomas has never been reported, so the possible biological role of this receptor in the control of pituitary hormone secretion remains unclear. The aim of this study was to evaluate the immunohistochemical expression of c-kit in normal human pituitary glands and in a series of 62 well-characterized pituitary adenomas. In normal adenohypophyses, several cells, mainly located in the central mucoid wedge, showed a c-kit immunoreactivity (IR). Double label immunostaining procedures showed that the c-kit-IR cells corresponded to ACTH cells. Out of 62 adenomas, 15 (24%) were c-kit-IR, including 7/16 (44%) ACTH cell, 3/7 (42%) null cell, 4/11 (36%) alpha-subunit cell, and 1/11 (10%) follicle-stimulating hormone-luteinizing hormone cell adenomas. By contrast, all ten prolactin cell and seven growth hormone cell adenomas were c-kit negative. These data suggest that, in normal conditions, c-kit may be involved in the pituitary-adrenal axis regulation.


Subject(s)
Adenoma/metabolism , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , Proto-Oncogene Proteins c-kit/biosynthesis , ACTH-Secreting Pituitary Adenoma/metabolism , ACTH-Secreting Pituitary Adenoma/pathology , Adenoma/pathology , Adolescent , Adult , Aged , Blotting, Western , Child , Female , Follicle Stimulating Hormone/blood , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Growth Hormone-Secreting Pituitary Adenoma/pathology , Humans , Immunohistochemistry , Luteinizing Hormone/blood , Male , Middle Aged , Paraffin Embedding , Pituitary Gland/cytology , Pituitary Neoplasms/pathology , Prolactinoma/metabolism , Prolactinoma/pathology , Tissue Fixation
7.
Pathol Res Pract ; 204(4): 219-33, 2008.
Article in English | MEDLINE | ID: mdl-18304753

ABSTRACT

Gastrointestinal stromal tumors (GISTs) are c-Kit-positive neoplasms of the digestive tract. Few studies have reported their real incidence and malignancy. Two systems of risk assessment of aggressive behavior were recently proposed. The aims of our study were (1) to ascertain the frequency of GISTs and their clinical behavior in a large series of cases with a long-term follow-up, (2) to evaluate the prognostic value of two well-known risk assessment classifications, and (3) to propose an alternative prognostic index based on our data. Statistical analyses were performed to identify possible predictors of malignant behavior. One hundred and eighteen out of 169 (70%) mesenchymal tumors were GISTs. They were located in the stomach (57%), small intestine (31%), colon/rectum (6%), and omentum/mesentery (6%). Eighteen cases (16%) showed malignant behavior, with local recurrence in eight cases and distant metastases in 11 cases. Fifteen of 114 (13%) patients died of disease within 74 months, whereas 63 (55%) patients were still alive after a median period of 78 months. At multivariate analysis, high-risk category (according to Fletcher's criteria), omental/colorectal site, and younger patient age were independent predictors of malignant behavior. In addition to the evaluation of risk category, tumor site and patient age helped to better identify patients requiring stricter monitoring.


Subject(s)
Gastrointestinal Stromal Tumors/epidemiology , Gastrointestinal Stromal Tumors/pathology , Neoplasm Recurrence, Local , Adult , Age Factors , Aged , Disease-Free Survival , Female , Gastrointestinal Stromal Tumors/classification , Gastrointestinal Stromal Tumors/mortality , Gastrointestinal Stromal Tumors/therapy , Health Status Indicators , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Proportional Hazards Models , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
8.
Ultrastruct Pathol ; 32(6): 246-51, 2008.
Article in English | MEDLINE | ID: mdl-19117266

ABSTRACT

BACE2 is a protease homologous to BACE1 protein, an enzyme involved in the amyloid formation of Alzheimer disease (AD). However, despite the high homology between these two proteins, the biological role of BACE2 is still controversial, even though a few studies have suggested a pathogenetic role in sporadic inclusion-body myositis and hereditary inclusion-body myopathy, which are characterized by vacuolization of muscular fibers with intracellular deposits of proteins similar to those found in the brain of AD patients. Although BACE2 has also been identified in the pancreas, its function remains unknown and its specific localization in different pancreatic cell types has not been definitively ascertained. For these reasons, the authors have investigated the cellular and subcellular localization of BACE2 in normal rodent pancreases. BACE2 immunoreactivity was found in secretory granules of beta cells, co-stored with insulin and IAPP, while it was lacking in the other endocrine and exocrine cell types. The presence of BACE2 in secretory granules of beta cells suggests that it may play a role in diabetes-associated amyloidogenesis.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Insulin-Secreting Cells/enzymology , Secretory Vesicles/enzymology , Amyloid Precursor Protein Secretases/ultrastructure , Animals , Aspartic Acid Endopeptidases/ultrastructure , Immunohistochemistry , Insulin-Secreting Cells/ultrastructure , Mice , Microscopy, Electron, Transmission , Rats , Secretory Vesicles/ultrastructure
9.
Diabetes Care ; 28(6): 1303-10, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15920043

ABSTRACT

OBJECTIVE: Diabetes, hypertension, infections, and nephrotoxicity of certain immunosuppressive drugs (i.e., calcineurin inhibitors) can reduce functional survival of the kidney graft. Our aim was to evaluate survival, hypertrophy, and vascular function of the kidney graft in end-stage renal disease (ESRD) type 1 diabetic patients after transplant. RESEARCH DESIGN AND METHODS: The study population consisted of 234 ESRD type 1 diabetic patients who underwent kidney-pancreas (KP; 166 patients), successful kidney-islet (KI-s; 24 patients), and kidney (KD; 44 patients) transplant. Kidney size, graft survival, vascular function, and microalbuminuria were evaluated prospectively yearly for 6 years. Sixty-eight protocol kidney biopsies were performed routinely between 1993 and 1998 cross-sectionally (3.2 +/- 0.3 years from kidney transplant). RESULTS: The KP and KI-s groups had better cumulative kidney graft survival at 6 years than did the KD group (KP: 73%; KI-s: 86%; KD: 42%, P < 0.01). The KP group but not the KI-s/KD groups showed a persistent kidney graft hypertrophy up to 6 years of follow-up. A significant increase in creatinine levels from baseline to year 6 was evident in the KD group (1.58 +/- 0.08 to 2.78 +/- 0.44 mg/dl, P < 0.05) but not in the KP/KI-s groups. The KP/KI-s groups only showed a reduction of renal resistance index from baseline to year 6 (KP at baseline: 0.74 +/- 0.01 to 0.68 +/- 0.01%, P < 0.01; KI-s at baseline: 0.72 +/- 0.02 to 0.69 +/- 0.02%, P < 0.05). At year 6, an increase from baseline in urinary albumin excretion was observed only in the KD group (31.4 +/- 9.0 to 82.9 +/- 33.6 mg/l, P < 0.05). Preliminary data suggested that graft nitric oxide (NO) expression was higher in the KP/KI-s groups than in the KD group (data not shown). CONCLUSIONS: In ESRD type 1 diabetic patients, KP and KI-s compared with KD resulted in enhanced kidney graft survival, hypertrophy, and vascular function.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Diabetic Nephropathies/surgery , Graft Survival/physiology , Islets of Langerhans Transplantation/physiology , Kidney Failure, Chronic/surgery , Kidney Transplantation/physiology , Adult , Biopsy , C-Peptide/blood , Follow-Up Studies , Glycated Hemoglobin/analysis , Humans , Hypertrophy , Immunosuppression Therapy/methods , Islets of Langerhans Transplantation/mortality , Islets of Langerhans Transplantation/pathology , Kidney Transplantation/mortality , Kidney Transplantation/pathology , Survival Analysis , Time Factors , Treatment Outcome
10.
Diabetes ; 53(9): 2291-300, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331538

ABSTRACT

To evaluate the effects of kidney-pancreas transplantation on hemostatic abnormalities in uremic type 1 diabetic patients, we conducted a cross-sectional study involving 12 type 1 diabetic patients, 30 uremic type 1 diabetic patients, 27 uremic type 1 diabetic patients who had a kidney-pancreas transplant, 12 uremic type 1 diabetic patients who had a kidney-alone transplant, and 13 healthy control subjects. We evaluated platelet and clotting system. Platelets in the group of uremic type 1 diabetic patients were significantly larger than platelets in the other groups. Resting calcium levels were significantly higher in the uremic type 1 diabetic patients and uremic type 1 diabetic patients who had a kidney-alone transplant than in the type 1 diabetic patients who had a kidney-pancreas transplant and control subjects. CD41 expression was significantly reduced in platelets from the uremic type 1 diabetic patients compared with the other groups. Levels of hypercoagulability markers in the type 1 diabetic patients who had a kidney-pancreas transplant and, to a lesser extent, the uremic type 1 diabetic patients who had a kidney-alone transplant but not the uremic type 1 diabetic patients were similar to those of the control subjects. A reduction in natural anticoagulants was evident in the uremic type 1 diabetic patients, whereas near-normal values were observed in the type 1 diabetic patients who had a kidney-pancreas transplant and uremic type 1 diabetic patients who had a kidney-alone transplant. Hemostatic abnormalities were not observed in type 1 diabetic patients who had a kidney-pancreas transplant. This finding might explain the lower cardiovascular death rate observed in type 1 diabetic patients who had a kidney-pancreas transplant compared with uremic type 1 diabetic patients who had a kidney-alone transplant or uremic type 1 diabetic patients.


Subject(s)
Blood Coagulation Disorders/complications , Diabetes Mellitus, Type 1/blood , Kidney Transplantation , Pancreas Transplantation , Uremia/blood , Adult , Blood Coagulation , Blood Coagulation Disorders/blood , Blood Platelets/metabolism , Calcium/metabolism , Cross-Sectional Studies , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/surgery , Diabetic Nephropathies/blood , Diabetic Nephropathies/complications , Diabetic Nephropathies/surgery , Female , Humans , Male , Middle Aged , Partial Thromboplastin Time , Platelet Membrane Glycoprotein IIb/metabolism , Prothrombin Time , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Signal Transduction , Uremia/complications , Uremia/surgery
11.
FASEB J ; 17(10): 1340-2, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12738810

ABSTRACT

Recent evidence suggests that insulin signaling through the insulin receptor A type (Ex11-), regulates insulin gene transcription. Because chronic hyperglycemia negatively affects insulin receptor function and regulates alternative splicing of the insulin receptor, we inquired whether chronic exposure of pancreatic beta-cells to high glucose results in alterations in insulin signaling due to changes in insulin receptor expression and relative abundance of its spliced isoforms. Our results demonstrate that the insulin receptor is localized in insulin secretory vescicles in human pancreatic beta-cells. Furthermore, we find that alterations in insulin expression and secretion caused by chronic exposure to high glucose are paralleled by decreased insulin receptor expression and increased relative abundance of the Ex11+ isoform in both human islets and RIN beta-cells. PDX-1 and HMGI(Y) transcription factors are down-regulated by high glucose. These changes are associated with defects in insulin signaling involving insulin receptor-associated PI 3-kinase/Akt/PHAS-I pathway in RIN beta-cells. Re-expression in RIN beta-cells chronically exposed to high glucose of the Ex11-, but not the Ex11+, isoform restored insulin mRNA expression. These data suggest that changes in early steps of insulin receptor signaling may play a role in determining beta-cell dysfunction caused by chronic hyperglycemia.


Subject(s)
Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Protein Serine-Threonine Kinases , Receptor, Insulin/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , HMGA1a Protein/metabolism , Humans , Insulin/biosynthesis , Insulin Receptor Substrate Proteins , Insulin Secretion , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/drug effects , Models, Biological , Pancreas/chemistry , Pancreas/ultrastructure , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA Splicing , Receptor, Insulin/analysis , Receptor, Insulin/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Transcription, Genetic
12.
Transplantation ; 75(8): 1296-301, 2003 Apr 27.
Article in English | MEDLINE | ID: mdl-12717219

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the effects of islet transplantation on patient survival and diabetic vascular complications. METHODS: Thirty-seven type 1 uremic diabetic kidney transplant patients underwent islet transplantation (KI group). Uremic type 1 diabetic kidney-pancreas (KP group, n=162), kidney-alone (KD group, n=42) transplant patients, and uremic type 1 diabetic patients still on hemodialysis (HD+DM group, n=196) constituted the control groups for survival and endothelial morphology. RESULTS: Patient survival was similar in the KI and KP groups and higher than in the HD+DM group (P<0.05). Patients experiencing long-term islet function (KI-successful [KI-s], n=24) showed a better survival (100%, 100%, and 90%) than those in the KI group who lost islet function (KI-unsuccessful [KI-u], n=13) (84%, 75%, and 45%) at 1, 4 and 7 years, respectively (P=0.02). The cardiovascular death rate for the KI group (18%) was similar to the KD group (19%) but lower when the KI-s group is considered alone (5%), and showed a cardiovascular death rate similar to the KP group (8%). The KI-s group showed a good metabolic profile, with reduction of exogenous insulin requirement and persistent C-peptide secretion, as compared with the KI-u group. The endothelial morphology was evaluated with a skin biopsy obtained in all groups. The KI-s and the KP groups demonstrated decreased signs of endothelial injury compared with the KI-u and HD+DM groups. The KI group showed a better atherothrombotic profile than the HD+DM group, with higher levels of natural anticoagulant protein. CONCLUSIONS: Successful islet transplantation improves survival, atherothrombotic profile, and endothelial morphology in uremic type 1 diabetic kidney transplant patients.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetic Angiopathies/surgery , Islets of Langerhans Transplantation , Kidney Transplantation , Adult , Antithrombin III/analysis , Arteriosclerosis/etiology , Biopsy , Cardiovascular Diseases/mortality , Humans , Kidney/physiopathology , Middle Aged , Protein C/analysis , Risk Factors , Skin/pathology , Survival Analysis , Thrombosis/etiology
13.
Pancreas ; 40(6): 883-95, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21705949

ABSTRACT

OBJECTIVES: Serotonin-producing tumors of the pancreas are rare endocrine neoplasms composed of enterochromaffin (EC) cells that have been mainly described in the literature as case reports. This study analyzes the clinicopathologic features of a series of pancreatic EC cell neoplasms and their similarities to and differences from intestinal EC cell tumors. METHODS: The morphological, immunohistochemical, ultrastructural, and fluorescent in situ hybridization features of 15 pancreatic and 20 intestinal serotonin-producing neoplasms were compared. In addition, we reviewed the literature on pancreatic serotonin-producing tumors to better understand the clinicopathologic features of this rare tumor type. RESULTS: The lack of substance P and acidic fibroblast growth factor immunoreactivity; the low immunohistochemical expression of CDX2, vesicular monoamine transporter 1, connective tissue growth factor, and prostatic acid phosphatase; the lack of S100-positive sustentacular cells; the strong expression of vesicular monoamine transporter 2; and peculiar ultrastructural features characterize pancreatic EC cell tumors and differentiate them from intestinal ones, although both categories show similar chromosome 18 cytogenetic alterations. The review of the literature indicates that pancreatic functioning tumors associated with the carcinoid syndrome arise in younger patients and are larger, more frequently malignant, and more aggressive neoplasms than pancreatic nonfunctioning ones. CONCLUSIONS: Pancreatic EC cell tumors show several different morphological features compared with related intestinal tumors despite similar cytogenetic alterations on chromosome 18.


Subject(s)
Carcinoid Tumor/metabolism , Carcinoid Tumor/pathology , Enterochromaffin Cells/metabolism , Enterochromaffin Cells/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Serotonin/biosynthesis , Adult , Aged , Chromosomes, Human, Pair 18/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Intestinal Neoplasms/genetics , Kaplan-Meier Estimate , Male , Malignant Carcinoid Syndrome/genetics , Malignant Carcinoid Syndrome/metabolism , Malignant Carcinoid Syndrome/pathology , Microscopy, Electron, Transmission , Middle Aged , Pancreatic Neoplasms/genetics
14.
PLoS One ; 6(11): e28050, 2011.
Article in English | MEDLINE | ID: mdl-22140505

ABSTRACT

Insulin resistance, reduced ß-cell mass, and hyperglucagonemia are consistent features in type 2 diabetes mellitus (T2DM). We used pancreas and islets from humans with T2DM to examine the regulation of insulin signaling and cell-cycle control of islet cells. We observed reduced ß-cell mass and increased α-cell mass in the Type 2 diabetic pancreas. Confocal microscopy, real-time PCR and western blotting analyses revealed increased expression of PCNA and down-regulation of p27-Kip1 and altered expression of insulin receptors, insulin receptor substrate-2 and phosphorylated BAD. To investigate the mechanisms underlying these findings, we examined a mouse model of insulin resistance in ß-cells--which also exhibits reduced ß-cell mass, the ß-cell-specific insulin receptor knockout (ßIRKO). Freshly isolated islets and ß-cell lines derived from ßIRKO mice exhibited poor cell-cycle progression, nuclear restriction of FoxO1 and reduced expression of cell-cycle proteins favoring growth arrest. Re-expression of insulin receptors in ßIRKO ß-cells reversed the defects and promoted cell cycle progression and proliferation implying a role for insulin-signaling in ß-cell growth. These data provide evidence that human ß- and α-cells can enter the cell-cycle, but proliferation of ß-cells in T2DM fails due to G1-to-S phase arrest secondary to defective insulin signaling. Activation of insulin signaling, FoxO1 and proteins in ß-cell-cycle progression are attractive therapeutic targets to enhance ß-cell regeneration in the treatment of T2DM.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Receptor, Insulin/metabolism , Signal Transduction , Aged , Aged, 80 and over , Animals , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , G1 Phase/genetics , Gene Expression Regulation , Humans , Insulin/metabolism , Male , Mice , Middle Aged , Models, Biological , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Insulin/deficiency , S Phase/genetics , Signal Transduction/genetics , Tissue Donors
15.
Virchows Arch ; 457(4): 497-507, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20694477

ABSTRACT

Thyroid transcription factor-1 (TTF1) regulates lung morphogenesis and differentiation, and its immunohistochemical expression is used for identifying lung neoplasms. The 8G7G3/1 antibody has been used in previous studies, but a different and more sensitive anti-TTF1 antibody, named SPT24, has become commercially available. Since the immunohistochemical expression of TTF1 in normal lung neuroendocrine (NE) cells has not been previously investigated and its utility in the diagnosis of lung NE tumors is a controversial issue, we studied the TTF1 expression in normal adult and fetal lungs, in 83 pulmonary NE neoplasms, in 131 non-lung NE tumors and in 36 metastases from these neoplasms using these two antibodies. A TTF1 immunoreactivity was demonstrated in normal fetal and adult NE cells when using the SPT24 clone. Conversely, using the 8G7G3/1 antibody, only rare fetal neuroendocrine cells were TTF1 positive while adult NE cells were negative. The SPT24 clone identified TTF1 expression in more carcinoids, most of them peripherally located, and poorly differentiated NE carcinomas than the 8G7G3/1 clone. Non-pulmonary well-differentiated NE tumors were negative for both antibodies. Among the 45 non-pulmonary poorly differentiated NE carcinomas 11% were positive for 8G7G3/1 and 18% for SPT24. TTF1 expression in metastases perfectly reflected that detected in the related primary tumors. Our results indicate that the SPT24 antibody is more sensitive than the 8G7G3/1 clone for labeling lung carcinoids and it appears particularly useful in detecting peripheral neoplasms. In addition, the expression of TTF1 in normal NE cells suggests a possible role for the transcription factor in their development and differentiation.


Subject(s)
Antibodies, Monoclonal/immunology , DNA-Binding Proteins/analysis , Lung Neoplasms/chemistry , Lung/chemistry , Neuroendocrine Cells/chemistry , Adult , Fetus/chemistry , Humans , Immunohistochemistry , Neuroendocrine Tumors/chemistry , Transcription Factors
16.
J Histochem Cytochem ; 58(10): 881-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20566755

ABSTRACT

Carboxyl ester lipase (CEL) is an enzyme that hydrolyzes a wide variety of lipid substrates, including ceramides, which are known to show inhibitory regulation of pituitary hormone secretion in experimental models. Because no studies on CEL expression in human pituitary and pituitary adenomas have been reported in the literature, we investigated CEL expression in 10 normal pituitary glands and 86 well-characterized pituitary adenomas [12 FSH/LH cell, 17 α-subunit/null cell, 6 TSH cell, 21 ACTH cell, 11 prolactin (PRL) cell, and 19 GH cell adenomas] using IHC, immunoelectron microscopy, Western blotting, and quantitative RT-PCR. In normal adenohypophysis, CEL was localized in GH, ACTH, and TSH cells. In adenomas, it was mainly found in functioning GH, ACTH, and TSH tumors, whereas its expression was poor in the corresponding silent adenomas and was lacking in FSH/LH cell, null cell, and PRL cell adenomas. Ultrastructurally, CEL was localized in secretory granules close to their membranes. This is the first study demonstrating CEL expression in normal human pituitary glands and in functioning GH, ACTH, and TSH adenomas. Considering that CEL hydrolyzes ceramides, inactivating their inhibitory function on pituitary hormone secretion, our findings suggest a possible role of CEL in the regulation of hormone secretion in both normal and adenomatous pituitary cells.


Subject(s)
Adenoma/enzymology , Lipase/biosynthesis , Pituitary Gland/enzymology , Pituitary Neoplasms/enzymology , Cell Line , Cell Line, Tumor , Humans , Immunohistochemistry , Microscopy, Immunoelectron , Reverse Transcriptase Polymerase Chain Reaction
17.
PLoS One ; 5(3): e9923, 2010 Mar 29.
Article in English | MEDLINE | ID: mdl-20360867

ABSTRACT

BACKGROUND: In type 1 diabetes (T1D) vascular complications such as accelerated atherosclerosis and diffused macro-/microangiopathy are linked to chronic hyperglycemia with a mechanism that is not yet well understood. End-stage renal disease (ESRD) worsens most diabetic complications, particularly, the risk of morbidity and mortality from cardiovascular disease is increased several fold. METHODS AND FINDINGS: We evaluated protein regulation and expression in skin biopsies obtained from T1D patients with and without ESRD, to identify pathways of persistent cellular changes linked to diabetic vascular disease. We therefore examined pathways that may be normalized by restoration of normoglycemia with kidney-pancreas (KP) transplantation. Using proteomic and ultrastructural approaches, multiple alterations in the expression of proteins involved in oxidative stress (catalase, superoxide dismutase 1, Hsp27, Hsp60, ATP synthase delta chain, and flavin reductase), aerobic and anaerobic glycolysis (ACBP, pyruvate kinase muscle isozyme, and phosphoglycerate kinase 1), and intracellular signaling (stratifin-14-3-3, S100-calcyclin, cathepsin, and PPI rotamase) as well as endothelial vascular abnormalities were identified in T1D and T1D+ESRD patients. These abnormalities were reversed after KP transplant. Increased plasma levels of malondialdehyde were observed in T1D and T1D+ESRD patients, confirming increased oxidative stress which was normalized after KP transplant. CONCLUSIONS: Our data suggests persistent cellular changes of anti-oxidative machinery and of aerobic/anaerobic glycolysis are present in T1D and T1D+ESRD patients, and these abnormalities may play a key role in the pathogenesis of hyperglycemia-related vascular complications. Restoration of normoglycemia and removal of uremia with KP transplant can correct these abnormalities. Some of these identified pathways may become potential therapeutic targets for a new generation of drugs.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Gene Expression Regulation , Kidney Transplantation/methods , Oxygen/chemistry , Pancreas Transplantation/methods , Proteomics/methods , Skin/metabolism , Adult , Case-Control Studies , Diabetes Mellitus, Type 1/therapy , Female , Glycolysis , Humans , Kidney Failure, Chronic/metabolism , Male , Middle Aged , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Leuk Lymphoma ; 49(7): 1321-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18604721

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease. The immunohistochemistry-based algorithms for the determination of the cell of origin of DLBCL have been proposed as a practical method to validate and surrogate results obtained by gene expression profiling. We studied 71 patients with primary nodal DLBCL at diagnosis, who received anthracycline-based therapy with or without rituximab. Immunohistochemistry was performed using anti-CD10, Bcl-6, MUM1 and Bcl-2 antibodies in order to assess the ontogenic profile of neoplastic cells and to verify its relation with clinical outcome. Survival data were analysed using an explorative Cox model. The immunohistochemistry-based algorithms for the determination of the cell of origin of DLBCL were not associated with prognosis. By contrast, Bcl-6 expression was associated with a longer lymphoma-free survival while immunoreactivities for MUM1 or Bcl-2 were not significantly related to patient outcome. Bcl-6 expression alone proved to be a prognostic marker in primary nodal DLBCL and seemed to be more reliable to predict clinical outcome in these disorders than the immunohistochemical algorithms for the detection of the germinal centre/non-germinal centre immunophenotype.


Subject(s)
Germinal Center/pathology , Lymphoma, Large B-Cell, Diffuse/diagnosis , Predictive Value of Tests , Proto-Oncogene Proteins c-bcl-6/analysis , Adult , Aged , Aged, 80 and over , Algorithms , Anthracyclines/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Disease-Free Survival , Female , Humans , Immunophenotyping , Interferon Regulatory Factors/analysis , Lymphoma, Large B-Cell, Diffuse/drug therapy , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins c-bcl-2/analysis , Rituximab , Treatment Outcome
19.
Ultrastruct Pathol ; 29(6): 525-33, 2005.
Article in English | MEDLINE | ID: mdl-16316953

ABSTRACT

Islet transplantation is a new therapeutic approach to type 1 diabetes mellitus. However, in several patients insulin levels are not restored and the glycemic control is inadequate. To clarify the cause of graft failure, the authors investigated with light and electron microscopy some human islet grafts before and after transplantation under the kidney capsule of streptozotocin-induced diabetic nude mice. In isolated islets, both pre- and post-transplantation, the endocrine component was scarcely represented, the beta/alpha cell ratio was reduced, and beta cells showed degenerative aspects such as apoptosis, immature secretory granules, and amylin fibrils deposition. The authors conclude that islet graft failure may be due to an insufficient beta cell mass related to their distress probably caused by anoxia and/or overstimulation.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Graft Rejection/pathology , Islets of Langerhans Transplantation , Islets of Langerhans/ultrastructure , Animals , Humans , Mice , Mice, Nude , Microscopy, Electron, Transmission
20.
Transpl Int ; 18(9): 1036-47, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16101724

ABSTRACT

End-stage renal disease (ESRD) is characterized by several atherothrombotic abnormalities, and kidney transplant seems to improve most of them. However, because it is not clear which mechanism is responsible for such improvement, our purpose was to clarify that point.We conducted a cross sectional study involving 30 ESRD patients, 30 ESRD kidney-transplanted patients (Ktx) and 30 healthy controls (C) to evaluate platelet morphology and function, atherothrombotic profile, endothelial abnormalities and cytokine levels involved in the insulin resistance/endothelial dysfunction. (i) Platelet morphology: The ESRD group showed platelet size similar to the other two groups (ESRD=3518x10(3)+/-549x10(3) nm2, C=3075x10(3)+/-197x10(3) nm2, Ktx=2862x10(3)+/-205x10(3) nm2) with similar platelet granules and number. (ii) Platelet surface glycoprotein: The CD41 and P-Selectin were similar between groups. (iii) Platelet intracellular calcium: Resting intracellular calcium was statistically higher in ESRD compared to the C group (ESRD=182.1+/-34.5, Ktx=126.7+/-14.1, C=72.0+/-11.0 nM, P<0.01). (iv) Hypercoagulability markers and natural anticoagulants: The Ktx and ESRD groups showed higher levels of hypercoagulability markers compared to the C group. A reduction in antithrombin activity was evident in ESRD compared to the Ktx group (P=0.03). (v) Endothelial morphology: The ESRD group showed a thickened vessel basal membrane compared to the Ktx and C groups with more endothelial sufference. (vi) Insulin resistance and pro-inflammatory cytokine profile: The ESRD showed a higher homeostasis model assessment provided equations for estimating insulin resistance (HOMA-IR) compared to the Ktx and C groups (ESRD=2.6+/-0.3, Ktx=1.8+/-0.2, C=1.1+/-0.1, P=0.005) and increased soluble tumor neurosis factor alpha (sTNFalpha) (P<0.05) and soluble vascular cell adhesion molecule (sVCAM) levels (P<0.01). Positive correlations were evident among HOMA-IR and sTNFalpha (P<0.001) and sVCAM (P=0.01), respectively. In a small subgroup of ESRD who underwent Ktx (five pts), our findings were confirmed at 1 year of follow-up, suggesting an improvement of almost haemostatic abnormalities. Kidney transplant is associated with a better atherothrombotic profile in ESRD, platelet intracellular calcium and cytokines seem to be most influenced by the transplant, while most morphological abnormalities are retained.


Subject(s)
Hemostasis , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/surgery , Kidney Transplantation , Adult , Blood Platelets/pathology , Blood Platelets/ultrastructure , Calcium/blood , Cross-Sectional Studies , Endothelium, Vascular/pathology , Humans , Insulin Resistance , Interleukin-18/blood , Kidney Failure, Chronic/pathology , Kidney Failure, Chronic/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL