Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Transplantation ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38637923

ABSTRACT

BACKGROUND: Pancreatogenic diabetes, a consequence of pancreatic tissue loss following pancreatectomy, poses a significant challenge for patients undergoing pancreatic surgery. Islet autotransplantation (IAT) offers a promising approach to prevent or alleviate pancreatogenic diabetes, but its application has been limited to individuals with painful chronic pancreatitis. METHODS: This study presents a 15-y clinical experience with the Milan Protocol, which expands IAT after pancreatectomy to a broader spectrum of patients with malignant and nonmalignant pancreatic diseases. The analysis evaluates feasibility, efficacy, and safety of IAT. Modified Igls criteria validated through the arginine test and mixed meal tolerance tests were used to assess long-term metabolic outcomes. RESULTS: Between November 2008 and June 2023, IAT procedures were performed on 114 of 147 candidates. IAT-related complications occurred in 19 of 114 patients (16.7%), with 5 being potentially serious. Patients exhibited sustained C-peptide secretion over the 10-y follow-up period, demonstrating a prevalence of optimal and good beta-cell function. Individuals who underwent partial pancreatectomy demonstrated superior metabolic outcomes, including sustained C-peptide secretion and a reduced risk of developing diabetes or insulin dependence compared with those who underwent total pancreatectomy. For patients who had total pancreatectomy, the quantity of infused islets and tissue volume were identified as critical factors influencing metabolic outcomes. An increased risk of recurrence or progression of baseline diseases was not observed in subjects with neoplasms. CONCLUSIONS: These findings provide valuable insights into the benefits and applications of IAT as a therapeutic option for pancreatogenic diabetes after pancreatic surgery, expanding its potential beyond painful chronic pancreatitis.

2.
Front Immunol ; 14: 1323439, 2023.
Article in English | MEDLINE | ID: mdl-38077372

ABSTRACT

Type 1 diabetes (T1D) presents a persistent medical challenge, demanding innovative strategies for sustained glycemic control and enhanced patient well-being. Beta cells are specialized cells in the pancreas that produce insulin, a hormone that regulates blood sugar levels. When beta cells are damaged or destroyed, insulin production decreases, which leads to T1D. Allo Beta Cell Transplantation has emerged as a promising therapeutic avenue, with the goal of reinstating glucose regulation and insulin production in T1D patients. However, the path to success in this approach is fraught with complex immunological hurdles that demand rigorous exploration and resolution for enduring therapeutic efficacy. This exploration focuses on the distinct immunological characteristics inherent to Allo Beta Cell Transplantation. An understanding of these unique challenges is pivotal for the development of effective therapeutic interventions. The critical role of glucose regulation and insulin in immune activation is emphasized, with an emphasis on the intricate interplay between beta cells and immune cells. The transplantation site, particularly the liver, is examined in depth, highlighting its relevance in the context of complex immunological issues. Scrutiny extends to recipient and donor matching, including the utilization of multiple islet donors, while also considering the potential risk of autoimmune recurrence. Moreover, unanswered questions and persistent gaps in knowledge within the field are identified. These include the absence of robust evidence supporting immunosuppression treatments, the need for reliable methods to assess rejection and treatment protocols, the lack of validated biomarkers for monitoring beta cell loss, and the imperative need for improved beta cell imaging techniques. In addition, attention is drawn to emerging directions and transformative strategies in the field. This encompasses alternative immunosuppressive regimens and calcineurin-free immunoprotocols, as well as a reevaluation of induction therapy and recipient preconditioning methods. Innovative approaches targeting autoimmune recurrence, such as CAR Tregs and TCR Tregs, are explored, along with the potential of stem stealth cells, tissue engineering, and encapsulation to overcome the risk of graft rejection. In summary, this review provides a comprehensive overview of the inherent immunological obstacles associated with Allo Beta Cell Transplantation. It offers valuable insights into emerging strategies and directions that hold great promise for advancing the field and ultimately improving outcomes for individuals living with diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Insulins , Islets of Langerhans Transplantation , Humans , Insulin-Secreting Cells/metabolism , Glucose
3.
Ann Surg ; 277(6): 894-903, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36177837

ABSTRACT

OBJECTIVE: To compare pancreaticoduodenectomy (PD) and total pancreatectomy (TP) with islet autotransplantation (IAT) in patients at high risk of postoperative pancreatic fistula (POPF). BACKGROUND: Criteria to predict the risk of POPF occurrence after PD are available. However, even when a high risk of POPF is predicted, TP is not currently accepted as an alternative to PD, because of its severe consequences on glycaemic control. Combining IAT with TP may mitigate such consequences. METHODS: Randomized, open-label, controlled, bicentric trial (NCT01346098). Candidates for PD at high-risk pancreatic anastomosis (ie, soft pancreas and duct diameter ≤3 mm) were randomly assigned (1:1) to undergo either PD or TP-IAT. The primary endpoint was the incidence of complications within 90 days after surgery. RESULTS: Between 2010 and 2019, 61 patients were assigned to PD (n=31) or TP-IAT (n=30). In the intention-to-treat analysis, morbidity rate was 90·3% after PD and 60% after TP-IAT ( P =0.008). According to complications' severity, PD was associated with an increased risk of grade ≥2 [odds ratio (OR)=7.64 (95% CI: 1.35-43.3), P =0.022], while the OR for grade ≥3 complications was 2.82 (95% CI: 0.86-9.24, P =0.086). After TP-IAT, the postoperative stay was shorter [median: 10.5 vs 16.0 days; P <0.001). No differences were observed in disease-free survival, site of recurrence, disease-specific survival, and overall survival. TP-IAT was associated with a higher risk of diabetes [hazard ratio=9.1 (95% CI: 3.76-21.9), P <0.0001], but most patients maintained good metabolic control and showed sustained C-peptide production over time. CONCLUSIONS: TP-IAT may become the standard treatment in candidates for PD, when a high risk of POPF is predicted.


Subject(s)
Islets of Langerhans Transplantation , Pancreatitis, Chronic , Humans , Pancreatectomy/adverse effects , Pancreaticojejunostomy , Pancreaticoduodenectomy/adverse effects , Prospective Studies , Transplantation, Autologous , Pancreatitis, Chronic/surgery , Treatment Outcome , Islets of Langerhans Transplantation/adverse effects , Postoperative Complications/epidemiology , Postoperative Complications/prevention & control , Postoperative Complications/etiology , Pancreatic Fistula/epidemiology , Pancreatic Fistula/etiology , Pancreatic Fistula/prevention & control
4.
J Clin Endocrinol Metab ; 107(3): e1009-e1019, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34718627

ABSTRACT

PURPOSE: To assess whether dysglycemia diagnosed during severe acute respiratory syndrome coronavirus 2 pneumonia may become a potential public health problem after resolution of the infection. In an adult cohort with suspected coronavirus disease 2019 (COVID-19) pneumonia, we integrated glucose data upon hospital admission with fasting blood glucose (FBG) in the year prior to COVID-19 and during postdischarge follow-up. METHODS: From February 25 to May 15, 2020, 660 adults with suspected COVID-19 pneumonia were admitted to the San Raffaele Hospital (Milan, Italy). Through structured interviews/ medical record reviews, we collected demographics, clinical features, and laboratory tests upon admission and additional data during hospitalization or after discharge and in the previous year. Upon admission, we classified participants according to American Diabetes Association criteria as having (1) preexisting diabetes, (2) newly diagnosed diabetes, (3) hyperglycemia not in the diabetes range, or (4) normoglycemia. FBG prior to admission and during follow-up were classified as normal or impaired fasting glucose and fasting glucose in the diabetes range. RESULTS: In patients with confirmed COVID (n = 589), the proportion with preexisting or newly diagnosed diabetes, hyperglycemia not in the diabetes range and normoglycemia was 19.6%, 6.7%, 43.7%, and 30.0%, respectively. Patients with dysglycemia associated to COVID-19 had increased markers of inflammation and organs' injury and poorer clinical outcome compared to those with normoglycemia. After the infection resolved, the prevalence of dysglycemia reverted to preadmission frequency. CONCLUSIONS: COVID-19-associated dysglycemia is unlikely to become a lasting public health problem. Alarmist claims on the diabetes risk after COVID-19 pneumonia should be interpreted with caution.


Subject(s)
Blood Glucose/analysis , COVID-19/metabolism , Hyperglycemia/etiology , SARS-CoV-2 , Aged , Aged, 80 and over , COVID-19/complications , Fasting/metabolism , Female , Humans , Male , Middle Aged
5.
Biology (Basel) ; 10(8)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34439986

ABSTRACT

AIM: The aim of the current study was to compare clinical characteristics, laboratory findings, and major outcomes of patients hospitalized for COVID-19 pneumonia with COVID-associated hyperglycaemia or pre-existing diabetes. METHODS: A cohort of 176 adult patients with a diagnosis of pre-existing diabetes (n = 112) or COVID-associated hyperglycaemia (n = 55) was studied. RESULTS: Patients with COVID-associated hyperglycaemia had lower BMI, significantly less comorbidities, and higher levels of inflammatory markers and indicators of multi-organ injury than those with pre-existing diabetes. No differences between pre-existing diabetes and COVID-associated hyperglycaemia were evident for symptoms at admission, the humoral response against SARS-CoV-2, or autoantibodies to glutamic acid decarboxylase or interferon alpha-4. COVID-associated hyperglycaemia was independently associated with the risk of adverse clinical outcome, which was defined as ICU admission or death (HR 2.11, 95% CI 1.34-3.31; p = 0.001), even after adjustment for age, sex, and other selected variables associated with COVID-19 severity. Furthermore, at the same time, we documented a negative association (HR 0.661, 95% CI 0.43-1.02; p = 0.063) between COVID-associated hyperglycaemia to swab negativization. CONCLUSIONS: Recognizing hyperglycaemia as a specific clinical entity associated with COVID-19 pneumonia is relevant for early and appropriate patient management and close monitoring for the progression of disease severity.

6.
Metabolism ; 116: 154463, 2021 03.
Article in English | MEDLINE | ID: mdl-33309713

ABSTRACT

OBJECTIVES: GDI1 gene encodes for αGDI, a protein controlling the cycling of small GTPases, reputed to orchestrate vesicle trafficking. Mutations in human GDI1 are responsible for intellectual disability (ID). In mice with ablated Gdi1, a model of ID, impaired working and associative short-term memory was recorded. This cognitive phenotype worsens if the deletion of αGDI expression is restricted to neurons. However, whether astrocytes, key homeostasis providing neuroglial cells, supporting neurons via aerobic glycolysis, contribute to this cognitive impairment is unclear. METHODS: We carried out proteomic analysis and monitored [18F]-fluoro-2-deoxy-d-glucose uptake into brain slices of Gdi1 knockout and wild type control mice. d-Glucose utilization at single astrocyte level was measured by the Förster Resonance Energy Transfer (FRET)-based measurements of cytosolic cyclic AMP, d-glucose and L-lactate, evoked by agonists selective for noradrenaline and L-lactate receptors. To test the role of astrocyte-resident processes in disease phenotype, we generated an inducible Gdi1 knockout mouse carrying the Gdi1 deletion only in adult astrocytes and conducted behavioural tests. RESULTS: Proteomic analysis revealed significant changes in astrocyte-resident glycolytic enzymes. Imaging [18F]-fluoro-2-deoxy-d-glucose revealed an increased d-glucose uptake in Gdi1 knockout tissue versus wild type control mice, consistent with the facilitated d-glucose uptake determined by FRET measurements. In mice with Gdi1 deletion restricted to astrocytes, a selective and significant impairment in working memory was recorded, which was rescued by inhibiting glycolysis by 2-deoxy-d-glucose injection. CONCLUSIONS: These results reveal a new astrocyte-based mechanism in neurodevelopmental disorders and open a novel therapeutic opportunity of targeting aerobic glycolysis, advocating a change in clinical practice.


Subject(s)
Deoxyglucose/pharmacology , Glycolysis/drug effects , Guanine Nucleotide Dissociation Inhibitors/genetics , Intellectual Disability/genetics , Memory Disorders/prevention & control , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Deoxyglucose/therapeutic use , Down-Regulation/drug effects , Glucose/metabolism , Guanine Nucleotide Dissociation Inhibitors/deficiency , Intellectual Disability/drug therapy , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Maze Learning/drug effects , Memory/drug effects , Memory Disorders/genetics , Mice , Mice, Knockout
7.
Diabetologia ; 63(12): 2548-2558, 2020 12.
Article in English | MEDLINE | ID: mdl-33029657

ABSTRACT

AIMS/HYPOTHESIS: The aim of the study was to characterise the humoral response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in patients with diabetes. Demonstrating the ability to mount an appropriate antibody response in the presence of hyperglycaemia is relevant for the comprehension of mechanisms related to the observed worse clinical outcome of coronavirus disease 2019 (COVID-19) pneumonia in patients with diabetes and for the development of any future vaccination campaign to prevent SARS-CoV-2 infection. METHODS: Using a highly specific and sensitive measurement of antibodies by fluid-phase luciferase immunoprecipitation assays, we characterised the IgG, IgM and IgA response against multiple antigens of SARS-CoV-2 in a cohort of 509 patients with documented diagnosis of COVID-19, prospectively followed at our institution. We analysed clinical outcomes and antibody titres according to the presence of hyperglycaemia, i.e., either diagnosed or undiagnosed diabetes, at the time of, or during, hospitalisation. RESULTS: Among patients with confirmed COVID-19, 139 (27.3%) had diabetes: 90 (17.7%) had diabetes diagnosed prior to the hospital admission (comorbid diabetes) while 49 (9.6%) had diabetes diagnosed at the time of admission (newly diagnosed). Diabetes was associated with increased levels of inflammatory biomarkers and hypercoagulopathy, as well as leucocytosis and neutrophilia. Diabetes was independently associated with risk of death (HR 2.32 [95% CI 1.44, 3.75], p = 0.001), even after adjustment for age, sex and other relevant comorbidities. Moreover, a strong association between higher glucose levels and risk of death was documented irrespective of diabetes diagnosis (HR 1.14 × 1.1 mmol/l [95% CI 1.08, 1.21], p < 0.001). The humoral response against SARS-CoV-2 in patients with diabetes was present and superimposable, as for timing and antibody titres, to that of non-diabetic patients, with marginal differences, and was not influenced by glucose levels. Of the measured antibody responses, positivity for IgG against the SARS-CoV-2 spike receptor-binding domain (RBD) was predictive of survival rate, both in the presence or absence of diabetes. CONCLUSIONS/INTERPRETATION: The observed increased severity and mortality risk of COVID-19 pneumonia in patients with hyperglycaemia was not the result of an impaired humoral response against SARS-CoV-2. RBD IgG positivity was associated with a remarkable protective effect, allowing for a cautious optimism about the efficacy of future vaccines against SARs-COV-2 in people with diabetes. Graphical abstract.


Subject(s)
Antibody Formation , Antigens, Viral/immunology , Coronavirus Infections/immunology , Diabetes Mellitus/immunology , Pneumonia, Viral/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Biomarkers/analysis , Blood Coagulation Disorders/complications , Blood Coagulation Disorders/immunology , Blood Glucose/analysis , COVID-19 , Cohort Studies , Coronavirus Infections/mortality , Female , Humans , Immunity, Humoral , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Male , Middle Aged , Pandemics , Pneumonia, Viral/mortality , Risk Factors , Survival Analysis
8.
Diabetes ; 68(8): 1541-1543, 2019 08.
Article in English | MEDLINE | ID: mdl-31331988

ABSTRACT

In December 2018, Diabetes and Diabetologia began requiring authors of papers reporting data obtained from studies on human islets to report critical characteristics of the human islets used for research. The islet community was asked to provide feedback on it. Here is the contribution by the European Consortium for Islet Transplantation.


Subject(s)
Diabetes Mellitus/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation
9.
Transplantation ; 103(4): 839-851, 2019 04.
Article in English | MEDLINE | ID: mdl-30130323

ABSTRACT

BACKGROUND: Results in murine and nonhuman primate suggested that the bone marrow (BM) might be an alternative site for pancreatic islet transplantation. METHODS: We report the results of 2 clinical studies in patients with type 1 diabetes receiving an intra-BM allogeneic islet transplantation: a feasibility study in patients with hepatic contraindications for liver islet allotransplantation receiving a single intra-BM islet infusion (n = 4) and a pilot randomized trial (1:1 allocation using blocks of size 6) in which patients were randomized to receive islets into either the liver (n = 6) or BM (n = 3) to evaluate islet transplant function and survival. RESULTS: We observed no adverse events related to the intrabone injection procedure or the presence of islets in the BM. None of the recipient of an intra-BM allogeneic islet transplantation had a primary nonfunction, as shown by measurable posttransplantation C-peptide levels and histopathological evidence of insulin-producing cells or molecular markers of endocrine tissue in BM biopsy samples collected during follow-up. All patients receiving islets in the BM except 1 lost islet function during the first 4 months after infusion (2 with an early graft loss). Based on biopsies and immunomonitoring, we concluded that the islet loss was primarily caused by the recurrence of autoimmunity. CONCLUSIONS: Bone marrow is not a suitable alternative site for pancreatic islet allotransplantation in patients with type 1 diabetes.


Subject(s)
Bone Marrow/surgery , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation/methods , Biopsy , Bone Marrow/pathology , Diabetes Mellitus, Type 1/immunology , Humans , Pilot Projects , Transplantation, Homologous
10.
Am J Transplant ; 19(3): 920-928, 2019 03.
Article in English | MEDLINE | ID: mdl-30549450

ABSTRACT

Islet autotransplant is particularly attractive to prevent diabetes after extended pancreatectomy for benign or borderline/malignant pancreas disease. Between 2008 and 2018, 25 patients underwent left extended pancreatectomy (>60%) and islet autotransplant for a neoplasm located in the pancreatic neck or proximal body. Overall, disease-free and diabetes-free survivals were estimated and compared with those observed in 68 nondiabetic patients who underwent distal pancreatectomy for pancreatic neoplasms without islet autotransplant. Median follow-up was 4 years. We observed no deaths and a low morbidity (nonserious procedure-related complications in 2 of 25 patients). Patient and insulin-independent survival rates at 4 years were 100% and 96%, respectively. Glucose homeostasis remained within a nondiabetic range at all times for 19 (73%) of 25 patients. Preoperative glycemic level and insulin resistance were major predictors of diabetes development in these patients. Patients undergoing islet autotransplant had a longer diabetes-free survival than did patients without islet autotransplant (P = .04). In conclusion, islet autotransplant after extended pancreatic resection for neoplasms is a safe and successful procedure for preventing diabetes.


Subject(s)
Diabetes Mellitus/mortality , Islets of Langerhans Transplantation/mortality , Pancreatectomy/mortality , Pancreatic Neoplasms/mortality , Autografts , Case-Control Studies , Combined Modality Therapy , Diabetes Mellitus/prevention & control , Feasibility Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Prognosis , Risk Factors
11.
J Clin Endocrinol Metab ; 103(12): 4343-4356, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30203067

ABSTRACT

Context: Recent studies have suggested that influenza A virus (IAV) might be involved in the etiology of diabetes. Objective and Methods: To address this question, we tested the ability of H1N1 pandemic IAV to infect, replicate, and damage human ß cells/pancreatic islets in vitro and induce pancreatic damage and/or glucose metabolism alterations in chemical and autoimmune models of ß cell damage in vivo. Moreover, we looked for direct and/or indirect evidence of correlation between IAV infection and autoimmunity/diabetes in humans. Results: Human H1N1 A/California/2009-derived viruses infected human pancreatic islets in vitro, inducing a proinflammatory response associated with substantial increases of CXCL9 and CXCL10 release. In vivo, infected mice showed a clear susceptibility to the virus, with its localization also found in extrapulmonary organs, including the pancreas. Infection was able to induce mild modifications of glycemia in C57B6 mice after chemical damage of islets but did not modulate the autoimmune damage of islets in NOD mice. One of 69 nasopharyngeal swabs collected from patients at the onset of type 1 diabetes yielded positive results for IAV. Pancreas sections from 17 organ donors available from the Network for Pancreatic Organ Donors With Diabetes showed the persistence of CXCL10-positive cells in islet autoimmunity-positive subjects; however, extremely rare cells stained for viral RNA and not preferentially in autoimmune subjects. Conclusion: Influenza H1N1 pdm strains are able to infect and replicate in mammalian pancreatic cells both in vitro and in vivo but did not cause any functional impairment consistent with diabetes.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/virology , Adolescent , Adult , Animals , Blood Glucose , Cell Line , Cell Line, Tumor , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Child , Child, Preschool , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/virology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/virology , Dogs , Female , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/epidemiology , Influenza, Human/immunology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/virology , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pandemics , Primary Cell Culture , RNA, Viral/isolation & purification , Young Adult
12.
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
13.
Transplantation ; 101(10): 2492-2500, 2017 10.
Article in English | MEDLINE | ID: mdl-28358727

ABSTRACT

BACKGROUND: To assess feasibility, safety, and metabolic outcome of islet auto transplantation (IAT) in patients undergoing completion pancreatectomy because of sepsis or bleeding after pancreatic surgery. METHODS: From November 2008 to October 2016, approximately 22 patients were candidates to salvage IAT during emergency relaparotomy because of postpancreatectomy sepsis (n = 11) or bleeding (n = 11). Feasibility, efficacy, and safety of salvage IAT were compared with those documented in a cohort of 36 patients who were candidate to simultaneous IAT during nonemergency preemptive completion pancreatectomy through the pancreaticoduodenectomy. RESULTS: The percentage of candidates that received the infusion of islets was significantly lower in salvage IAT than simultaneous IAT (59.1% vs 88.9%, P = 0.008), mainly because of a higher rate of inadequate islet preparations. Even if microbial contamination of islet preparation was significantly higher in candidates to salvage IAT than in those to simultaneous IAT (78.9% vs 20%, P < 0.001), there was no evidence of a higher rate of complications related to the procedure. Median follow-up was 5.45 ± 0.52 years. Four (36%) of 11 patients reached insulin independence, 6 patients (56%) had partial graft function, and 1 patient (9%) had primary graft nonfunction. At the last follow-up visit, median fasting C-peptide was 0.43 (0.19-0.93) ng/mL; median insulin requirement was 0.38 (0.04-0.5) U/kg per day, and median HbA1c was 6.6% (5.9%-8.1%). Overall mortality, in-hospital mortality, metabolic outcome, graft survival, and insulin-free survival after salvage IAT were not different from those documented after simultaneous IAT. CONCLUSIONS: Our data demonstrate the feasibility, efficacy, and safety of salvage IAT after relaparotomy.


Subject(s)
Graft Survival , Islets of Langerhans Transplantation/methods , Pancreatectomy/methods , Pancreatitis, Chronic/surgery , Postoperative Complications/prevention & control , Salvage Therapy/methods , Aged , Female , Follow-Up Studies , Humans , Italy/epidemiology , Male , Middle Aged , Pancreatitis, Chronic/mortality , Retrospective Studies , Survival Rate/trends , Transplantation, Autologous
14.
Transplantation ; 101(5): 1046-1055, 2017 05.
Article in English | MEDLINE | ID: mdl-27575689

ABSTRACT

BACKGROUND: The aim of this study was to characterize the immune response against intrabone marrow (BM-Tx) or intraliver (liver-Tx) transplanted islets in the presence or in the absence of immunosuppression. METHODS: Less (C57BL/6 in Balb/c) and highly (Balb/c in C57BL/6) stringent major histocompatibility complex fully mismatched mouse models were used to evaluate the alloimmune response. Single antigen-mismatched mouse model (C57BL/6 RIP-GP in C57BL/6) was used to evaluate the antigen-specific immune response. Mice received tacrolimus (FK-506, 0.1 mg/kg per day)/mycophenolate mofetil (MMF, 60 mg/kg per day), and anti-CD3 (50 µg/day) either alone or in combination. RESULTS: Transplant site did not impact the timing nor the kinetics of the alloimmune and single antigen-specific memory T cell responses in the absence of immunosuppression or in the presence of MMF/FK-506 combination. On the other hand, the median time to graft rejection was 28 ± 5.2 days and 16 ± 2.6 days (P = 0.14) in the presence of anti-CD3 treatment, 50 ± 12.5 days and 10 ± 1.3 days (P = 0.003) in the presence of anti-CD3/MMF/FK-506 treatment for liver-Tx and BM-Tx, respectively. Anti-CD3 did not differentially reach BM and liver tissues but was more effective in reducing graft associated T cell responses in liver-Tx than in BM-Tx. CONCLUSIONS: Islets infused in the BM appear less protected from the adaptive immune response in the presence of the anti-CD3 treatment. This result raises some concerns over the potential of the BM as a site for islet allotransplantation.


Subject(s)
Bone Marrow/surgery , Graft Rejection/immunology , Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/methods , Liver/surgery , Adaptive Immunity , Animals , Biomarkers/metabolism , Bone Marrow/immunology , Drug Therapy, Combination , Graft Rejection/prevention & control , Isoantibodies/metabolism , Isoantigens/immunology , Liver/immunology , Male , Mice , Mice, Inbred BALB C , T-Lymphocytes/metabolism
15.
Cell Transplant ; 25(3): 491-501, 2016.
Article in English | MEDLINE | ID: mdl-26102316

ABSTRACT

Islet volume and endocrine pancreas architecture (islet size distribution) may be independent determinants of ß-cell function. Furthermore, the accuracy of homeostatic model assessment (HOMA) indexes in predicting ß-cell mass has never been assessed. Here we investigated the relationships between islet volume, islet density, and islet size distribution, estimated after pancreatic tissue digestion, with established indexes of ß-cell function in humans. We included in this study 42 patients who were candidates for islet autotransplantation and had well-characterized glucose metabolism. Indexes of insulin secretion were calculated and compared with the islet volume, as a surrogate of ß-cell mass, obtained after digestion of pancreas. Islet counting analysis showed considerable interindividual variation in islet density and size. Islet volume, but not density nor size, was the only independent determinant of ß-cell function assessed by insulin HOMA ß-cell. Islet volume was significantly reduced in the patients with overt hyperglycemia, but not in patients with impaired fasting glucose. Insulin HOMA ß-cell predicted islet volume better than other measures of fasting insulin secretion. In conclusion, the present study documented a close direct relationship between indexes of ß-cell function and islet volume in humans. The insulin HOMA ß-cell provides a more reliable estimate of pancreatic islet volume than fasting glucose before islet isolation.


Subject(s)
Insulin-Secreting Cells/metabolism , Islets of Langerhans/anatomy & histology , Islets of Langerhans/physiology , Adult , Aged , Female , Glucose/metabolism , Homeostasis , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Male , Middle Aged , Organ Size
16.
Acta Diabetol ; 52(6): 1025-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25733399

ABSTRACT

AIMS: New sources of insulin-secreting cells are strongly required for the cure of diabetes. Recent successes in differentiating embryonic stem cells, in combination with the discovery that it is possible to derive human induced pluripotent stem cells (iPSCs) from somatic cells, have raised the possibility that patient-specific beta cells might be derived from patients through cell reprogramming and differentiation. In this study, we aimed to obtain insulin-producing cells from human iPSCs and test their ability to secrete insulin in vivo. METHODS: Human iPSCs, derived from both fetal and adult fibroblasts, were differentiated in vitro into pancreas-committed cells and then transplanted into immunodeficient mice at two different stages of differentiation (posterior foregut and endocrine cells). RESULTS: IPSCs were shown to differentiate in insulin-producing cells in vitro, following the stages of pancreatic organogenesis. At the end of the differentiation, the production of INSULIN mRNA was highly increased and 5 ± 2.9 % of the cell population became insulin-positive. Terminally differentiated cells also produced C-peptide in vitro in both basal and stimulated conditions. In vivo, mice transplanted with pancreatic cells secreted human C-peptide in response to glucose stimulus, but transplanted cells were observed to lose insulin secretion capacity during the time. At histological evaluation, the grafts resulted to be composed of a mixed population of cells containing mature pancreatic cells, but also pluripotent and some neuronal cells. CONCLUSION: These data overall suggest that human iPSCs have the potential to generate insulin-producing cells and that these differentiated cells can engraft and secrete insulin in vivo.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Insulin-Secreting Cells/physiology , Insulin-Secreting Cells/transplantation , Pancreas Transplantation , Animals , C-Peptide/analysis , C-Peptide/metabolism , Cell Differentiation , Fetus/cytology , Fibroblasts , Fluorometry , Humans , Insulin/analysis , Insulin/metabolism , Mice, Inbred NOD , Mice, SCID
18.
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
19.
Transplantation ; 98(12): 1301-9, 2014 Dec 27.
Article in English | MEDLINE | ID: mdl-25286053

ABSTRACT

BACKGROUND: Our final objective is to develop an adoptive therapy with tolerogenic donor-specific type 1 T regulatory cells for patients with type 1 diabetes undergoing islet transplantation. The achievement of this objective depends on the availability of an immunosuppressive treatment compatible with the survival, function, and expansion of type 1 T regulatory cells. METHODS: For this purpose, we designed a single-group, phase 1 to 2 trial with an immunosuppression protocol including: (i) rapamycin treatment before the first islet infusion (starting ≥ 30 days before transplantation); (ii) induction therapy with anti-thymocyte globulin (ATG) instead of anti-interleukin-2Ra monoclonal antibody (after the first islet infusion only); (iii) short-term treatment with steroids and interleukin-1Ra (right before and for 2 weeks after each infusion); rapamycin+mycophenolate mofetil treatment as maintenance therapy. The target enrollment was 10 patients. RESULTS: Ten of 15 patients who started the pretransplant rapamycin treatment completed it. Nine of 10 patients did not complete the induction therapy with ATG, and three of 10 required adaptation of maintenance immunosuppression caused by side effects. Four of 10 patients acquired insulin independence which can be maintained up to year 3 after last infusion. All six other patients have lost their graft, and the early graft loss was associated with lower dose of ATG during induction. CONCLUSION: This protocol resulted feasible, safe but less efficient in maintaining graft survival during the time than other T-cell depletion-based protocols. An adequate induction at the first infusion should be considered to improve the overall clinical outcome.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation , Adult , Antilymphocyte Serum/chemistry , Antilymphocyte Serum/therapeutic use , Calcineurin Inhibitors/chemistry , Female , Glomerular Filtration Rate , Graft Survival , Humans , Immunosuppression Therapy/methods , Immunosuppressive Agents/chemistry , Insulin/metabolism , Interleukin 1 Receptor Antagonist Protein/chemistry , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Male , Middle Aged , Sirolimus/chemistry , Sirolimus/therapeutic use , Steroids/chemistry , Steroids/therapeutic use , T-Lymphocytes, Regulatory/cytology , Treatment Outcome
20.
World J Gastroenterol ; 20(14): 4030-6, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24744593

ABSTRACT

AIM: To evaluate the safety and feasibility of laparoscopic spleen-preserving distal pancreatectomy (LSPDP) with autologous islet transplantation (AIT) for benign tumors of the pancreatic body-neck. METHODS: Three non-diabetic, female patients (age 37, 44 and 35 years, respectively) were declared candidates for surgery, between May and September 2011, because of pancreatic body/neck cystic lesions. The planned operation was an LSPDP associated with AIT from the normal pancreas distal to the neoplasm. Islets isolation was performed on the residual pancreatic parenchyma after frozen section examination of the margin. Purified autologous islets were infused into the portal vein by a percutaneous transhepatic approach the day after surgery. RESULTS: The procedure was performed successfully in all the three cases, and the spleen was preserved along with its vessels. Mean operation time was 283 ± 52 min and average blood loss was 133 ± 57 mL. Residual pancreas weights were 33, 22 and 30 g, and 105.200, 40.390 and 94.790 islet equivalents were isolated, respectively. Surgical complications occurred in one patient (grade A pancreatic fistula). Postoperative stays were 6, 6 and 7 d, respectively. Histopathological evaluation revealed mucinous cystic neoplasm in cases 1 and 3, and serous cystic neoplasm in patient 2. No postoperative insulin administration was required. One patient developed a transient partial portal thrombosis 2 mo after islet infusion. Patients are insulin independent at a mean follow up of 8 ± 2 mo. CONCLUSION: Combination of LSPDP and AIT is feasible and could be effective to minimize the surgical impact for benign neoplasm of pancreatic body-neck.


Subject(s)
Laparoscopy/methods , Pancreatectomy/methods , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms/therapy , Spleen/surgery , Abdominal Pain/diagnostic imaging , Abdominal Pain/pathology , Adult , Female , Humans , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Pancreas/pathology , Renal Colic/diagnostic imaging , Renal Colic/pathology , Tomography, X-Ray Computed , Transplantation , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...