1501
|
Sima C, Van Dyke TE. Therapeutic Targets for Management of Periodontitis and Diabetes. Curr Pharm Des 2016; 22:2216-37. [PMID: 26881443 PMCID: PMC4854768 DOI: 10.2174/1381612822666160216150338] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/15/2016] [Indexed: 11/22/2022]
Abstract
The increasing incidence of diabetes mellitus (DM) and chronic periodontitis (CP) worldwide imposes a rethinking of individualized therapy for patients with both conditions. Central to bidirectional links between DM and CP is deregulated systemic inflammation and dysfunctional immune responses to altered-self and non-self. Control of blood glucose levels and metabolic imbalances associated with hyperglycemia in DM, and disruption of pathogenic subgingival biofilms in CP are currently the main therapeutic approaches for these conditions. Mounting evidence suggests the need to integrate immune modulatory therapeutics in treatment regimens that address the unresolved inflammation associated with DM and CP. The current review discusses the pathogenesis of DM and CP with emphasis on deregulated inflammation, current therapeutic approaches and the novel pro-resolution lipid mediators derived from Ω-3 polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Corneliu Sima
- Corneliu Sima, 245 First Street, Room 5145, Cambridge, MA, 02142, USA.
| | | |
Collapse
|
1502
|
Gourdy P, Bourgeois EA, Levescot A, Pham L, Riant E, Ahui ML, Damotte D, Gombert JM, Bayard F, Ohlsson C, Arnal JF, Herbelin A. Estrogen Therapy Delays Autoimmune Diabetes and Promotes the Protective Efficiency of Natural Killer T-Cell Activation in Female Nonobese Diabetic Mice. Endocrinology 2016; 157:258-67. [PMID: 26485613 DOI: 10.1210/en.2015-1313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Therapeutic strategies focused on restoring immune tolerance remain the main avenue to prevent type 1 diabetes (T1D). Because estrogens potentiate FoxP3+ regulatory T cells (Treg) and invariant natural killer T (iNKT) cells, two regulatory lymphocyte populations that are functionally deficient in nonobese diabetic (NOD) mice, we investigated whether estradiol (E2) therapy influences the course of T1D in this model. To this end, female NOD mice were sc implanted with E2- or placebo-delivering pellets to explore the course of spontaneous and cyclophosphamide-induced diabetes. Treg-depleted and iNKT-cell-deficient (Jα18(-/-)) NOD mice were used to assess the respective involvement of these lymphocyte populations in E2 effects. Early E2 administration (from 4 wk of age) was found to preserve NOD mice from both spontaneous and cyclophosphamide-induced diabetes, and a complete protection was also observed throughout treatment when E2 treatment was initiated after the onset of insulitis (from 12 wk of age). This delayed E2 treatment remained fully effective in Treg-depleted mice but failed to entirely protect Jα18(-/-) mice. Accordingly, E2 administration was shown to restore the cytokine production of iNKT cells in response to in vivo challenge with the cognate ligand α-galactosylceramide. Finally, transient E2 administration potentiated the previously described protective action of α-galactosylceramide treatment in NOD females. This study provides original evidence that E2 therapy strongly protects NOD mice from T1D and reveals the estrogen/iNKT cell axis as a new effective target to counteract diabetes onset at the stage of insulitis. Estrogen-based therapy should thus be considered for T1D prevention.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/prevention & control
- Cytokines/blood
- Cytokines/metabolism
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/prevention & control
- Drug Implants
- Estradiol/administration & dosage
- Estradiol/therapeutic use
- Estrogen Replacement Therapy
- Estrogens/administration & dosage
- Estrogens/therapeutic use
- Female
- Galactosylceramides/agonists
- Galactosylceramides/pharmacology
- Galactosylceramides/therapeutic use
- Immune Tolerance/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/drug effects
- Lymphocyte Depletion/adverse effects
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Mutant Strains
- Ovariectomy/adverse effects
- Prediabetic State/drug therapy
- Prediabetic State/immunology
- Prediabetic State/metabolism
- Prediabetic State/prevention & control
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Pierre Gourdy
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elvire A Bourgeois
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Anaïs Levescot
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Linh Pham
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elodie Riant
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Marie-Louise Ahui
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Diane Damotte
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-Marc Gombert
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Francis Bayard
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Claes Ohlsson
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-François Arnal
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - André Herbelin
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| |
Collapse
|
1503
|
Balakhadze M, Giorgadze E, Lomidze M. The Frequency of Langerhans Islets β-Cells Autoantibodies (Anti-GAD) in Georgian Children and Adolescents with Chronic Autoimmune Thyroiditis. Int J Endocrinol 2016; 2016:6597091. [PMID: 27429616 PMCID: PMC4939347 DOI: 10.1155/2016/6597091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/31/2016] [Accepted: 06/06/2016] [Indexed: 11/17/2022] Open
Abstract
Aim. Chronic autoimmune thyroiditis and type 1 diabetes mellitus are organ-specific autoimmune diseases. There is large evidence that autoimmunity against the thyroid gland in patients with type 1 diabetes mellitus is increased, but little is known about anti-islet cell autoimmune status in patients with chronic autoimmune thyroiditis. We evaluated the concentration of antibodies against glutamic acid decarboxylase (GAD) which are widely used as a diagnostic and predictive tool for type 1 diabetes mellitus, in school-aged Georgian children with chronic autoimmune thyroiditis. Methods. The frequency of anti-GAD antibodies was measured in Georgian school-aged children with chronic autoimmune thyroiditis and compared to healthy age and sex matched controls. Results. Of the 41 patients with chronic autoimmune thyroiditis 4 (9.8%) were positive for GAD antibodies. The frequency of GAD positivity in the chronic autoimmune thyroiditis group was significantly higher than in the control subjects (P = 0.036). Conclusion. In the study we found that the frequency of GAD antibody positivity in autoimmune thyroiditis patients was significantly higher (9.8%, P = 0.036) than in the control group. Our findings support the concept that patients with autoimmune thyroid disease may develop type 1 diabetes mellitus in future life.
Collapse
Affiliation(s)
- Mariam Balakhadze
- Ivane Javakhishvili Tbilisi State University, 0179 Tbilisi, Georgia
- V. Iverieli Endocrinology Metabology Dietology Center “ENMEDIC”, 9 Tsinandali Street, 0144 Tbilisi, Georgia
- *Mariam Balakhadze:
| | - Elene Giorgadze
- Ivane Javakhishvili Tbilisi State University, 0179 Tbilisi, Georgia
- National Institute of Endocrinology, 2/6 Lubliana Street, 0159 Tbilisi, Georgia
| | - Marina Lomidze
- National Institute of Endocrinology, 2/6 Lubliana Street, 0159 Tbilisi, Georgia
| |
Collapse
|
1504
|
Risk factors for sudden death and cardiac arrest at the onset of fulminant type 1 diabetes mellitus. Diabetol Int 2015; 7:281-288. [PMID: 30603275 DOI: 10.1007/s13340-015-0247-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/01/2015] [Indexed: 01/23/2023]
Abstract
Aims The onset of fulminant type 1 diabetes mellitus is sometimes accompanied by sudden death or cardiac arrest. The aim of this study was to determine the risk factors for the development of these conditions at the onset of fulminant type 1 diabetes mellitus. Methods We conducted a search of the literature on fulminant type 1 diabetes and sudden death or cardiac arrest published up to 2012 in PubMed and Ichushi (a Japanese article database), and a questionnaire survey was administered to the authors of the articles and to diabetes specialists affiliated to the Japan Diabetes Society. We analyzed the clinical data at disease onset of 17 patients with fulminant type 1 diabetes mellitus who experienced sudden death or cardiac arrest, and those of 257 patients who did not develop these conditions. Results Patients with sudden death or cardiac arrest were younger, had a higher rate of impaired consciousness, more severe acidosis, hyperglycemia, hyponatremia, hyperkalemia, and hypochloremia, a higher serum blood urea nitrogen level, a higher serum creatinine level, and a higher plasma osmolality level than the other patients. In multiple logistic regression analyses, plasma glucose level was positively associated with sudden death or cardiac arrest. Receiver operating characteristic curve analyses showed that patients with a plasma glucose level over 1000 mg/dl (55.5 mmol/l) were at a high risk of cardiac arrest. Conclusions Severe metabolic derangement, especially a high plasma glucose level, is associated with sudden death or cardiac arrest at the onset of fulminant type 1 diabetes mellitus.
Collapse
|
1505
|
Marchand L, Nicolino M, Fabien N, Roep BO, Thivolet C. Multiple autoantibodies at onset do not accurately predict long-term pancreatic beta-cell fate in a 13-year-old obese child with immediate insulin-requiring diabetes. DIABETES & METABOLISM 2015; 42:69-70. [PMID: 26619919 DOI: 10.1016/j.diabet.2015.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/08/2015] [Accepted: 10/14/2015] [Indexed: 01/14/2023]
Affiliation(s)
- L Marchand
- Department of Endocrinology and Diabetes Lyon-Sud Hospital, hospices civils de Lyon, Claude-Bernard University, 69495 Pierre-Bénite cedex, France
| | - M Nicolino
- Department of Pediatric Endocrinology, hospices civils de Lyon, 69677 Bron, France
| | - N Fabien
- Immunology Laboratory, Lyon-Sud Hospital, hospices civils de Lyon, 69495 Lyon, France
| | - B O Roep
- Leiden University Medical Center, Department of Immunohaematology & Blood Transfusion, Leiden, The Netherlands
| | - C Thivolet
- Department of Endocrinology and Diabetes Lyon-Sud Hospital, hospices civils de Lyon, Claude-Bernard University, 69495 Pierre-Bénite cedex, France.
| |
Collapse
|
1506
|
Semenkovich CF, Danska J, Darsow T, Dunne JL, Huttenhower C, Insel RA, McElvaine AT, Ratner RE, Shuldiner AR, Blaser MJ. American Diabetes Association and JDRF Research Symposium: Diabetes and the Microbiome. Diabetes 2015; 64:3967-77. [PMID: 26420863 PMCID: PMC4876761 DOI: 10.2337/db15-0597] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/13/2015] [Indexed: 12/12/2022]
Abstract
From 27-29 October 2014, more than 100 people gathered in Chicago, IL, to participate in a research symposium titled "Diabetes and the Microbiome," jointly sponsored by the American Diabetes Association and JDRF. The conference brought together international scholars and trainees from multiple disciplines, including microbiology, bioinformatics, endocrinology, metabolism, and immunology, to share the current understanding of host-microbe interactions and their influences on diabetes and metabolism. Notably, this gathering was the first to assemble specialists with distinct expertise in type 1 diabetes, type 2 diabetes, immunology, and microbiology with the goal of discussing and defining potential pathophysiologies linking the microbiome and diabetes. In addition to reviewing existing evidence in the field, speakers presented their own original research to provide a comprehensive view of the current understanding of the topics under discussion.Presentations and discussions throughout the conference reflected a number of important concepts. The microbiota in any host represent a complex ecosystem with a high degree of interindividual variability. Different microbial communities, comprising bacteria, archaea, viruses, and fungi, occupy separate niches in and on the human body. Individually and collectively, these microbes provide benefits to the host-including nutrient harvest from food and protection against pathogens. They are dynamically regulated by both host genes and the environment, and they critically influence both physiology and lifelong health. The objective of the symposium was to discuss the relationship between the host and the microbiome-the combination of microbiota and their biomolecular environment and ecology-specifically with regard to metabolic and immunological systems and to define the critical research needed to understand and potentially target the microbiome in the prevention and treatment of diabetes. In this report, we present meeting highlights in the following areas: 1) relationships between diabetes and the microbiome, 2) bioinformatic tools, resources, and study design considerations, 3) microbial programming of the immune system, 4) the microbiome and energy balance, 5) interventions, and 6) limitations, unanswered questions, and resource and policy needs.
Collapse
Affiliation(s)
| | | | | | | | - Curtis Huttenhower
- Harvard T.H. Chan School of Public Health, Boston, MA, and Broad Institute of MIT and Harvard, Cambridge, MA
| | | | | | | | | | | |
Collapse
|
1507
|
Simmons KM, Michels AW. Alternate Ways to Quantify Antibodies. Diabetes Technol Ther 2015; 17:854-6. [PMID: 26544921 PMCID: PMC4677111 DOI: 10.1089/dia.2015.0328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Kimber M Simmons
- Barbara Davis Center for Childhood Diabetes, University of Colorado , Aurora, Colorado
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado , Aurora, Colorado
| |
Collapse
|
1508
|
Mellerio H, Guilmin-Crépon S, Jacquin P, Labéguerie M, Lévy-Marchal C, Alberti C. Long-term impact of childhood-onset type 1 diabetes on social life, quality of life and sexuality. DIABETES & METABOLISM 2015; 41:489-97. [PMID: 25869639 DOI: 10.1016/j.diabet.2014.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/19/2014] [Accepted: 12/28/2014] [Indexed: 02/07/2023]
Abstract
AIM This study describes the socio-professional outcomes, health-related quality of life (HRQOL) and sexuality of adults with childhood-onset type 1 diabetes (T1D). METHODS The study participants (n=388), recruited from a nationwide registry (age: 28.5 ± 3.1 years; T1D duration: 17.0 ± 2.7 years), completed a questionnaire (198 items); the results were compared with the French general population using standardized incidence ratios (SIRs) and Z scores matched for age, gender and period with/without education levels and patterns of family life. Linear regression models also investigated correlates of SF-36 Physical (PCS) and Mental Composite Scores (MCS). RESULTS Compared with the French general population, education levels of people with T1D were similar, with 68.6% having at least a high-school diploma or higher (SIR: 1.06, 95% CI: 0.93; 1.20), as were also their patterns of family life. Unemployment was higher in T1D women (15.3%, SIR: 1.50, 1.00; 2.05), but not in T1D men (8.6%, SIR: 0.96, 0.51; 1.57). Social discrimination was more common (SIR: 5.64, 4.64; 6.62), and frequency of daily alcohol consumption was higher (SIR: men, 3.34, 2.38; 4.54; women, 6.53, 4.57; 12.99). PCS and MCS were decreased moderately (mean ± SD: 52.0 ± 7.5; mean Z score: -0.2, 95% CI: -0.3; -0.1) and substantially (mean ± SD: 42.1 ± 12.4; mean Z score: -0.7, -0.8; -0.6), respectively. Fatigue and abandoning sports were predictive of a lower HRQOL. Both men and women were more frequently dissatisfied with their sex life. Prevalence of sexual problems was higher in women (SIR for: dysorgasmia, 1.91, 1.21-2.88; decreased/loss of desire: 2.11, 1.35-3.08), but similar in men. Participants with T1D-related complications had preserved social outcomes, but altered HRQOL. CONCLUSION Young adults with T1D have satisfactory social participation. However, their higher alcohol consumption, lower MCS and frequent dissatisfaction with sexuality suggest a heavy impact of the disease on morale, especially in women. Improving the everyday well-being of these young adults represents a key challenge for diabetes healthcare.
Collapse
Affiliation(s)
- H Mellerio
- Univ Paris Diderot, Sorbonne Paris Cité, ECEVE, UMRS 1123, 75010 Paris, France; Inserm, ECEVE U1123 et CIC-EC, CIC 1426, 75010 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, Unité d'épidémiologie clinique, 75019 Paris, France.
| | - S Guilmin-Crépon
- Univ Paris Diderot, Sorbonne Paris Cité, ECEVE, UMRS 1123, 75010 Paris, France; Inserm, ECEVE U1123 et CIC-EC, CIC 1426, 75010 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, Unité d'épidémiologie clinique, 75019 Paris, France
| | - P Jacquin
- Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, Médecine de l'adolescent, 75019 Paris, France
| | - M Labéguerie
- Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, Unité d'épidémiologie clinique, 75019 Paris, France
| | - C Lévy-Marchal
- Inserm, CIC-EC, CIC 1426, 75010 Paris, France; Inserm, Pôle Recherche clinique, 75013 Paris, France
| | - C Alberti
- Univ Paris Diderot, Sorbonne Paris Cité, ECEVE, UMRS 1123, 75010 Paris, France; Inserm, ECEVE U1123 et CIC-EC, CIC 1426, 75010 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, Unité d'épidémiologie clinique, 75019 Paris, France
| |
Collapse
|
1509
|
Abstract
Type 1 diabetes is a chronic autoimmune disease resulting from T cell-mediated destruction of insulin-producing beta cells within pancreatic islets. Disease incidence has increased significantly in the last two decades, especially in young children. Type 1 diabetes is now predictable in humans with the measurement of serum islet autoantibodies directed against insulin and beta cell proteins. Knowledge regarding the presentation of insulin and islet antigens to T cells has increased dramatically over the last several years. Here, we review the trimolecular complex in diabetes, which consists of a major histocompatibility molecule,self-peptide, and T cell receptor, with a focus on insulin peptide presentation to T cells. With this increased understanding of how antigens are presented to T cells comes the hope for improved therapies for type 1 diabetes prevention.
Collapse
Affiliation(s)
- Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kimberly M Simmons
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
1510
|
Van Allen J, Steele RG, Nelson MB, Peugh J, Egan A, Clements M, Patton SR. A Longitudinal Examination of Hope and Optimism and Their Role in Type 1 Diabetes in Youths. J Pediatr Psychol 2015; 41:741-9. [PMID: 26628250 DOI: 10.1093/jpepsy/jsv113] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/28/2015] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES To test the longitudinal associations between hope and optimism and health outcomes (i.e., HbA1c and self-monitored blood glucose [SMBG]) among youths with Type 1 diabetes mellitus (T1DM) over a 6-month period. METHODS A total of 110 participants (aged 10-16 years) completed study measures at Time 1, and 81 completed measures at Time 2. Analyses examined hope and optimism as predictors of change in health outcomes, and examined SMBG as a mediator of the relationship between hope and optimism, and HbA1c. RESULTS Change in hope, but not optimism, was associated with change in SMBG and HbA1c. Change in SMBG mediated the relationship between change in hope and HbA1c, but not between optimism and HbA1c. CONCLUSIONS It may be beneficial to assess hope in pediatric T1DM patients to identify youths who may be at risk for poor diabetes management, and to test the benefit of hope-based intervention efforts in clinical studies.
Collapse
Affiliation(s)
- Jason Van Allen
- Clinical Psychology Program, Department of Psychological Sciences, Texas Tech University,
| | - Ric G Steele
- Clinical Child Psychology Program, University of Kansas
| | | | - James Peugh
- Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center
| | - Anna Egan
- Division of Developmental and Behavioral Sciences, Children's Mercy Kansas City
| | - Mark Clements
- Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Mercy Kansas City, and
| | - Susana R Patton
- Department of Pediatrics, University of Kansas Medical Center
| |
Collapse
|
1511
|
Zaccardi F, Webb DR, Yates T, Davies MJ. Pathophysiology of type 1 and type 2 diabetes mellitus: a 90-year perspective. Postgrad Med J 2015; 92:63-9. [DOI: 10.1136/postgradmedj-2015-133281] [Citation(s) in RCA: 326] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 11/09/2015] [Indexed: 12/11/2022]
|
1512
|
Merchant AT, Nahhas GJ, Wadwa RP, Zhang J, Tang Y, Johnson LR, Maahs DM, Bishop F, Teles R, Morrato EH. Periodontal Microorganisms and Cardiovascular Risk Markers in Youth With Type 1 Diabetes and Without Diabetes. J Periodontol 2015; 87:376-84. [PMID: 26616842 DOI: 10.1902/jop.2015.150531] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND A subset of periodontal microorganisms has been associated with cardiovascular disease (CVD), which is the leading complication of type 1 diabetes (t1DM). The authors therefore evaluated the association between periodontal microorganism groups and early markers of CVD in youth with t1DM. METHODS A cross-sectional analysis was conducted among youth aged 12 to 19 years at enrollment; 105 had t1DM for ≥5 years and were seeking care at the Barbara Davis Center, University of Colorado, from 2009 to 2011, and 71 did not have diabetes. Subgingival plaque samples were assessed for counts of 41 periodontal microorganisms using DNA-DNA hybridization. Microorganisms were classified using cluster analysis into four groups named red-orange, orange-green, blue/other, and yellow/other, modified from Socransky's color scheme for periodontal microorganisms. Subsamples (54 with t1DM and 48 without diabetes) also received a periodontal examination at the University of Colorado School of Dental Medicine. RESULTS Participants were ≈15 years old on average, and 74% were white. Mean periodontal probing depth was 2 mm (SE 0.02), and 17% had bleeding on probing. In multivariable analyses, glycated hemoglobin (HbA1c) was inversely associated with the yellow/other cluster (microorganisms that are not associated with periodontal disease) among youth with t1DM. Blood pressure, triglycerides, low-density lipoprotein, high-density lipoprotein, and total cholesterol were not associated with microorganism clusters in this group. HbA1c was not associated with periodontal microorganism clusters among youth without diabetes. CONCLUSION Among youth with t1DM who had good oral health, periodontal microorganisms were not associated with CVD risk factors.
Collapse
Affiliation(s)
- Anwar T Merchant
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC
| | - Georges J Nahhas
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC
| | - R Paul Wadwa
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Jiajia Zhang
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC
| | - Yifan Tang
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC
| | | | - David M Maahs
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | | | | | | |
Collapse
|
1513
|
Comprehensive Survey of miRNA-mRNA Interactions Reveals That Ccr7 and Cd247 (CD3 zeta) are Posttranscriptionally Controlled in Pancreas Infiltrating T Lymphocytes of Non-Obese Diabetic (NOD) Mice. PLoS One 2015; 10:e0142688. [PMID: 26606254 PMCID: PMC4659659 DOI: 10.1371/journal.pone.0142688] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/26/2015] [Indexed: 01/14/2023] Open
Abstract
In autoimmune type 1 diabetes mellitus (T1D), auto-reactive clones of CD4+ and CD8+ T lymphocytes in the periphery evolve into pancreas-infiltrating T lymphocytes (PILs), which destroy insulin-producing beta-cells through inflammatory insulitis. Previously, we demonstrated that, during the development of T1D in non-obese diabetic (NOD) mice, a set of immune/inflammatory reactivity genes were differentially expressed in T lymphocytes. However, the posttranscriptional control involving miRNA interactions that occur during the evolution of thymocytes into PILs remains unknown. In this study, we postulated that miRNAs are differentially expressed during this period and that these miRNAs can interact with mRNAs involved in auto-reactivity during the progression of insulitis. To test this hypothesis, we used NOD mice to perform, for the first time, a comprehensive survey of miRNA and mRNA expression as thymocytes mature into peripheral CD3+ T lymphocytes and, subsequently, into PILs. Reconstruction of miRNA-mRNA interaction networks for target prediction revealed the participation of a large set of miRNAs that regulate mRNA targets related to apoptosis, cell adhesion, cellular regulation, cellular component organization, cellular processes, development and the immune system, among others. The interactions between miR-202-3p and the Ccr7 chemokine receptor mRNA or Cd247 (Cd3 zeta chain) mRNA found in PILs are highlighted because these interactions can contribute to a better understanding of how the lack of immune homeostasis and the emergence of autoimmunity (e.g., T1D) can be associated with the decreased activity of Ccr7 or Cd247, as previously observed in NOD mice. We demonstrate that these mRNAs are controlled at the posttranscriptional level in PILs.
Collapse
|
1514
|
Immune Depletion in Combination with Allogeneic Islets Permanently Restores Tolerance to Self-Antigens in Diabetic NOD Mice. PLoS One 2015; 10:e0142318. [PMID: 26580221 PMCID: PMC4651367 DOI: 10.1371/journal.pone.0142318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022] Open
Abstract
The destruction of beta cells in type 1 diabetes (T1D) results in loss of insulin production and glucose homeostasis. Treatment of non-obese diabetic (NOD) mice with immune-depleting/modulating agents (e.g., anti-CD3, murine anti-thymocyte-globulin (mATG)) can lead to diabetes reversal. However, for preclinical studies with these and other agents seeking to reverse disease at onset, the necessity for exogenous insulin administration is debated. Spontaneously diabetic NOD mice were treated with a short-course of mATG and insulin provided as drug therapy or by way of allogeneic islet implants. Herein we demonstrate that exogenous insulin administration is required to achieve disease reversal with mATG in NOD mice. Unexpectedly, we also observed that provision of insulin by way of allogeneic islet implantation in combination with mATG leads to a pronounced reversal of diabetes as well as restoration of tolerance to self-islets. Expansion/induction of regulatory cells was observed in NOD mice stably cured with mATG and allogeneic islets. These data suggest that transient provision of allogeneic insulin-producing islets might provide a temporary window for immune depletion to be more effective and instilling stable tolerance to endogenous beta cells. These findings support the use of a never before explored approach for preserving beta cell function in patients with recent onset T1D.
Collapse
|
1515
|
Li J, Zhang Z, Liu X, Wang Y, Mao F, Mao J, Lu X, Jiang D, Wan Y, Lv JY, Cao G, Zhang J, Zhao N, Atkinson M, Greiner DL, Prud'homme GJ, Jiao Z, Li Y, Wang Q. Study of GABA in Healthy Volunteers: Pharmacokinetics and Pharmacodynamics. Front Pharmacol 2015; 6:260. [PMID: 26617516 PMCID: PMC4639630 DOI: 10.3389/fphar.2015.00260] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/20/2015] [Indexed: 01/13/2023] Open
Abstract
Preclinical studies show that GABA exerts anti-diabetic effects in rodent models of type 1 diabetes. Because little is known about its absorption and effects in humans, we investigated the pharmacokinetics and pharmacodynamics of GABA in healthy volunteers. Twelve subjects were subjected to an open-labeled, three-period trial involving sequential oral administration of placebo, 2 g GABA once, and 2 g GABA three times/day for 7 days, with a 7-day washout between each period. GABA was rapidly absorbed (Tmax: 0.5 ~ 1 h) with the half-life (t1/2) of 5 h. No accumulation was observed after repeated oral GABA administration for 7 days. Remarkably, GABA significantly increased circulating insulin levels in the subjects under either fasting (1.6-fold, single dose; 2.0-fold, repeated dose; p < 0.01) or fed conditions (1.4-fold, single dose; 1.6-fold, repeated dose; p < 0.01). GABA also increased glucagon levels only under fasting conditions (1.3-fold, single dose, p < 0.05; 1.5-fold, repeated dose, p < 0.01). However, there were no significant differences in the insulin-to-glucagon ratio and no significant change in glucose levels in these healthy subjects during the study period. Importantly, GABA significantly decreased glycated albumin levels in the repeated dosing period. Subjects with repeated dosing showed an elevated incidence of minor adverse events in comparison to placebo or the single dosing period, most notably transient discomforts such as dizziness and sore throat. However, there were no serious adverse events observed throughout the study. Our data show that GABA is rapidly absorbed and tolerated in human beings; its endocrine effects, exemplified by increasing islet hormonal secretion, suggest potential therapeutic benefits for diabetes.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Xiaoxia Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Yi Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Fei Mao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Junjun Mao
- Department of Pharmacy, Huashan Hospital, Fudan University Shanghai, China
| | - Xiaolan Lu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Dongdong Jiang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Jia-Ying Lv
- Department of Biostatistics, School of Public Health, Fudan University Shanghai, China
| | - Guoying Cao
- Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Institute of Antibiotics, Huashan Hospital, Fudan University Shanghai, China
| | - Jing Zhang
- Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Institute of Antibiotics, Huashan Hospital, Fudan University Shanghai, China
| | - Naiqing Zhao
- Department of Biostatistics, School of Public Health, Fudan University Shanghai, China
| | - Mark Atkinson
- Department of Pathology, College of Medicine, University of Florida Gainesville, FL, USA
| | - Dale L Greiner
- Department of Molecular Medicine, University of Massachusetts Medical School Worcester, MA, USA
| | - Gerald J Prud'homme
- Department of Laboratory Medicine and Pathobiology, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, University of Toronto Toronto, ON, Canada
| | - Zheng Jiao
- Department of Pharmacy, Huashan Hospital, Fudan University Shanghai, China
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China ; Division of Endocrinology and Metabolism, The Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital Toronto, ON, Canada ; Department of Physiology and Medicine, University of Toronto ON, Canada
| |
Collapse
|
1516
|
Shulman R, Miller FA, Daneman D, Guttmann A. Valuing technology: A qualitative interview study with physicians about insulin pump therapy for children with type 1 diabetes. Health Policy 2015; 120:64-71. [PMID: 26563632 DOI: 10.1016/j.healthpol.2015.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 09/28/2015] [Accepted: 10/16/2015] [Indexed: 11/24/2022]
Abstract
Insulin pumps for children with type 1 diabetes have been broadly adopted despite equivocal evidence about comparative effectiveness. To understand why and inform policy related to public funding for new technologies, we explored how physicians interpret the value of pumps. We conducted open-ended, semi-structured interviews with 16 physicians from a pediatric diabetes network in Ontario, Canada, and analyzed the data using interpretive description. Respondents recognized that pumps fell short of expectations because they required hard work, as well as family and school support. Yet, pumps were valued for their status as new technologies and as a promising step in developing future technology. In addition, they were valued for their role within a therapeutic relationship, given the context of chronic childhood disease. These findings identify the types of beliefs that influence the adoption and diffusion of technologies. Some beliefs bear on hopes for new technology that may inappropriately hasten adoption, creating excess cost with little benefit. On the other hand, some beliefs identify potential benefits that are not captured in effectiveness studies, but may warrant consideration in resource allocation decisions. Still others suggest the need for remediation, such as those bearing on disparity in pump use by socioeconomic status. Understanding how technologies are valued can help stakeholders decide how to address such beliefs and expectations in funding decisions and implementation protocols.
Collapse
Affiliation(s)
- Rayzel Shulman
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Fiona A Miller
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Toronto Health Economics and Technology Assessment (THETA) Collaborative, Canada.
| | - Denis Daneman
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Astrid Guttmann
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada; Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| |
Collapse
|
1517
|
Does type 1 diabetes mellitus affect Achilles tendon response to a 10 km run? A case control study. BMC Musculoskelet Disord 2015; 16:345. [PMID: 26556589 PMCID: PMC4641391 DOI: 10.1186/s12891-015-0803-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/03/2015] [Indexed: 11/10/2022] Open
Abstract
Background Achilles tendon structure deteriorates 2-days after maximal loading in elite athletes. The load-response behaviour of tendons may be altered in type 1 diabetes mellitus (T1DM) as hyperglycaemia accelerates collagen cross-linking. This study compared Achilles tendon load-response in participants with T1DM and controls. Methods Achilles tendon structure was quantified at day-0, day-2 and day-4 after a 10 km run. Ultrasound tissue characterisation (UTC) measures tendon structural integrity by classifying pixels as echo-type I, II, III or IV. Echo-type I has the most aligned collagen fibrils and IV has the least. Results Participants were 7 individuals with T1DM and 10 controls. All regularly ran distances greater than 5 km and VISA-A scores indicated good tendon function (T1DM = 94 ± 11, control = 94 ± 10). There were no diabetic complications and HbA1c was 8.7 ± 2.6 mmol/mol for T1DM and 5.3 ± 0.4 mmol/mol for control groups. Baseline tendon structure was similar in T1DM and control groups – UTC echo-types (I-IV) and anterior-posterior thickness were all p > 0.05. No response to load was seen in either T1DM or control group over the 4-days post exercise. Conclusion Active individuals with T1DM do not have a heightened Achilles tendon response to load, which suggests no increased risk of tendon injury. We cannot extrapolate these findings to sedentary individuals with T1DM.
Collapse
|
1518
|
Looney BM, Xia CQ, Concannon P, Ostrov DA, Clare-Salzler MJ. Effects of type 1 diabetes-associated IFIH1 polymorphisms on MDA5 function and expression. Curr Diab Rep 2015; 15:96. [PMID: 26385483 DOI: 10.1007/s11892-015-0656-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent evidence has highlighted the role of the innate immune system in type 1 diabetes (T1D) pathogenesis. Specifically, aberrant activation of the interferon response prior to seroconversion of T1D-associated autoantibodies supports a role for the interferon response as a precipitating event toward activation of autoimmunity. Melanoma differentiation-associated protein 5 (MDA5), encoded by IFIH1, mediates the innate immune system's interferon response to certain viral species that form double-stranded RNA (dsRNA), the MDA5 ligand, during their life cycle. Extensive research has associated single nucleotide polymorphisms (SNPs) within the coding region of IFIH1 with T1D. This review discusses the different risk and protective IFIH1 alleles in the context of recent structural and functional analysis that relate to MDA5 regulation of interferon responses. These studies have provided a functional hypothesis for IFIH1 T1D-associated SNPs' effects on MDA5-mediated interferon responses as well as supporting the genome-wide association (GWA) studies that first associated IFIH1 with T1D.
Collapse
Affiliation(s)
- Benjamin M Looney
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine Interdisciplinary Program in Biomedical Sciences, University of Florida, 1600 SW Archer Rd., Gainesville, FL, 32610, USA.
| | - Chang-Qing Xia
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, 1600 SW Archer Rd., Gainesville, FL, 32610, USA.
| | - Patrick Concannon
- University of Florida Genetics Institute, 2033 Mowry Rd., P.O. Box 103610, Gainesville, FL, 32611, USA.
| | - David A Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 2033 Mowry Rd., P.O. Box 103633, Gainesville, FL, 32611, USA.
| | - Michael J Clare-Salzler
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 2033 Mowry Rd., P.O. Box 103633, Gainesville, FL, 32611, USA.
- Center for Immunology and Transplantation, University of Florida, 1600 SW Archer Rd., P.O. Box 100275, Gainesville, FL, 32610, USA.
| |
Collapse
|
1519
|
Abstract
Type 1 diabetes (T1D) affects 1.93 in 1000 youth in the USA. Over the last 40 years, a combination of genetic and immunological markers has been developed allowing for the accurate prediction of progression to T1D. Despite our abilities to predict disease and the marked improvement in our understanding of the natural history of T1D, therapies capable of preventing or reversing T1D remain elusive. This article will review recent and ongoing efforts to understand the causes of T1D and related efforts to study potential therapies aimed at preventing T1D.
Collapse
Affiliation(s)
| | - Michael J Haller
- University of Florida, PO Box 100296, Gainesville, FL, 32610, USA.
| |
Collapse
|
1520
|
Michels A, Zhang L, Khadra A, Kushner JA, Redondo MJ, Pietropaolo M. Prediction and prevention of type 1 diabetes: update on success of prediction and struggles at prevention. Pediatr Diabetes 2015; 16. [PMID: 26202050 PMCID: PMC4592445 DOI: 10.1111/pedi.12299] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is the archetypal example of a T cell-mediated autoimmune disease characterized by selective destruction of pancreatic β cells. The pathogenic equation for T1DM presents a complex interrelation of genetic and environmental factors, most of which have yet to be identified. On the basis of observed familial aggregation of T1DM, it is certain that there is a decided heritable genetic susceptibility for developing T1DM. The well-known association of T1DM with certain human histocompatibility leukocyte antigen (HLA) alleles of the major histocompatibility complex (MHC) was a major step toward understanding the role of inheritance in T1DM. Type 1 diabetes is a polygenic disease with a small number of genes having large effects (e.g., HLA) and a large number of genes having small effects. Risk of T1DM progression is conferred by specific HLA DR/DQ alleles [e.g., DRB1*03-DQB1*0201 (DR3/DQ2) or DRB1*04-DQB1*0302 (DR4/DQ8)]. In addition, the HLA allele DQB1*0602 is associated with dominant protection from T1DM in multiple populations. A concordance rate lower than 100% between monozygotic twins indicates a potential involvement of environmental factors on disease development. The detection of at least two islet autoantibodies in the blood is virtually pre-diagnostic for T1DM. The majority of children who carry these biomarkers, regardless of whether they have an a priori family history of the disease, will develop insulin-requiring diabetes. Facilitating pre-diagnosis is the timing of seroconversion which is most pronounced in the first 2 yr of life. Unfortunately the significant progress in improving prediction of T1DM has not yet been paralleled by safe and efficacious intervention strategies aimed at preventing the disease. Herein we summarize the chequered history of prediction and prevention of T1DM, describing successes and failures alike, and thereafter examine future trends in the exciting, partially explored field of T1DM prevention.
Collapse
Affiliation(s)
- Aaron Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado
| | - Li Zhang
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, QC Canada
| | - Jake A. Kushner
- Division of Diabetes Pediatric Endocrinology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Maria J. Redondo
- Division of Diabetes Pediatric Endocrinology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Massimo Pietropaolo
- Division of Diabetes, Endocrinology and Metabolism, McNair Medical Institute, Baylor College of Medicine, Houston, Texas,To Whom Correspondence May be Addressed: Massimo Pietropaolo, M.D., Division of Diabetes, Endocrinology and Metabolism, Alkek Building for Biomedical Research, R 609, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030
| |
Collapse
|
1521
|
Mathews CE, Xue S, Posgai A, Lightfoot YL, Li X, Lin A, Wasserfall C, Haller MJ, Schatz D, Atkinson MA. Acute Versus Progressive Onset of Diabetes in NOD Mice: Potential Implications for Therapeutic Interventions in Type 1 Diabetes. Diabetes 2015; 64:3885-90. [PMID: 26216853 PMCID: PMC4613974 DOI: 10.2337/db15-0449] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/21/2015] [Indexed: 12/20/2022]
Abstract
Most natural history models for type 1 diabetes (T1D) propose that overt hyperglycemia results after a progressive loss of insulin-secreting β-cell mass and/or function. To experimentally address this concept, we prospectively determined morning blood glucose measurements every other day in multiple cohorts (total n = 660) of female NOD/ShiLtJ mice starting at 8 weeks of age until diabetes onset or 26 weeks of age. Consistent with this notion, a majority of mice that developed diabetes (354 of 489 [72%]) displayed a progressive increase in blood glucose with transient excursions >200 mg/dL, followed by acute and persistent hyperglycemia at diabetes onset. However, 135 of the 489 (28%) diabetic animals demonstrated normal glucose values followed by acute (i.e., sudden) hyperglycemia. Interestingly, diabetes onset occurred earlier in mice with acute versus progressive disease onset (15.37 ± 0.3207 vs. 17.44 ± 0.2073 weeks of age, P < 0.0001). Moreover, the pattern of onset (i.e., progressive vs. acute) dramatically influenced the ability to achieve reversal of T1D by immunotherapeutic intervention, with increased effectiveness observed in situations of a progressive deterioration in euglycemia. These studies highlight a novel natural history aspect in this animal model, one that may provide important guidance for the selection of subjects participating in human trials seeking disease reversal.
Collapse
Affiliation(s)
| | - Song Xue
- Department of Pathology, University of Florida, Gainesville, FL
| | - Amanda Posgai
- Department of Pathology, University of Florida, Gainesville, FL
| | | | - Xia Li
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital and the Diabetes Center, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
| | - Andrea Lin
- Department of Pathology, University of Florida, Gainesville, FL
| | | | - Michael J Haller
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Desmond Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Mark A Atkinson
- Department of Pathology, University of Florida, Gainesville, FL
| |
Collapse
|
1522
|
Vitamin D fortification and seasonality of birth in type 1 diabetic cases: D-tect study. J Dev Orig Health Dis 2015; 7:114-9. [PMID: 26503327 DOI: 10.1017/s2040174415007849] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Fortification of margarine with vitamin D was mandatory in Denmark during 1961-1985. The aim of the study was to assess whether gestational and early infancy exposure to margarine fortification was associated with seasonality of birth in Danish type 1 diabetes (T1D) patients. The risks of T1D in Danes born during various exposure periods around margarine fortification termination in 1985 were analyzed. As expected, the T1D hazards in males unexposed to margarine fortification and born in spring were higher than in males born in autumn: relevant hazard ratios (95% confidence intervals) in various exposure groups ranged from 1.74 (1.112/2.708) to 37.43 (1.804/776.558). There were no indications of seasonality of birth in males exposed to fortification, nor in both exposed and unexposed females. The study suggests that early life exposure to low-dose vitamin D from fortified food eliminates seasonality of birth in T1D male patients. Further studies are required to investigate the identified gender differences.
Collapse
|
1523
|
New approaches for predicting T cell-mediated drug reactions: A role for inducible and potentially preventable autoimmunity. J Allergy Clin Immunol 2015; 136:252-7. [PMID: 26254052 DOI: 10.1016/j.jaci.2015.06.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/23/2015] [Accepted: 06/25/2015] [Indexed: 12/20/2022]
Abstract
Adverse drug reactions (ADRs) are commonplace and occur when a drug binds to its intended pharmacologic target (type A ADR) or an unintended target (type B ADR). Immunologically mediated type B ADRs, such as drug hypersensitivity syndrome, drug reaction with eosinophilia and systemic symptoms syndrome, and Stevens-Johnson syndrome/toxic epidermal necrolysis, can be severe and result in a diverse set of clinical manifestations that include fever and rash, as well as multiple organ failure (liver, kidney, lungs, and/or heart) in the case of drug hypersensitivity syndrome. There is increasing evidence that specific HLA alleles influence the risk of drug reactions. Several features of T cell-mediated ADRs are strikingly similar to those displayed by patients with autoimmune diseases like type I diabetes, such as strong HLA association, organ-specific adaptive immune responses, viral involvement, and activation of innate immunity. There is a need to better predict patient populations at risk for immunologically mediated type B ADRs. Because methods to predict type 1 diabetes by using genetic and immunologic biomarkers have been developed to a high level of accuracy (predicting 100% of subjects likely to progress), new research strategies based on these methods might also improve the ability to predict drug hypersensitivity.
Collapse
|
1524
|
D'Addio F, La Rosa S, Maestroni A, Jung P, Orsenigo E, Nasr MB, Tezza S, Bassi R, Finzi G, Marando A, Vergani A, Frego R, Albarello L, Andolfo A, Manuguerra R, Viale E, Staudacher C, Corradi D, Batlle E, Breault D, Secchi A, Folli F, Fiorina P. Circulating IGF-I and IGFBP3 Levels Control Human Colonic Stem Cell Function and Are Disrupted in Diabetic Enteropathy. Cell Stem Cell 2015; 17:486-498. [PMID: 26431183 PMCID: PMC4826279 DOI: 10.1016/j.stem.2015.07.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 06/02/2015] [Accepted: 07/19/2015] [Indexed: 01/10/2023]
Abstract
The role of circulating factors in regulating colonic stem cells (CoSCs) and colonic epithelial homeostasis is unclear. Individuals with long-standing type 1 diabetes (T1D) frequently have intestinal symptoms, termed diabetic enteropathy (DE), though its etiology is unknown. Here, we report that T1D patients with DE exhibit abnormalities in their intestinal mucosa and CoSCs, which fail to generate in vitro mini-guts. Proteomic profiling of T1D+DE patient serum revealed altered levels of insulin-like growth factor 1 (IGF-I) and its binding protein 3 (IGFBP3). IGFBP3 prevented in vitro growth of patient-derived organoids via binding its receptor TMEM219, in an IGF-I-independent manner, and disrupted in vivo CoSC function in a preclinical DE model. Restoration of normoglycemia in patients with long-standing T1D via kidney-pancreas transplantation or in diabetic mice by treatment with an ecto-TMEM219 recombinant protein normalized circulating IGF-I/IGFBP3 levels and reestablished CoSC homeostasis. These findings demonstrate that peripheral IGF-I/IGFBP3 controls CoSCs and their dysfunction in DE.
Collapse
Affiliation(s)
- Francesca D'Addio
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston 02115, MA, USA
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Stefano La Rosa
- Department of Pathology, Ospedale di Circolo, Varese 21100, Italy
| | - Anna Maestroni
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Peter Jung
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona 08028, Spain
| | - Elena Orsenigo
- Surgery, Protein Microsequencing Facility, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston 02115, MA, USA
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Sara Tezza
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston 02115, MA, USA
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Roberto Bassi
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston 02115, MA, USA
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Giovanna Finzi
- Department of Pathology, Ospedale di Circolo, Varese 21100, Italy
| | | | - Andrea Vergani
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston 02115, MA, USA
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Roberto Frego
- Gastroenterology, Protein Microsequencing Facility, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Luca Albarello
- Pathology Unit, Protein Microsequencing Facility, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Annapaola Andolfo
- ProMiFa, Protein Microsequencing Facility, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Roberta Manuguerra
- Department of Biomedical, Biotechnological and Translational Sciences, Unit of Pathology, University of Parma, Parma 43121, Italy
| | - Edi Viale
- Gastroenterology, Protein Microsequencing Facility, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Carlo Staudacher
- Surgery, Protein Microsequencing Facility, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Domenico Corradi
- Department of Biomedical, Biotechnological and Translational Sciences, Unit of Pathology, University of Parma, Parma 43121, Italy
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona 08028, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08028, Spain
| | - David Breault
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Antonio Secchi
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
- Vita Salute San Raffaele University, Milano 20132, Italy
| | - Franco Folli
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio 78229, Texas, USA
- Department of Internal Medicine, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas, São Paulo 13100, Brazil
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston 02115, MA, USA
- Transplant Medicine, IRCCS Ospedale San Raffaele, Milan 20132, Italy
| |
Collapse
|
1525
|
Insel RA, Dunne JL, Atkinson MA, Chiang JL, Dabelea D, Gottlieb PA, Greenbaum CJ, Herold KC, Krischer JP, Lernmark Å, Ratner RE, Rewers MJ, Schatz DA, Skyler JS, Sosenko JM, Ziegler AG. Staging presymptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association. Diabetes Care 2015; 38:1964-74. [PMID: 26404926 PMCID: PMC5321245 DOI: 10.2337/dc15-1419] [Citation(s) in RCA: 693] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Insights from prospective, longitudinal studies of individuals at risk for developing type 1 diabetes have demonstrated that the disease is a continuum that progresses sequentially at variable but predictable rates through distinct identifiable stages prior to the onset of symptoms. Stage 1 is defined as the presence of β-cell autoimmunity as evidenced by the presence of two or more islet autoantibodies with normoglycemia and is presymptomatic, stage 2 as the presence of β-cell autoimmunity with dysglycemia and is presymptomatic, and stage 3 as onset of symptomatic disease. Adoption of this staging classification provides a standardized taxonomy for type 1 diabetes and will aid the development of therapies and the design of clinical trials to prevent symptomatic disease, promote precision medicine, and provide a framework for an optimized benefit/risk ratio that will impact regulatory approval, reimbursement, and adoption of interventions in the early stages of type 1 diabetes to prevent symptomatic disease.
Collapse
Affiliation(s)
| | | | - Mark A Atkinson
- UF Diabetes Institute, University of Florida, Gainesville, FL
| | | | - Dana Dabelea
- Colorado School of Public Health, University of Colorado, Denver, CO
| | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO
| | | | - Kevan C Herold
- Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Jeffrey P Krischer
- Department of Pediatrics, Pediatric Epidemiology Center, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Åke Lernmark
- Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | | | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO
| | | | - Jay S Skyler
- Diabetes Research Institute, University of Miami, Miami, FL
| | - Jay M Sosenko
- Diabetes Research Institute, University of Miami, Miami, FL
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Munich and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, Neuherberg, Germany
| |
Collapse
|
1526
|
Narendran P. Alternative Approach to Immunomodulation for Type 1 Diabetes: Antigen-Specific Immunotherapy In Utero. Diabetes 2015; 64:3347-9. [PMID: 26405274 DOI: 10.2337/db15-0825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Parth Narendran
- University of Birmingham, and Queen Elizabeth Hospital Birmingham, Birmingham, U.K.
| |
Collapse
|
1527
|
Xiang L, Mittwede PN, Clemmer JS. Glucose Homeostasis and Cardiovascular Alterations in Diabetes. Compr Physiol 2015; 5:1815-39. [PMID: 26426468 DOI: 10.1002/cphy.c150001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
1528
|
George NM, Boerner BP, Mir SUR, Guinn Z, Sarvetnick NE. Exploiting Expression of Hippo Effector, Yap, for Expansion of Functional Islet Mass. Mol Endocrinol 2015; 29:1594-607. [PMID: 26378466 DOI: 10.1210/me.2014-1375] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Loss of pancreas β-cell function is the precipitating factor in all forms of diabetes. Cell replacement therapies, such as islet transplantation, remain the best hope for a cure; however, widespread implementation of this method is hampered by availability of donor tissue. Thus, strategies that expand functional β-cell mass are crucial for widespread usage in diabetes cell replacement therapy. Here, we investigate the regulation of the Hippo-target protein, Yes-associated protein (Yap), during development of the endocrine pancreas and its function after reactivation in human cadaveric islets. Our results demonstrate that Yap expression is extinguished at the mRNA level after neurogenin-3-dependent specification of the pancreas endocrine lineage, correlating with proliferation decreases in these cells. Interestingly, when a constitutively active form of Yap was expressed in human cadaver islets robust increases in proliferation were noted within insulin-producing β-cells. Importantly, proliferation in these cells occurs without negatively affecting β-cell differentiation or functional status. Finally, we show that the proproliferative mammalian target of rapamycin pathway is activated after Yap expression, providing at least one explanation for the observed increases in β-cell proliferation. Together, these results provide a foundation for manipulating Yap activity as a novel approach to expand functional islet mass for diabetes regenerative therapy.
Collapse
Affiliation(s)
- Nicholas M George
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Brian P Boerner
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Shakeel U R Mir
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Zachary Guinn
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Nora E Sarvetnick
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| |
Collapse
|
1529
|
Godehardt AW, Schilling-Leiß D, Sanzenbacher R, Tönjes RR. [Xenogeneic cell therapeutics: Treatment of type 1 diabetes using porcine pancreatic islets and islet cells]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2015; 58:1281-8. [PMID: 26369761 DOI: 10.1007/s00103-015-2246-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In view of the existing shortage of human donor organs and tissues, xenogeneic cell therapeutics (xCT) offer an alternative for adequate treatment. In particular, porcine pancreatic islets and islet cells have already entered the field of experimental therapy for type-1 diabetes mellitus (T1DM) patients. Thereby, xCT depict challenging products with a glance on medical, ethical, and regulatory questions. With cross-species transplantation (xenotransplantation), the risk of immunological graft rejection as well as the risk of infectious transmission of microbial and viral pathogens must be considered. This includes the bidirectional transmission of microorganisms from graft to host as well as from host to graft. Crossing the border of species requires a critical risk-benefit evaluation as well as a thorough longtime surveillance of transplant recipients after treatment. The international legal and regulatory requirements for xCT are inter alia based on the World Health Organization criteria summarized in the Changsha Communiqué (2008). In the European Union, they were reflected by the European Medicines Agency (EMA) Guideline on Xenogeneic Cell-based Medicinal Products following the implementation of the Regulation on Advanced Therapies (ATMP). On the basis of this regulation, the first non-clinical and clinical experiences were obtained for porcine islets. The results suggest that supportive treatment of T1DM risk patients with xCT may be an alternative to established allogeneic organ transplantation in the future.
Collapse
Affiliation(s)
- Antonia W Godehardt
- Abteilung Medizinische Biotechnologie, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Deutschland.
| | - Dagmar Schilling-Leiß
- Abteilung Medizinische Biotechnologie, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Deutschland
| | - Ralf Sanzenbacher
- Abteilung Medizinische Biotechnologie, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Deutschland
| | - Ralf R Tönjes
- Abteilung Medizinische Biotechnologie, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Deutschland
| |
Collapse
|
1530
|
Chujo D, Nguyen TS, Foucat E, Blankenship D, Banchereau J, Nepom GT, Chaussabel D, Ueno H. Adult-onset type 1 diabetes patients display decreased IGRP-specific Tr1 cells in blood. Clin Immunol 2015; 161:270-7. [PMID: 26341315 DOI: 10.1016/j.clim.2015.08.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 11/26/2022]
Abstract
The breakdown of immune tolerance against islet antigens causes type 1 diabetes (T1D). The antigens associated with adult-onset T1D (AT1D) remain largely undefined. It is possible that AT1D patients display a unique type of CD4(+) T cells specific for a certain islet antigen. Here we analyzed the cytokine production profiles of CD4(+) helper T (Th) cells that are specific for three islet antigens; GAD65, preproinsulin, and IGRP in patients with AT1D, juvenile-onset T1D (JT1D), and age-, gender- and human leukocyte antigen (HLA)-matched control adults. While IGRP-specific Th cells in AT1D patients were dominantly Th1 cells, IGRP-specific Th cells in control adults and JT1D patients were dominantly Th2 and T regulatory type 1 (Tr1) cells. Notably, the frequency of IGRP-specific Tr1 cells was significantly lower in AT1D patients than in control adults and JT1D patients. In conclusion, our study suggests that IGRP-specific Th cells play a unique pathogenic role in AT1D.
Collapse
Affiliation(s)
- Daisuke Chujo
- Baylor Institute for Immunology Research, Dallas, TX, USA; National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Emile Foucat
- Baylor Institute for Immunology Research, Dallas, TX, USA
| | | | | | - Gerald T Nepom
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | | | - Hideki Ueno
- Baylor Institute for Immunology Research, Dallas, TX, USA.
| |
Collapse
|
1531
|
Wang B, Hawa MI, Rijsdijk FV, Fain PR, Paschou SA, Boehm BO, Steck AK, Snieder H, Leslie RDG. Heritability of thyroid peroxidase autoantibody levels in type 1 diabetes: evidence from discordant twin pairs. Diabetologia 2015; 58:2079-86. [PMID: 26070305 PMCID: PMC4886710 DOI: 10.1007/s00125-015-3664-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/20/2015] [Indexed: 01/22/2023]
Abstract
AIMS/HYPOTHESIS The discordance status of (autoimmune) type 1 diabetes within monozygotic twin pairs points to the importance of environmental factors. The aim of this study was to investigate whether the environmental events causing type 1 diabetes influence thyroid autoimmunity. METHODS Monozygotic and dizygotic twins discordant for type 1 diabetes from the UK and USA were tested for thyroid peroxidase autoantibodies (TPOA) by radioimmunoassay. Using quantitative genetic model fitting of a liability-threshold model we estimated the contribution of genetic (heritability) and environmental factors to TPOA. RESULTS TPOA positivity was higher in females than in males in both cohorts and was associated with later age at diagnosis in the UK and combined cohorts (p < 0.01). TPOA did not specifically segregate with type 1 diabetes in the twin pairs (p > 0.2 in all groups). The best-fitting models showed heritability (95% CI) estimates for TPOA of 63% (37%, 80%) for the UK and 80% (51%, 92%) for US twins, while the best-fitting meta-analysis model of the two twin cohorts combined included additive genetic and unique environmental factors with a heritability estimate of 69% (50%, 82%). CONCLUSIONS/INTERPRETATION Risk of thyroid autoimmunity, defined by TPOA, in the context of autoimmune diabetes is, substantially, genetically determined in discordant twin pairs. Environmental factors leading to type 1 diabetes were not the same as those involved with thyroid autoimmunity. It follows that it is as important to investigate for thyroid autoimmunity in relatives of type 1 diabetes patients as it is in the patients themselves.
Collapse
Affiliation(s)
- Bin Wang
- Department of Epidemiology, University of Groningen, University Medical Center, Groningen, the Netherlands
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, People’s Republic of China
| | - Mohammed I. Hawa
- Centre for Diabetes and Metabolic Medicine, The Blizard Institute, Queen Mary, University of London, London E1 2AT, UK
| | - Frühling V. Rijsdijk
- Institute of Psychiatry, Department of Psychosis Studies, King’s College London, London, UK
| | - Pamela R. Fain
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, USA
| | - Stavroula A. Paschou
- Centre for Diabetes and Metabolic Medicine, The Blizard Institute, Queen Mary, University of London, London E1 2AT, UK
| | - Bernhard O. Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore and Imperial College London, London, UK
- Department of Internal Medicine 1, Ulm University Medical Centre, Ulm, Germany
| | - Andrea K. Steck
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center, Groningen, the Netherlands
| | - R. David G. Leslie
- Centre for Diabetes and Metabolic Medicine, The Blizard Institute, Queen Mary, University of London, London E1 2AT, UK
| |
Collapse
|
1532
|
Wherrett DK. Trials in the prevention of type 1 diabetes: current and future. Can J Diabetes 2015; 38:279-84. [PMID: 25092646 DOI: 10.1016/j.jcjd.2014.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 05/04/2014] [Accepted: 05/05/2014] [Indexed: 12/19/2022]
Abstract
A major thrust in type 1 diabetes research is stopping the destruction of beta cells that leads to type 1 diabetes. Research over the past 30 years has defined genetic factors and evidence of autoimmunity that have led to the development of robust prediction models in those at high risk for type 1 diabetes. The ability to identify those at risk and the development of new agents and of collaborative research networks has led to multiple trials aimed at preventing beta cell loss. Trials at all stages of beta cell loss have been conducted: primary prevention (prior to the development of autoimmunity); secondary prevention (after autoantibodies are found) and tertiary prevention (intervening after diagnosis to maintain remaining beta cells). Studies have shown mixed results; evidence of maintained insulin secretion after the time of diagnosis has been described in a number of studies, and primary and secondary prevention is proving to be elusive. Much has been learned from the increasing number of studies in the field in terms of network creation, study design and choice of intervention that will facilitate new avenues of investigation.
Collapse
Affiliation(s)
- Diane K Wherrett
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
1533
|
The Role of Gluten in Celiac Disease and Type 1 Diabetes. Nutrients 2015; 7:7143-62. [PMID: 26343710 PMCID: PMC4586524 DOI: 10.3390/nu7095329] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/10/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022] Open
Abstract
Celiac disease (CD) and type 1 diabetes (T1D) are autoimmune conditions in which dietary gluten has been proven or suggested to play a pathogenic role. In CD; gluten is established as the instigator of autoimmunity; the autoimmune process is halted by removing gluten from the diet; which allows for resolution of celiac autoimmune enteropathy and subsequent normalization of serological markers of the disease. However; an analogous causative agent has not yet been identified for T1D. Nevertheless; the role of dietary gluten in development of T1D and the potentially beneficial effect of removing gluten from the diet of patients with T1D are still debated. In this review; we discuss the comorbid occurrence of CD and T1D and explore current evidences for the specific role of gluten in both conditions; specifically focusing on current evidence on the effect of gluten on the immune system and the gut microbiota.
Collapse
|
1534
|
Burch TC, Morris MA, Campbell-Thompson M, Pugliese A, Nadler JL, Nyalwidhe JO. Proteomic Analysis of Disease Stratified Human Pancreas Tissue Indicates Unique Signature of Type 1 Diabetes. PLoS One 2015; 10:e0135663. [PMID: 26302420 PMCID: PMC4547762 DOI: 10.1371/journal.pone.0135663] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/09/2015] [Indexed: 11/18/2022] Open
Abstract
Type 1 diabetes (T1D) and type 2 diabetes (T2D) are associated with functional beta cell loss due to ongoing inflammation. Despite shared similarities, T1D is an autoimmune disease with evidence of autoantibody production, as well as a role for exocrine pancreas involvement. Our hypothesis is that differential protein expression occurs in disease stratified pancreas tissues and regulated proteins from endocrine and exocrine tissues are potential markers of disease and potential therapeutic targets. The study objective was to identify novel proteins that distinguish the pancreas from donors with T1D from the pancreas from patients with T2D, or autoantibody positive non-diabetic donors. Detailed quantitative comprehensive proteomic analysis was applied to snap frozen human pancreatic tissue lysates from organ donors without diabetes, with T1D-associated autoantibodies in the absence of diabetes, with T1D, or with T2D. These disease-stratified human pancreas tissues contain exocrine and endocrine tissues (with dysfunctional islets) in the same microenvironment. The expression profiles of several of the proteins were further verified by western blot. We identified protein panels that are significantly and uniquely upregulated in the three disease-stratified pancreas tissues compared to non-disease control tissues. These proteins are involved in inflammation, metabolic regulation, and autoimmunity, all of which are pathways linked to, and likely involved in, T1 and T2 diabetes pathogenesis. Several new proteins were differentially upregulated in prediabetic, T1D, and T2D pancreas. The results identify proteins that could serve as novel prognostic, diagnostic, and therapeutic tools to preserve functional islet mass in Type 1 Diabetes.
Collapse
Affiliation(s)
- Tanya C. Burch
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States of America
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States of America
| | - Margaret A. Morris
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States of America
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, United States of America
- Strelitz Diabetes Research Center, Eastern Virginia Medical School, Norfolk, VA, United States of America
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Gainesville, FL, United States of America
| | - Alberto Pugliese
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States of America
| | - Jerry L. Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, United States of America
- Strelitz Diabetes Research Center, Eastern Virginia Medical School, Norfolk, VA, United States of America
- * E-mail: (JLN); (JON)
| | - Julius O. Nyalwidhe
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States of America
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States of America
- * E-mail: (JLN); (JON)
| |
Collapse
|
1535
|
Hanes WM, Olofsson PS, Kwan K, Hudson LK, Chavan SS, Pavlov VA, Tracey KJ. Galantamine Attenuates Type 1 Diabetes and Inhibits Anti-Insulin Antibodies in Nonobese Diabetic Mice. Mol Med 2015; 21:702-708. [PMID: 26322849 DOI: 10.2119/molmed.2015.00142] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/17/2015] [Indexed: 01/01/2023] Open
Abstract
Type 1 diabetes in mice is characterized by autoimmune destruction of insulin-producing pancreatic β-cells. Disease pathogenesis involves invasion of pancreatic islets by immune cells, including macrophages and T cells, and production of antibodies to self-antigens, including insulin. Activation of the inflammatory reflex, the neural circuit that inhibits inflammation, culminates on cholinergic receptor signals on immune cells to attenuate cytokine release and inhibit B-cell antibody production. Here, we show that galantamine, a centrally acting acetylcholinesterase inhibitor and an activator of the inflammatory reflex, attenuates murine experimental type 1 diabetes. Administration of galantamine to animals immunized with keyhole limpet hemocyanin (KLH) significantly suppressed splenocyte release of immunoglobulin G (IgG) and interleukin (IL)-4 and IL-6 during KLH challenge ex vivo. Administration of galantamine beginning at 1 month of age in nonobese diabetic (NOD) mice significantly delayed the onset of hyperglycemia, attenuated immune cell infiltration in pancreatic islets and decreased anti-insulin antibodies in serum. These observations indicate that galantamine attenuates experimental type 1 diabetes in mice and suggest that activation of the inflammatory reflex should be further studied as a potential therapeutic approach.
Collapse
Affiliation(s)
- William M Hanes
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America.,Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Peder S Olofsson
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Kevin Kwan
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - LaQueta K Hudson
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Sangeeta S Chavan
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Valentin A Pavlov
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Kevin J Tracey
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| |
Collapse
|
1536
|
Presa M, Chen YG, Grier AE, Leiter EH, Brehm MA, Greiner DL, Shultz LD, Serreze DV. The Presence and Preferential Activation of Regulatory T Cells Diminish Adoptive Transfer of Autoimmune Diabetes by Polyclonal Nonobese Diabetic (NOD) T Cell Effectors into NSG versus NOD-scid Mice. THE JOURNAL OF IMMUNOLOGY 2015; 195:3011-9. [PMID: 26283479 DOI: 10.4049/jimmunol.1402446] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 07/23/2015] [Indexed: 01/18/2023]
Abstract
NOD-scid.Il2rg(null) (NSG) mice are currently being used as recipients to screen for pathogenic autoreactive T cells in type 1 diabetes (T1D) patients. We questioned whether the restriction of IL-2R γ-chain (Il-2rγ)-dependent cytokine signaling only to donor cells in NSG recipients differently influenced the activities of transferred diabetogenic T cells when they were introduced as a monoclonal/oligoclonal population versus being part of a polyclonal repertoire. Unexpectedly, a significantly decreased T1D transfer by splenocytes from prediabetic NOD donors was observed in Il-2rγ(null)-NSG versus Il-2rγ-intact standard NOD-scid recipients. In contrast, NOD-derived monoclonal/oligoclonal TCR transgenic β cell-autoreactive T cells in either the CD8 (AI4, NY8.3) or CD4 (BDC2.5) compartments transferred disease significantly more rapidly to NSG than to NOD-scid recipients. The reduced diabetes transfer efficiency by polyclonal T cells in NSG recipients was associated with enhanced activation of regulatory T cells (Tregs) mediated by NSG myeloid APC. This enhanced suppressor activity was associated with higher levels of Treg GITR expression in the presence of NSG than NOD-scid APC. These collective results indicate NSG recipients might be efficiently employed to test the activity of T1D patient-derived β cell-autoreactive T cell clones and lines, but, when screening for pathogenic effectors within polyclonal populations, Tregs should be removed from the transfer inoculum to avoid false-negative results.
Collapse
Affiliation(s)
| | - Yi-Guang Chen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226; and
| | | | | | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Dale L Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | | | | |
Collapse
|
1537
|
Wen H, Jung H, Li X. Drug Delivery Approaches in Addressing Clinical Pharmacology-Related Issues: Opportunities and Challenges. AAPS JOURNAL 2015; 17:1327-40. [PMID: 26276218 DOI: 10.1208/s12248-015-9814-9] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 08/06/2015] [Indexed: 12/27/2022]
Abstract
Various drug delivery approaches can be used to maximize therapeutic efficacy and minimize side effects, by impacting absorption, distribution, metabolism, and elimination (ADME) of a drug compound. For those drugs with poor water solubility or low permeability, techniques such as amorphous solid dispersion, liposomes, and complexations have been used to improve their oral bioavailability. Modified release (MR) formulations have been widely used to improve patient compliance, as well as to reduce side effects, especially for those drugs with short half-lives or narrow therapeutic windows. More than ten drugs using sterile long-acting release (LAR) formulations with clear clinical benefit have been successfully marketed. Furthermore, drug delivery systems have been used in delaying drug clearance processes. Additionally, modifying the in vivo drug distribution using targeted delivery systems has significantly improved oncology treatments. All the drug delivery approaches have their advantages and limitations. For both brand and generic drugs, the achievement of consistent quality and therapeutic performance using drug delivery systems can also pose serious challenges in developing a drug for the market, which requires close collaboration among industry, academia, and regulatory agencies. With the advent of personalized medicines, there will be great opportunities and challenges in utilizing drug delivery systems to provide better products and services for patients.
Collapse
Affiliation(s)
- Hong Wen
- Office of Generic Drugs, CDER, FDA, Silver Spring, Maryland, 20993, USA.
| | - Huijeong Jung
- Office of Generic Drugs, CDER, FDA, Silver Spring, Maryland, 20993, USA
| | - Xuhong Li
- Office of Pharmaceutical Quality, CDER, FDA, Silver Spring, Maryland, 20993, USA
| |
Collapse
|
1538
|
Tallapragada DSP, Bhaskar S, Chandak GR. New insights from monogenic diabetes for "common" type 2 diabetes. Front Genet 2015; 6:251. [PMID: 26300908 PMCID: PMC4528293 DOI: 10.3389/fgene.2015.00251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/13/2015] [Indexed: 01/17/2023] Open
Abstract
Boundaries between monogenic and complex genetic diseases are becoming increasingly blurred, as a result of better understanding of phenotypes and their genetic determinants. This had a large impact on the way complex disease genetics is now being investigated. Starting with conventional approaches like familial linkage, positional cloning and candidate genes strategies, the scope of complex disease genetics has grown exponentially with scientific and technological advances in recent times. Despite identification of multiple loci harboring common and rare variants associated with complex diseases, interpreting and evaluating their functional role has proven to be difficult. Information from monogenic diseases, especially related to the intermediate traits associated with complex diseases comes handy. The significant overlap between traits and phenotypes of monogenic diseases with related complex diseases provides a platform to understand the disease biology better. In this review, we would discuss about one such complex disease, type 2 diabetes, which shares marked similarity of intermediate traits with different forms of monogenic diabetes.
Collapse
Affiliation(s)
| | | | - Giriraj R. Chandak
- Genomic Research on Complex Diseases Laboratory, Council of Scientific and Industrial Research-Centre for Cellular and Molecular BiologyHyderabad, India
| |
Collapse
|
1539
|
Abstract
PURPOSE OF REVIEW The purpose of this review was to discuss the evidence suggesting that our definition of type 1 diabetes (T1D) needs to change to incorporate new data about risk of disease and how we should intervene in this wider spectrum of disease. RECENT FINDINGS Autoantibodies can predict the development of T1D within defined periods of time. Other biomarkers are being developed that may further characterize stages of disease, as well as help define new pathways for treatment to prevent the development of hyperglycemia and full-blown diabetes. SUMMARY We have entered a new age in which prevention of disease is now possible based on changing the definition of diabetes to one that incorporates immunological markers that bring with them high risk. Further advances in our knowledge will continue to refine our ability to predict and hopefully eventually prevent T1D.
Collapse
Affiliation(s)
- Peter A Gottlieb
- Pediatrics and Medicine, Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
1540
|
Niinistö S, Takkinen HM, Uusitalo L, Rautanen J, Vainio N, Ahonen S, Nevalainen J, Kenward MG, Lumia M, Simell O, Veijola R, Ilonen J, Knip M, Virtanen SM. Maternal intake of fatty acids and their food sources during lactation and the risk of preclinical and clinical type 1 diabetes in the offspring. Acta Diabetol 2015; 52:763-72. [PMID: 25563476 DOI: 10.1007/s00592-014-0673-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 10/20/2014] [Indexed: 12/30/2022]
Abstract
AIMS We examined maternal dietary intake of fatty acids and foods which are sources of fatty acids during lactation and whether they are associated with the risk of preclinical and clinical type 1 diabetes in the offspring. METHODS The subjects comprised a cohort of 2,939 mother-child pairs from the prospective Type 1 Diabetes Prediction and Prevention Study. Composition of maternal diet during the third month of lactation was assessed by a validated food frequency questionnaire. Among the children with HLA-conferred susceptibility to type 1 diabetes, 172 developed preclinical and 81 clinical diabetes. Average follow-up for preclinical type 1 diabetes was 7.5 years (range 0.2-14.0 years) and for clinical type 1 diabetes 7.7 years (0.2-14.0 years). RESULTS Maternal intake of fatty acids during lactation was not associated with the risk of type 1 diabetes in the offspring. After adjusting for putative confounders, maternal total consumption of red meat and meat products during lactation was associated both with increased risk for preclinical [hazard ratio (HR) 1.19, 95 % CI 1.02-1.40, p = 0.038] and clinical type 1 diabetes (HR 1.27, 95 % CI 1.06-1.52, p = 0.025). In particular, consumption of processed meat products showed an association with increased risk for type 1 diabetes (HR 1.23, 95 % CI 1.02-1.48, p = 0.045). Maternal use of vegetable oils was associated with increased risk for preclinical type 1 diabetes (HR 1.21, 95 % CI 1.03-1.41, p = 0.023). CONCLUSIONS Maternal consumption of red meat, especially processed meat, during lactation may increase the risk of type 1 diabetes.
Collapse
Affiliation(s)
- S Niinistö
- Nutrition Unit, Department of Lifestyle and Participation, National Institute for Health and Welfare, P.O. Box 30, 00271, Helsinki, Finland,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1541
|
Insel RA, Dunne JL, Ziegler AG. General population screening for type 1 diabetes: has its time come? Curr Opin Endocrinol Diabetes Obes 2015; 22:270-6. [PMID: 26087338 DOI: 10.1097/med.0000000000000173] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review was to describe the potential for general childhood population-based screening of risk of symptomatic type 1 diabetes (T1D) RECENT FINDINGS: The earliest stages of T1D can be identified and risk and rate of progression to symptomatic disease can be estimated by the presence of multiple islet autoantibodies and glucose intolerance (dysglycemia) in individuals screened for risk. Screening for human leukocyte antigen risk genotypes in neonates with follow-up detection of islet autoantibodies in childhood has been explored. An alternative approach of general childhood population-based detection of autoantibodies at well child visits provides an approach to detect a high proportion of children who will develop T1D. The Fr1da study was launched in Bavaria in 2015 to explore this concept. SUMMARY General childhood population-based screening for risk of T1D will allow detection of an at-risk population that can participate in natural history studies to better understand disease pathogenesis and intervention trials to prevent symptomatic disease and will provide a framework for public health-based prevention of childhood-onset T1D.
Collapse
Affiliation(s)
- Richard A Insel
- aJDRF, New York, USA bInstitute of Diabetes Research, Helmholtz Zentrum München, Munich cForschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, Neuherberg, Germany
| | | | | |
Collapse
|
1542
|
Poudel A, Savari O, Striegel DA, Periwal V, Taxy J, Millis JM, Witkowski P, Atkinson MA, Hara M. Beta-cell destruction and preservation in childhood and adult onset type 1 diabetes. Endocrine 2015; 49:693-702. [PMID: 25605478 PMCID: PMC4511725 DOI: 10.1007/s12020-015-0534-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/12/2015] [Indexed: 01/09/2023]
Abstract
Previous studies describing the symptomatic onset of type 1 diabetes (T1D) and rate of beta-cell loss (C-peptide) support the notion that childhood onset T1D exhibits more severe beta-cell depletion compared to adult onset T1D. To test this notion, we performed whole pancreas analyses in two T1D cases, one of childhood onset (7-year old, onset at 1.5-year) along with an adult onset case (43-year old with onset at 27-year). Both cases were matched for age and gender with control subjects. Striking regional differences in beta-cell loss were observed in both T1D cases, with severity of loss in the order of tail > body > head regions. In contrast, pancreatic alpha- and delta-cell mass was similar in controls and T1D patients. In the childhood onset T1D case, no intra-islet beta-cells were detected while in the adult onset case, beta-cell containing islets were found, exclusively in the head region. In the latter case, considerable numbers of small cellular clusters negative for three major endocrine hormones were observed, in islets with or without beta-cells. Ultrastructural analysis suggests these cells correspond to degenerating beta-cells, with empty granular membranes and abnormal morphology of nuclei with intranuclear pseudo-inclusions, adjacent to healthy alpha- and delta-cells. These results support a hypothesis that during T1D development in childhood, beta-cells are more susceptible to autoimmune destruction or immune attack is more severe, while beta-cell death in the adult onset T1D may be more protracted and incomplete. In addition, T1D may be associated with the formation of "empty" beta-cells, an interesting population of cells that may represent a key facet to the disorder's pathogenesis.
Collapse
Affiliation(s)
- Ananta Poudel
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Omid Savari
- Department of Surgery, The University of Chicago, Chicago, Illinois
| | - Deborah A. Striegel
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Vipul Periwal
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jerome Taxy
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, Illinois
| | | | - Piotr Witkowski
- Department of Surgery, The University of Chicago, Chicago, Illinois
| | - Mark A. Atkinson
- Department of Pathology, The University of Florida, Gainesville, Florida
| | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|
1543
|
Kaddis JS, Pugliese A, Atkinson MA. A run on the biobank: what have we learned about type 1 diabetes from the nPOD tissue repository? Curr Opin Endocrinol Diabetes Obes 2015; 22:290-5. [PMID: 26087339 DOI: 10.1097/med.0000000000000171] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Since the inaugural year of its biobank in 2007, the Network for Pancreatic Organ Donors with Diabetes program has provided 70 370 human samples to 127 investigators worldwide for projects focused on the pathogenesis of type 1 diabetes (T1D). The purpose of this review was to highlight major advances in our understanding of T1D using works that contain original data from experiments utilizing biospecimens provided by the Network for Pancreatic Organ Donors with Diabetes program. A total of 15 studies, published between 1 June 2013 and 31 December 2014, were selected using various search and filter strategies. RECENT FINDINGS The type and frequency of B and/or T-cell immune markers in both the endocrine and exocrine compartments vary in T1D. Enterovirus signals have been identified as having new proteins in the extracellular matrix around infiltrated islets. Novel genes within human islet cell types have been shown to play a role in immunity, infiltration, inflammation, disease progression, cell mass and function. Various cytokines and a complement degradation product have also been detected in the blood or surrounding pancreatic ducts/vasculature. SUMMARY These findings, from T1D donors across the disease spectrum, emphasize the notion that pathogenic heterogeneity is a hallmark of the disorder.
Collapse
Affiliation(s)
- John S Kaddis
- aDepartment of Information Sciences, City of Hope, Duarte, California bDiabetes Research Institute and Departments of Medicine, Microbiology and Immunology, University of Miami Miller School of Medicine, Miami cDepartments of Pathology and Pediatrics, University of Florida, Gainesville, Florida, USA
| | | | | |
Collapse
|
1544
|
Campbell-Thompson M. Organ donor specimens: What can they tell us about type 1 diabetes? Pediatr Diabetes 2015; 16:320-30. [PMID: 25998576 PMCID: PMC4718555 DOI: 10.1111/pedi.12286] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/04/2015] [Accepted: 05/01/2015] [Indexed: 12/16/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic disease resulting from the destruction of pancreatic beta cells, due to a poorly understood combination of genetic, environmental, and immune factors. The JDRF Network for Pancreatic Organ donors with Diabetes (nPOD) program recovers transplantation quality pancreas from organ donors throughout the USA. In addition to recovery of donors with T1D, non-diabetic donors include those with islet autoantibodies. Donors with type 2 diabetes and other conditions are also recovered to aid investigations directed at the full spectrum of pathophysiological mechanisms affecting beta cells. One central processing laboratory conducts standardized procedures for sample processing, storage, and distribution, intended for current and future cutting edge investigations. Baseline histology characterizations are performed on the pancreatic samples, with images of the staining results provided though whole-slide digital scans. Uniquely, these high-grade biospecimens are provided without expense to investigators, working worldwide, seeking methods for disease prevention and reversal strategies. Collaborative working groups are highly encouraged, bringing together multiple investigators with different expertise to foster collaborations in several areas of critical need. This mini-review will provide some key histopathological findings emanating from the nPOD collection, including the heterogeneity of beta cell loss and islet inflammation (insulitis), beta cell mass, insulin-producing beta cells in chronic T1D, and pancreas weight reductions at disease onset. Analysis of variations in histopathology observed from these organ donors could provide for mechanistic differences related to etiological agents and serve an important function in terms of identifying the heterogeneity of T1D.
Collapse
Affiliation(s)
- Martha Campbell-Thompson
- The Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida, College of Medicine, Gainesville, FL, USA
| |
Collapse
|
1545
|
Awasthi H, Nath R, Usman K, Mani D, Khattri S, Nischal A, Singh M, Sawlani KK. Effects of a standardized Ayurvedic formulation on diabetes control in newly diagnosed Type-2 diabetics; a randomized active controlled clinical study. Complement Ther Med 2015; 23:555-61. [DOI: 10.1016/j.ctim.2015.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 06/04/2015] [Accepted: 06/04/2015] [Indexed: 12/16/2022] Open
|
1546
|
Interleukin-35 administration counteracts established murine type 1 diabetes--possible involvement of regulatory T cells. Sci Rep 2015. [PMID: 26224624 PMCID: PMC4519737 DOI: 10.1038/srep12633] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The anti-inflammatory cytokine IL-35 is produced by regulatory T (Treg) cells to suppress autoimmune and inflammatory responses. The role of IL-35 in type 1 diabetes (T1D) remains to be answered. To elucidate this, we investigated the kinetics of Treg cell response in the multiple low dose streptozotocin induced (MLDSTZ) T1D model and measured the levels of IL-35 in human T1D patients. We found that Treg cells were increased in MLDSTZ mice. However, the Treg cells showed a decreased production of anti-inflammatory (IL-10, IL-35, TGF-β) and increased pro-inflammatory (IFN-γ, IL-2, IL-17) cytokines, indicating a phenotypic shift of Treg cells under T1D condition. IL-35 administration effectively both prevented development of, and counteracted established MLDSTZ T1D, seemingly by induction of Eos expression and IL-35 production in Treg cells, thus reversing the phenotypic shift of the Treg cells. IL-35 administration reversed established hyperglycemia in NOD mouse model of T1D. Moreover, circulating IL-35 levels were decreased in human T1D patients compared to healthy controls. These findings suggest that insufficient IL-35 levels play a pivotal role in the development of T1D and that treatment with IL-35 should be investigated in treatment of T1D and other autoimmune diseases.
Collapse
|
1547
|
Krogvold L, Skog O, Sundström G, Edwin B, Buanes T, Hanssen KF, Ludvigsson J, Grabherr M, Korsgren O, Dahl-Jørgensen K. Function of Isolated Pancreatic Islets From Patients at Onset of Type 1 Diabetes: Insulin Secretion Can Be Restored After Some Days in a Nondiabetogenic Environment In Vitro: Results From the DiViD Study. Diabetes 2015; 64:2506-2512. [PMID: 25677915 DOI: 10.2337/db14-1911] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/06/2015] [Indexed: 02/05/2023]
Abstract
The understanding of the etiology of type 1 diabetes (T1D) remains limited. One objective of the Diabetes Virus Detection (DiViD) study was to collect pancreatic tissue from living subjects shortly after the diagnosis of T1D. Here we report the insulin secretion ability by in vitro glucose perifusion and explore the expression of insulin pathway genes in isolated islets of Langerhans from these patients. Whole-genome RNA sequencing was performed on islets from six DiViD study patients and two organ donors who died at the onset of T1D, and the findings were compared with those from three nondiabetic organ donors. All human transcripts involved in the insulin pathway were present in the islets at the onset of T1D. Glucose-induced insulin secretion was present in some patients at the onset of T1D, and a perfectly normalized biphasic insulin release was obtained after some days in a nondiabetogenic environment in vitro. This indicates that the potential for endogenous insulin production is good, which could be taken advantage of if the disease process was reversed at diagnosis.
Collapse
Affiliation(s)
- Lars Krogvold
- Paediatric Department, Oslo University Hospital and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Görel Sundström
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Bjørn Edwin
- Intervention Centre and Department of Surgery, Oslo University Hospital and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trond Buanes
- Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kristian F Hanssen
- Department of Endocrinology, Oslo University Hospital and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Johnny Ludvigsson
- Division of Paediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Manfred Grabherr
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Knut Dahl-Jørgensen
- Paediatric Department, Oslo University Hospital and Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
1548
|
Abstract
No treatment to halt the progressive loss of insulin-producing beta-cells in type 1 diabetes mellitus has yet been clinically introduced. Strategies tested have at best only transiently preserved beta-cell function and in many cases with obvious side effects of drugs used. Several studies have suggested that mesenchymal stromal cells exert strong immunomodulatory properties with the capability to prevent or halt diabetes development in animal models of type 1 diabetes. A multitude of mechanisms has been forwarded to exert this effect. Recently, we translated this strategy into a first clinical phase I/IIa trial and observed no side effects, and preserved or even increased C-peptide responses to a mixed meal tolerance test during the first year after treatment. Future blinded, larger studies, with extended follow-up, are clearly of interest to investigate this treatment concept.
Collapse
Affiliation(s)
- Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Box 571, 75123, Uppsala, Sweden,
| | | | | |
Collapse
|
1549
|
Davis-Richardson AG, Triplett EW. A model for the role of gut bacteria in the development of autoimmunity for type 1 diabetes. Diabetologia 2015; 58:1386-93. [PMID: 25957231 PMCID: PMC4473028 DOI: 10.1007/s00125-015-3614-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/27/2015] [Indexed: 01/05/2023]
Abstract
Several lines of evidence suggest a role for the gut microbiome in type 1 diabetes. Treating diabetes-prone rodents with probiotics or antibiotics prevents the development of the disorder. Diabetes-prone rodents also have a distinctly different gut microbiome compared with healthy rodents. Recent studies in children with a high genetic risk for type 1 diabetes demonstrate significant differences in the gut microbiome between children who develop autoimmunity for the disease and those who remain healthy. However, the differences in microbiome composition between autoimmune and healthy children are not consistent across all studies because of the strong environmental influences on microbiome composition, particularly diet and geography. Controlling confounding factors of microbiome composition uncovers bacterial associations with disease. For example, in a human cohort from a single Finnish city where geography is confined, a strong association between one dominant bacterial species, Bacteroides dorei, and type 1 diabetes was discovered (Davis-Richardson et al. Front Microbiol 2014;5:678). Beyond this, recent DNA methylation analyses suggest that a thorough epigenetic analysis of the gut microbiome may be warranted. These studies suggest a testable model whereby a diet high in fat and gluten and low in resistant starch may be the primary driver of gut dysbiosis. This dysbiosis may cause a lack of butyrate production by gut bacteria, which, in turn, leads to the development of a permeable gut followed by autoimmunity. The bacterial community responsible for these changes in butyrate production may vary around the world, but bacteria of the genus Bacteroides are thought to play a key role.
Collapse
Affiliation(s)
- Austin G. Davis-Richardson
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, 1355 Museum Road, PO Box 110700, Gainesville, FL 32611-0700 USA
| | - Eric W. Triplett
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, 1355 Museum Road, PO Box 110700, Gainesville, FL 32611-0700 USA
| |
Collapse
|
1550
|
Bellin MD, Moran A, Wilhelm JJ, O'Brien TD, Gottlieb PA, Yu L, Dunn TB. Development of Autoimmune-Mediated β Cell Failure After Total Pancreatectomy With Autologous Islet Transplantation. Am J Transplant 2015; 15:1991-4. [PMID: 25765064 DOI: 10.1111/ajt.13216] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/21/2014] [Accepted: 01/05/2015] [Indexed: 01/25/2023]
Abstract
Total pancreatectomy with islet autotransplantation (TPIAT) is performed for definitive treatment of chronic pancreatitis; patients are not diabetic before surgery, or have C-peptide positive pancreatogenous diabetes. Thus, TPIAT recipients are not traditionally considered at risk for autoimmune loss of the islet graft. We describe a 43-year-old female who underwent TPIAT with high mass islet graft of 6031 IEQ/kg, with no evidence of presurgical β cell autoimmunity who developed type 1 diabetes within the first year after TPIAT, resulting in complete loss of beta cell function. The patient had positive GAD and insulin autoantibodies at 1 year and 18 months after TPIAT, not present prior, and undetectable C-peptide after mixed meal and intravenous glucose tolerance testing at 18 months. Glucagon secretion was preserved, suggesting the transplanted alpha cell mass was intact. HLA typing revealed a DR3/DR4 class II haplotype. This case highlights the need to consider de novo type 1 diabetes in patients with unexpected islet graft failure after TPIAT.
Collapse
Affiliation(s)
- M D Bellin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN.,University of Minnesota, Schulze Diabetes Institute, Minneapolis, MN
| | - A Moran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - J J Wilhelm
- University of Minnesota, Schulze Diabetes Institute, Minneapolis, MN
| | - T D O'Brien
- University of Minnesota, College of Veterinary Medicine, St. Paul, MN
| | - P A Gottlieb
- Barbara Davis Center for Childhood Diabetes, Aurora, CO
| | - L Yu
- Barbara Davis Center for Childhood Diabetes, Aurora, CO
| | - T B Dunn
- Department of Surgery, University of Minnesota, Minneapolis, MN
| |
Collapse
|