Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Acta Diabetol ; 56(9): 1013-1022, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30989379

ABSTRACT

AIM: More than 40% of pancreatic ductal adenocarcinoma (PDAC) patients have glucose intolerance or diabetes. The association has led to two hypotheses: PDAC causes diabetes or diabetes shares risk factors for the development of PDAC. In order to elucidate the relationship between diabetes and PDAC, we investigated the glucose metabolism during tumorigenesis in the LSL-KrasG12D/+; LSL-Trp53R172H/+; and Pdx-1-Cre (KPC) mouse, a genetically engineered model of PDAC. METHODS: Male and female KPCs have been fed with standard diet (SD) or high-fat diet (HFD). The imaging-based 4-class tumor staging was used to follow pancreatic cancer development. Not fasting glycemia, 4-h fasting glycemia, insulin, C-peptide, glucose tolerance after OGTT and abdominal fat volume were measured during tumorigenesis. RESULTS: PDAC development did not lead to an overt diabetic phenotype or to any alterations in glucose tolerance in KPC fed with SD. Consumption of HFD induced higher body weight/abdominal fat volume and worsened glucose homeostasis both in control CRE mice and only in early tumorigenesis stages of the KPC mice, excluding that the cancer development itself acts as a trigger for the onset of dysmetabolic features. CONCLUSION: Our data demonstrate that carcinogenesis in KPC mice is not associated with paraneoplastic diabetes.


Subject(s)
Carbohydrate Metabolism/physiology , Carcinogenesis/metabolism , Glucose/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Carbohydrate Metabolism/genetics , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/metabolism , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Staging , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , Pancreatic Neoplasms
2.
Pancreatology ; 18(8): 945-953, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30293872

ABSTRACT

BACKGROUND: Despite the recent introduction of new drugs and the development of innovative multi-target treatments, the prognosis of pancreatic ductal adenocarcinoma (PDAC) remains very poor. Even when PDAC is resectable, the rate of local or widespread disease recurrence remains particularly high. Currently, reliable prognostic biomarkers of recurrence are lacking. We decided to explore the potential usefulness of pancreatic developmental regulators as biomarkers of PDAC relapse. METHODS: We analyzed by quantitative real-time PCR the mRNA of selected factors involved either in pancreatic organogenesis (ISL1, NEUROD1, NGN3, NKX2.2, NKX6.1, PAX4, PAX6, PDX1 and PTF1α) or associated with terminally committed pancreatic cells (CHGA, CHGB, GAD2, GCG, HNF6α, INS, KRT19, SYP) in 17 PDAC cell lines and in frozen tumor samples from 41 PDAC patients. RESULTS: High baseline levels of the ISL1, KRT19, PAX6 and PDX1 mRNAs in PDAC cell lines, were risk factors for time-dependent xenograft appearance after subcutaneous injection in CD1-Nude mice. Consistently, in human PDAC samples, high levels of KRT19 mRNA were associated with reduced overall survival and earlier recurrence. Higher levels of PDX1 or PAX6 mRNAs were instead associated with a higher frequency of local recurrence. CONCLUSIONS: Our findings suggest that selected factors associated with pancreas development or its terminal differentiation might be implicated in mechanisms of PDAC progression and/or metastatic spread and that the measurement of their mRNA in tumors might be potentially used to improve patient prognostic stratification and prediction of the relapse site.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/surgery , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Pancreas/embryology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/surgery , Adult , Aged , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Female , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Keratin-19/genetics , Male , Mice , Mice, Nude , Middle Aged , Nuclear Proteins , Organogenesis/genetics , Prognosis , Real-Time Polymerase Chain Reaction , Survival Analysis , Transcription Factors , Xenograft Model Antitumor Assays
3.
Carcinogenesis ; 39(9): 1197-1206, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30052815

ABSTRACT

Background: The widely used genetically engineered mouse LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre, termed KPC, spontaneously develops pancreatic cancer mirroring all phases of the carcinogenesis but in asynchronous manner. Preclinical studies need defined criteria for the enrollment of the KPC sharing the same stage of carcinogenesis. Aim: To define a tumor-staging criteria using magnetic resonance (MR) and ultrasound (US) and then to correlate the imaging stage with overall survival of KPC mice. Methods: Forty KPC (2- to 5-month-old mice) were imaged by axial fat-saturated T2-weighted sequences at MR and by brightness mode US to establish criteria for tumor staging. Immunohistopathology was used to validate imaging. A second cohort of 25 KPC was used to correlate imaging stage with survival by Kaplan-Meier analysis. Results: We defined a four-class tumor staging system ranking from stages 1 to 4. Stage 1 was described as radiologically healthy pancreas; precursor lesions were detectable in histology only. Cystic papillary neoplasms, besides other premalignant alterations, marked stage 2 in the absence of cancer nodules. Stages 3 and 4 identified mice affected by overt pancreatic cancer with size <5 or ≥5 mm, respectively. Regarding the prognosis, this staging system correlated with disease-related mortality whatever may be the KPC age when they staged. Conclusion: This imaging-based four-class tumor staging is an effective and safe method to stage pancreatic cancer development in KPC. As a result, regardless of their age, KPC mice can be synchronized based on prognosis or on a specific phase of tumorigenesis, such as the early but already radiologically detectable one (stage 2).


Subject(s)
Magnetic Resonance Imaging/methods , Pancreatic Neoplasms , Ultrasonography/methods , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Staging/methods , Pancreas/physiology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Precancerous Conditions/pathology
4.
Cancer Biomark ; 22(2): 351-357, 2018.
Article in English | MEDLINE | ID: mdl-29660901

ABSTRACT

BACKGROUND: Sensitive and specific biomarkers of Pancreatic Ductal Adenocarcinoma (PDAC) are desperately needed to allow early diagnosis and improve patient's survival. Ezrin autoantibodies were recently described as present in 93% of PDAC patients and 40% of healthy subjects who later developed PDAC. However, another prospective study failed to replicate these findings. Both studies were based on the use of a solid phase ELISA immunoassay. OBJECTIVE: We aimed at re-evaluating the usefulness of Ezrin autoantibodies as PDAC biomarkers using the Luciferase Immuno Precipitation System (LIPS), an alternative immunoassay format that found successful application for the measurement of autoantibodies against pancreatic autoantigens. METHODS: We produced a Nanoluciferase™ tagged Ezrin (NLuc-Ezrin). NLuc-Ezrin was then used as antigen in LIPS to test for Ezrin autoantibodies patients affected by PDAC (n= 40), other pancreatic diseases (OPD, n= 50), and healthy controls (n= 60). RESULTS: Overall, binding in liquid phase to Ezrin by serum antibodies was rare and low titer. Furthermore, we did not find statistically significant differences in the prevalence of Ezrin autoantibodies between patients affected by either PDAC or OPD compared to control. CONCLUSIONS: Our results do not confirm the usefulness of Ezrin autoAbs as biomarker of PDAC.


Subject(s)
Autoantibodies/blood , Autoantibodies/immunology , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/immunology , Cytoskeletal Proteins/immunology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/immunology , Adult , Aged , Autoantigens/immunology , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal/diagnosis , Case-Control Studies , Cloning, Molecular , Cytoskeletal Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Genes, Reporter , Humans , Immunoassay , Immunoprecipitation , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Pancreatic Neoplasms/diagnosis , ROC Curve , Pancreatic Neoplasms
5.
Acta Diabetol ; 55(3): 263-270, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29305766

ABSTRACT

AIMS: Insulin autoantibodies (IAA) are often the first marker of autoimmunity detected in children in the preclinical phase of type 1 diabetes (T1D). Currently, the vast majority of laboratories adopt the radiobinding micro-assay (RBA) for measuring IAA. Our aim was to replace RBA with a novel non-radioactive IAA Luciferase Immuno Precipitation System (LIPS) assay with improved performance. METHODS: We developed (pro)insulin antigens with alternative placements of a NanoLuc™ luciferase reporter (NLuc). Performance in LIPS was evaluated by testing sera from new onset T1D (n = 80), blood donors (n = 123), schoolchildren (n = 186), first-degree relatives (FDRs) from the Bart's Oxford family study (n = 53) and from the Belgian Diabetes Registry (n = 136), coded sera from the Islet Autoantibody Standardization Program (IASP) (T1D n = 50, blood donors n = 90). RESULTS: IAA LIPS based on B chain-NLuc proinsulin or B chain-NLuc insulin, in which NLuc was fused at the C-terminus of the insulin B chain, required only 2 µL of serum and a short incubation time, showed high concordance with RBA (Spearman r = 0.866 and 0.833, respectively), high assay performance (B chain-NLuc proinsulin ROC-AUC = 0.894 and B chain-NLuc insulin ROC-AUC = 0.916), and an adjusted sensitivity at 95% specificity ranking on par with the best assays submitted to the two most recent IASP workshops. In FDRs, the IAA LIPS showed improved discrimination of progressors to T1D compared to RBA. CONCLUSIONS: We established a novel high-performance non-radioactive IAA LIPS that might replace the current gold standard RBA and find wide application in the study of the IAA response in T1D.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Fluorescent Dyes/metabolism , Immunoprecipitation/methods , Insulin Antibodies/analysis , Luciferases/metabolism , Autoantibodies/blood , Biomarkers/analysis , Biomarkers/metabolism , Case-Control Studies , Child , Fluorescent Dyes/analysis , HEK293 Cells , Humans , Insulin/immunology , Insulin Antibodies/blood , Predictive Value of Tests , Prognosis , Proinsulin/immunology , Sensitivity and Specificity
6.
Sci Rep ; 7(1): 14051, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070792

ABSTRACT

miR-204 has been proposed to modulate insulin expression in human pancreatic islets by regulating the expression of the MAFA transcript, and in turn insulin transcription. We investigated miR-204 expression in pancreatic endocrine tumors (PET), a panel of human tissues, tissues derived from pancreatic islet purification, and in induced pluripotent stem cells (iPSCs) differentiated towards a pancreatic endocrine phenotype by quantitative real time RT-PCR or droplet digital PCR (ddPCR). In addition, we evaluated the effect of miR-204 up- or down-regulation in purified human islets and in the EndoC-ßH1 cell line, as an experimental model of human pancreatic ß cells. Our results confirm that miR-204 was enriched in insulin producing PET, in ß cells within healthy pancreatic islets, and highly expressed in EndoC-ßH1 cells. Moreover, in iPSCs miR-204 increased stepwise upon stimulated differentiation to insulin producing cells. However, up- or down-regulation of miR-204 in human islets and in EndoC-ßH1 cells resulted in modest and not significant changes of the MAFA and INS mRNAs measured by ddPCR or c-peptide release. Our data confirm the association of miR-204 with a ß cell endocrine phenotype in human pancreatic islets, but do not support its direct role in regulating the levels of insulin mRNA through MAFA.


Subject(s)
Endocrine Gland Neoplasms/genetics , Induced Pluripotent Stem Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Maf Transcription Factors, Large/metabolism , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , RNA, Messenger/metabolism , Cell Differentiation , Cells, Cultured , Endocrine Gland Neoplasms/metabolism , Endocrine Gland Neoplasms/pathology , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Insulin/genetics , Islets of Langerhans/cytology , Maf Transcription Factors, Large/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenotype , RNA, Messenger/genetics
7.
Dig Liver Dis ; 49(9): 1050-1056, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28734776

ABSTRACT

BACKGROUND: This study aimed to develop and validate a preoperative prognostic model for death within one year post-surgery in patients with resectable pancreatic ductal adenocarcinoma (PDAC). METHODS: A derivation cohort study of 296 patients who underwent surgical resection of PDAC was prospectively enrolled in an observational study. Preoperative predictors of one year mortality were used to develop a risk score which was then validated in an external cohort of 182 patients with resectable PDAC. RESULTS: Seventy-eight out of 296 patients (26%) died within the first year. Preoperative independent predictors of one year mortality were: nutritional status (Geriatric Nutritional Risk Index, OR 2.23, 1.14-4.38; p=0.02), American Society of Anaesthesiologists' score (OR 2.56, 1.1-5.98; p=0.03), abdominal or back pain at presentation (OR 2.51, 1.05-5.9; p=0.038) and non metastatic liver disease as comorbidity (OR 4.5, 1.05-19.3; p=0.043). A score ranging from 0 to 7 points was developed. In the validation cohort, the model was able to predict early mortality (OR 7.1, 3.9-12.7; p<0.0001), with a predictive ability of 53.5% (Nagelkerke R2), an area under the receiver operating characteristic curve of 88.7% and an acceptable calibration (goodness-of-fit test, p=0.403). CONCLUSIONS: Our new simple risk score proved reliable in forecasting one year mortality in patients with resectable PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/surgery , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Aged , Aged, 80 and over , Female , Humans , Italy , Logistic Models , Male , Middle Aged , Multivariate Analysis , Preoperative Period , Prognosis , Prospective Studies , ROC Curve , Risk Factors , Severity of Illness Index , Survival Analysis , Time Factors
9.
PLoS One ; 11(11): e0166008, 2016.
Article in English | MEDLINE | ID: mdl-27814399

ABSTRACT

AIM: To investigate the effect of diabetes mellitus (DM) on disease-free and overall post-resection survival of patients with pancreatic ductal adenocarcinoma (PDAC). METHODS: Prospective observational study on patients admitted for pancreatic disease from January 2008 to October 2012. DM was classified as recent-onset (<48 months before PDAC diagnosis), longstanding (≥48 months before PDAC) or new onset (after surgery). RESULTS: Of 296 patients, 140 had a diagnosis of DM prior to surgery (26 longstanding, 99 recent-onset, 15 with unknown duration). Median follow-up time was 5.4 ± 0.22 years. Patients with recent onset DM had poorer postoperative survival than patients without DM: disease-free survival and overall survival were 1.14±0.13 years and 1.52±0.12 years in recent onset DM, versus 1.3±0.15 years and 1.87±0.15 years in non-diabetic patients (p = 0.013 and p = 0.025, respectively). Longstanding DM and postoperative new onset DM had no impact on prognosis. Compared to cases without DM, patients with recent onset DM were more likely to have residual disease after surgery and to develop liver metastases during follow-up. Multivariate analysis confirmed recent onset DM was independently associated with PDAC relapse (hazard ratio 1.45 [1.06-1.99]). CONCLUSION: Preoperative recent onset DM has an impact on survival after the resection of PDAC.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/surgery , Diabetes Mellitus/mortality , Diabetes Mellitus/physiopathology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Adenocarcinoma/pathology , Aged , Disease-Free Survival , Female , Humans , Male , Pancreatic Neoplasms/pathology , Prognosis , Prospective Studies , Risk Factors , Pancreatic Neoplasms
10.
Acta Diabetol ; 53(6): 945-956, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27552832

ABSTRACT

AIMS: To study whether insulin resistance accelerates the development and/or the progression of pancreatic adenocarcinoma (PDAC), we hypothesized that patients with insulin resistance, compared with those without insulin resistance, show: (1) a younger age and more advanced PDAC stage at diagnosis and (2) a shorter disease-free and overall survival after PDAC diagnosis. METHODS: Prospective observational study of patients admitted to a referral center for pancreatic disease. Insulin resistance was defined as a HOMA-IR value greater than the 66th percentile value of the patients included in this study. Survival was estimated according to Kaplan-Meier and by Cox regression. RESULTS: Of 296 patients with PDAC, 99 (33 %) met criteria for being classified as insulin resistant at diagnosis. Median follow-up time after diagnosis was 5.27 ± 0.23 years. Patients with insulin resistance received a diagnosis of PDAC at a similar age compared to patients without insulin resistance (67.1 ± 9 vs. 66.8 ± 10 years, p = 0.68), but were more likely to have a cancer stage ≥3 (23.2 vs. 14.2 %, p = 0.053) and a residual disease after surgery (R1 56.4 vs. 38 %; p = 0.007). The median overall survival was 1.3 ± 0.14 and 1.79 ± 0.11 years for the patients with and without insulin resistance, respectively (p = 0.016). Results did not change when patients with diabetes at PDAC diagnosis were excluded from the analysis. Multivariate analysis showed that insulin resistance was independently associated with overall survival. CONCLUSIONS: Insulin resistance is associated with the aggressiveness of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Insulin/metabolism , Pancreatic Neoplasms , Age Factors , Aged , Carcinoma, Pancreatic Ductal/epidemiology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Disease Progression , Female , Humans , Insulin Resistance , Italy/epidemiology , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prospective Studies , Survival Analysis
11.
Pancreatology ; 16(5): 844-52, 2016.
Article in English | MEDLINE | ID: mdl-27546476

ABSTRACT

BACKGROUND: Identification of a specific diabetes signature associated to pancreatic ductal carcinoma (PDAC) could be a key to detect asymptomatic, early stage tumors. We aim to characterize the clinical signature and the pathogenetic factors of the different types of diabetes associated with PDAC, based on the time between diabetes and cancer diagnosis. METHODS: Prospective observational study on 364 PDAC patients admitted to a referral center for pancreatic disease. Hospital and/or outpatient medical records were reviewed. Blood biochemical values including fasting blood glucose, insulin and/or C-peptide, glycosylated hemoglobin and anti-islet antibodies were determined. Diabetes onset was assessed after surgery and during follow-up. RESULTS: The prevalence of diabetes in patients was 67%. Considering 174 patients (47.8%) already having diabetes when diagnosed with PDAC (long duration, short duration, concomitant), the clinical and biochemical profile was similar to that of patients with type 2 diabetes (T2D). Diabetes was associated with known risk factors (i.e., age, sex, family history for diabetes and increased BMI) and both beta-cell dysfunction and insulin resistance were present. Considering 70 patients (19.2%) with onset of diabetes after PDAC diagnosis (early and late onset), the strongest predictor was the loss of beta-cell mass following pancreatectomy in patients with risk factors for T2D. CONCLUSION: Different types of diabetes according to the time between diabetes and PDAC diagnosis are clinical entities widely overlapping with T2D. Therefore, the success of a strategy considering diabetes onset as a marker of asymptomatic PDAC will largely depend on our ability to identify new diabetes-unrelated biomarkers of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/surgery , Diabetes Complications/complications , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/surgery , Aged , Aged, 80 and over , Blood Glucose/analysis , Carcinoma, Pancreatic Ductal/epidemiology , Diabetes Complications/epidemiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Female , Follow-Up Studies , Humans , Insulin/blood , Male , Middle Aged , Pancreatectomy , Pancreatic Neoplasms/epidemiology , Prevalence , Prospective Studies , Risk Factors
12.
Stem Cells Transl Med ; 5(11): 1525-1537, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27405779

ABSTRACT

: ß-Cell replacement therapy represents the most promising approach to restore ß-cell mass and glucose homeostasis in patients with type 1 diabetes. Safety and ethical issues associated with pluripotent stem cells stimulated the search for adult progenitor cells with endocrine differentiation capacities. We have already described a model for expansion and differentiation of human pancreatic duct-derived cells (HDDCs) into insulin-producing cells. Here we show an innovative and robust in vitro system for large-scale production of ß-like cells from HDDCs using a nonintegrative RNA-based reprogramming technique. Synthetic modified RNAs for pancreatic transcription factors (pancreatic duodenal homeobox 1, neurogenin3, and V-Maf musculoaponeurotic fibrosarcoma oncogene homolog A [MAFA]) were manufactured and daily transfected in HDDCs without strongly affecting immune response and cell viability. MAFA overexpression was efficient and sufficient to induce ß-cell differentiation of HDDCs, which acquired a broad repertoire of mature ß-cell markers while downregulating characteristic epithelial-mesenchymal transition markers. Within 7 days, MAFA-reprogrammed HDDC populations contained 37% insulin-positive cells and a proportion of endocrine cells expressing somatostatin and pancreatic polypeptide. Ultrastructure analysis of differentiated HDDCs showed both immature and mature insulin granules with light-backscattering properties. Furthermore, in vitro HDDC-derived ß cells (called ß-HDDCs) secreted human insulin and C-peptide in response to glucose, KCl, 3-isobutyl-1-methylxanthine, and tolbutamide stimulation. Transplantation of ß-HDDCs into diabetic SCID-beige mice confirmed their functional glucose-responsive insulin secretion and their capacity to mitigate hyperglycemia. Our data describe a new, reliable, and fast procedure in adult human pancreatic cells to generate clinically relevant amounts of new ß cells with potential to reverse diabetes. SIGNIFICANCE: ß-Cell replacement therapy represents the most promising approach to restore glucose homeostasis in patients with type 1 diabetes. This study shows an innovative and robust in vitro system for large-scale production of ß-like cells from human pancreatic duct-derived cells (HDDCs) using a nonintegrative RNA-based reprogramming technique. V-Maf musculoaponeurotic fibrosarcoma oncogene homolog A overexpression was efficient and sufficient to induce ß-cell differentiation and insulin secretion from HDDCs in response to glucose stimulation, allowing the cells to mitigate hyperglycemia in diabetic SCID-beige mice. The data describe a new, reliable, and fast procedure in adult human pancreatic cells to generate clinically relevant amounts of new ß cells with the potential to reverse diabetes.

13.
Curr Diab Rep ; 16(6): 53, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27112957

ABSTRACT

Islet autoantibodies are the main markers of pancreatic autoimmunity in type 1 diabetes (T1D). Islet autoantibodies recognize insulin (IAA), glutamic acid decarboxylase (GADA), protein phosphatase-like IA-2 (IA-2A), and ZnT8 (ZnT8A), all antigens that are found on secretory granules within pancreatic beta cells. Islet antibodies, measured by sensitive and specific liquid phase assays, are the key parameters of the autoimmune response monitored for diagnostics or prognostics in patients with T1D or for disease prediction in at-risk individuals before T1D onset. Islet autoantibodies have been the main tool used to explore the natural history of T1D; this review summarizes the current knowledge about the autoantigens and the phenotype of islets autoantibodies acquired in large prospective studies from birth in children at risk of developing T1D.


Subject(s)
Autoantibodies/immunology , Islets of Langerhans/immunology , Animals , Autoantigens/immunology , Biomarkers/analysis , Diabetes Mellitus, Type 1/immunology , Humans , Insulin/immunology
14.
Diabetes ; 65(6): 1690-8, 2016 06.
Article in English | MEDLINE | ID: mdl-26953162

ABSTRACT

The presence of autoantibodies to multiple-islet autoantigens confers high risk for the development of type 1 diabetes. Four major autoantigens are established (insulin, glutamate decarboxylase, IA2, and zinc transporter-8), but the molecular identity of a fifth, a 38-kDa membrane glycoprotein (Glima), is unknown. Glima antibodies have been detectable only by immunoprecipitation from extracts of radiolabeled islet or neuronal cells. We sought to identify Glima to enable efficient assay of these autoantibodies. Mouse brain and lung were shown to express Glima. Membrane glycoproteins from extracts of these organs were enriched by detergent phase separation, lectin affinity chromatography, and SDS-PAGE. Proteins were also immunoaffinity purified from brain extracts using autoantibodies from the sera of patients with diabetes before SDS-PAGE. Eluates from gel regions equivalent to 38 kDa were analyzed by liquid chromatography-tandem mass spectrometry for protein identification. Three proteins were detected in samples from the brain and lung extracts, and in the immunoaffinity-purified sample, but not in the negative control. Only tetraspanin-7, a multipass transmembrane glycoprotein with neuroendocrine expression, had physical characteristics expected of Glima. Tetraspanin-7 was confirmed as an autoantigen by demonstrating binding to autoantibodies in type 1 diabetes. We identify tetraspanin-7 as a target of autoimmunity in diabetes, allowing its exploitation for diabetes prediction and immunotherapy.


Subject(s)
Autoantibodies/immunology , Diabetes Mellitus, Type 1/immunology , Membrane Glycoproteins/immunology , Tetraspanins/immunology , Adolescent , Adult , Animals , Autoantibodies/blood , Autoantigens/immunology , Brain/immunology , Humans , Lung/immunology , Mice , Middle Aged
15.
Clin Cancer Res ; 22(5): 1076-85, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26459175

ABSTRACT

PURPOSE: We aimed to assess the safety and efficacy of metformin for treating patients with metastatic pancreatic cancer and to identify endocrine and metabolic phenotypic features or tumor molecular markers associated with sensitivity to metformin antineoplastic action. EXPERIMENTAL DESIGN: We designed an open-label, randomized, phase II trial to assess the efficacy of adding metformin to a standard systemic therapy with cisplatin, epirubicin, capecitabine, and gemcitabine (PEXG) in patients with metastatic pancreatic cancer. Patients ages 18 years or older with metastatic pancreatic cancer were randomly assigned (1:1) to receive PEXG every 4 weeks in combination or not with 2 g oral metformin daily. The primary endpoint was 6-months progression-free survival (PFS-6) in the intention-to-treat population. RESULTS: Between August 2010 and January 2014, we randomly assigned 60 patients to receive PEXG with (n = 31) or without metformin (n = 29). At the preplanned interim analysis, the study was ended for futility. PFS-6 was 52% [95% confidence interval (CI), 33-69] in the control group and 42% (95% CI, 24-59) in the metformin group (P = 0.61). Furthermore, there was no difference in disease-free survival and overall survival between groups. Despite endocrine metabolic modifications induced by metformin, there was no correlation with the outcome. Single-nucleotide polymorphism rs11212617 predicted glycemic response, but not tumor response to metformin. Gene expression on tumor tissue did not predict tumor response to metformin. CONCLUSIONS: Addition of metformin at the dose commonly used in diabetes did not improve outcome in patients with metastatic pancreatic cancer treated with standard systemic therapy. See related commentary by Yang and Rustgi, p. 1031.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Metformin/administration & dosage , Pancreatic Neoplasms/drug therapy , Adult , Aged , Capecitabine/administration & dosage , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Epirubicin/administration & dosage , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Pancreatic Neoplasms/pathology , Treatment Outcome , Gemcitabine
16.
Curr Diab Rep ; 15(4): 16, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25702096

ABSTRACT

In the developed world, pancreatic surgery is becoming more common, with an increasing number of patients developing diabetes because of either partial or total pancreatectomy, with a significant impact on quality of life and survival. Although these patients are expected to consume increasing health care resources in the near future, many aspects of diabetes after pancreatectomy are still not well defined. The treatment of diabetes in these patients takes advantage of the therapies used in type 1 and 2 diabetes; however, no specific guidelines for its management, both immediately after pancreatic surgery or in the long term, have been developed. In this article, on the basis of both the literature and our clinical experience, we address the open issues and discuss the most appropriate therapeutic options for patients with diabetes after pancreatectomy.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 2/etiology , Pancreas/surgery , Pancreatectomy/adverse effects , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/therapy , Humans , Incidence , Prevalence
17.
Diabetes ; 64(4): 1329-40, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25315007

ABSTRACT

Chemokines and their receptors have been associated with or implicated in the pathogenesis of type 1 diabetes (T1D), but the identification of a single specific chemokine/receptor pathway that may constitute a suitable target for the development of therapeutic interventions is still lacking. Here, we used multiple low-dose (MLD) streptozotocin (STZ) injections and the NOD mouse model to investigate the potency of CXCR1/2 inhibition to prevent inflammation- and autoimmunity-mediated damage of pancreatic islets. Reparixin and ladarixin, noncompetitive allosteric inhibitors, were used to pharmacologically blockade CXCR1/2. Transient blockade of said receptors was effective in preventing inflammation-mediated damage in MLD-STZ and in preventing and reversing diabetes in NOD mice. Blockade of CXCR1/2 was associated with inhibition of insulitis and modification of leukocytes distribution in blood, spleen, bone marrow, and lymph nodes. Among leukocytes, CXCR2(+) myeloid cells were the most decreased subpopulations. Together these results identify CXCR1/2 chemokine receptors as "master regulators" of diabetes pathogenesis. The demonstration that this strategy may be successful in preserving residual ß-cells holds the potential to make a significant change in the approach to management of human T1D.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Islets of Langerhans/drug effects , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Sulfonamides/therapeutic use , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...