1
|
Aziz N, Dash B, Wal P, Kumari P, Joshi P, Wal A. New Horizons in Diabetic Neuropathies: An Updated Review on their Pathology, Diagnosis, Mechanism, Screening Techniques, Pharmacological, and Future Approaches. Curr Diabetes Rev 2024; 20:e201023222416. [PMID: 37867268 DOI: 10.2174/0115733998242299231011181615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/16/2023] [Accepted: 08/25/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND One of the largest problems for global public health is diabetes mellitus (DM) and its micro and macrovascular consequences. Although prevention, diagnosis, and treatment have generally improved, its incidence is predicted to keep rising over the coming years. Due to the intricacy of the molecular mechanisms, which include inflammation, oxidative stress, and angiogenesis, among others, discovering treatments to stop or slow the course of diabetic complications is still a current unmet need. METHODS The pathogenesis and development of diabetic neuropathies may be explained by a wide variety of molecular pathways, hexosamine pathways, such as MAPK pathway, PARP pathway, oxidative stress pathway polyol (sorbitol) pathway, cyclooxygenase pathway, and lipoxygenase pathway. Although diabetic neuropathies can be treated symptomatically, there are limited options for treating the underlying cause. RESULT Various pathways and screening models involved in diabetic neuropathies are discussed, along with their possible outcomes. Moreover, both medicinal and non-medical approaches to therapy are also explored. CONCLUSION This study highlights the probable involvement of several processes and pathways in the establishment of diabetic neuropathies and presents in-depth knowledge of new therapeutic approaches intended to stop, delay, or reverse different types of diabetic complications.
Collapse
Affiliation(s)
- Namra Aziz
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur 209305, UP, India
| | - Biswajit Dash
- Department of Pharmaceutical Technology, School of Medical Sciences, ADAMAS University, Kolkata 700 126, West Bengal, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur 209305, UP, India
| | - Prachi Kumari
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur 209305, UP, India
| | - Poonam Joshi
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, Uttarakhand, India
| | - Ankita Wal
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur 209305, UP, India
| |
Collapse
|
2
|
Yagihashi S. Contribution of animal models to diabetes research: Its history, significance, and translation to humans. J Diabetes Investig 2023; 14:1015-1037. [PMID: 37401013 PMCID: PMC10445217 DOI: 10.1111/jdi.14034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 07/05/2023] Open
Abstract
Diabetes mellitus is still expanding globally and is epidemic in developing countries. The combat of this plague has caused enormous economic and social burdens related to a lowered quality of life in people with diabetes. Despite recent significant improvements of life expectancy in patients with diabetes, there is still a need for efforts to elucidate the complexities and mechanisms of the disease processes to overcome this difficult disorder. To this end, the use of appropriate animal models in diabetes studies is invaluable for translation to humans and for the development of effective treatment. In this review, a variety of animal models of diabetes with spontaneous onset in particular will be introduced and discussed for their implication in diabetes research.
Collapse
Affiliation(s)
- Soroku Yagihashi
- Department of Exploratory Medicine for Nature, Life and HumansToho University School of MedicineChibaJapan
- Department of PathologyHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
3
|
Halliez C, Ibrahim H, Otonkoski T, Mallone R. In vitro beta-cell killing models using immune cells and human pluripotent stem cell-derived islets: Challenges and opportunities. Front Endocrinol (Lausanne) 2023; 13:1076683. [PMID: 36726462 PMCID: PMC9885197 DOI: 10.3389/fendo.2022.1076683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Type 1 diabetes (T1D) is a disease of both autoimmunity and β-cells. The β-cells play an active role in their own demise by mounting defense mechanisms that are insufficient at best, and that can become even deleterious in the long term. This complex crosstalk is important to understanding the physiological defense mechanisms at play in healthy conditions, their alterations in the T1D setting, and therapeutic agents that may boost such mechanisms. Robust protocols to develop stem-cell-derived islets (SC-islets) from human pluripotent stem cells (hPSCs), and islet-reactive cytotoxic CD8+ T-cells from peripheral blood mononuclear cells offer unprecedented opportunities to study this crosstalk. Challenges to develop in vitro β-cell killing models include the cluster morphology of SC-islets, the relatively weak cytotoxicity of most autoimmune T-cells and the variable behavior of in vitro expanded CD8+ T-cells. These challenges may however be highly rewarding in light of the opportunities offered by such models. Herein, we discuss these opportunities including: the β-cell/immune crosstalk in an islet microenvironment; the features that make β-cells more sensitive to autoimmunity; therapeutic agents that may modulate β-cell vulnerability; and the possibility to perform analyses in an autologous setting, i.e., by generating T-cell effectors and SC-islets from the same donor.
Collapse
Affiliation(s)
- Clémentine Halliez
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
- Department of Pediatrics, Helsinki University Hospital, Helsinki, Finland
| | - Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| |
Collapse
|
4
|
Pathak R, Sachan N, Chandra P. Mechanistic approach towards diabetic neuropathy screening techniques and future challenges: A review. Biomed Pharmacother 2022; 150:113025. [PMID: 35658222 DOI: 10.1016/j.biopha.2022.113025] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
Diabetic neuropathy, also called peripheral diabetic neuropathy (PDN), is among the most significant diabetes health consequences, alongside diabetic nephropathy, diabetic cardiomyopathy and diabetic retinopathy. Diabetic neuropathy is the existence of signs and indications of peripheral nerve damage in patients with diabetes after other causes have been governed out. Diabetic neuropathy is a painful and severe complication of diabetes that affects roughly 20% of people. The development of diabetic neuropathy is regulated by blood arteries that nourish the peripheral nerves and metabolic problems such as increased stimulation of polyol pathway, loss of myo-inositol and enhanced non-enzymatic glycation. It's divided into four types based on where neurons are most affected: autonomic, peripheral, proximal, and focal, with each kind presenting different symptoms like numbing, gastrointestinal disorders, and heart concerns. Pharmacotherapy for neuropathic pain is complex and for many patients, effective treatment is lacking; as a result, scientific proof recommendations are crucial. As a result, the current demand is to give the most vital medications or combinations of drugs that work directly on the nerves to help diabetic neuropathy patients feel less pain without causing any adverse effects. In diabetic neuropathy research, animal models are ubiquitous, with rats and mice being the most typically chosen for various reasons. This review covers the epidemiology, clinical features, pathology, clinical symptom, mechanism of diabetic neuropathy development, diagnosis, screening models of animals, diabetic neuropathy pharmacotherapy.
Collapse
Affiliation(s)
- Rashmi Pathak
- School of Pharmaceutical Sciences, IFTM University, Lodhipur Rajput Delhi Road (NH-24), Moradabad, UP 244102, India
| | - Neetu Sachan
- School of Pharmaceutical Sciences, IFTM University, Lodhipur Rajput Delhi Road (NH-24), Moradabad, UP 244102, India
| | - Phool Chandra
- School of Pharmaceutical Sciences, IFTM University, Lodhipur Rajput Delhi Road (NH-24), Moradabad, UP 244102, India.
| |
Collapse
|
5
|
Bouali-Benazzouz R, Landry M, Benazzouz A, Fossat P. Neuropathic pain modeling: Focus on synaptic and ion channel mechanisms. Prog Neurobiol 2021; 201:102030. [PMID: 33711402 DOI: 10.1016/j.pneurobio.2021.102030] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022]
Abstract
Animal models of pain consist of modeling a pain-like state and measuring the consequent behavior. The first animal models of neuropathic pain (NP) were developed in rodents with a total lesion of the sciatic nerve. Later, other models targeting central or peripheral branches of nerves were developed to identify novel mechanisms that contribute to persistent pain conditions in NP. Objective assessment of pain in these different animal models represents a significant challenge for pre-clinical research. Multiple behavioral approaches are used to investigate and to validate pain phenotypes including withdrawal reflex to evoked stimuli, vocalizations, spontaneous pain, but also emotional and affective behaviors. Furthermore, animal models were very useful in investigating the mechanisms of NP. This review will focus on a detailed description of rodent models of NP and provide an overview of the assessment of the sensory and emotional components of pain. A detailed inventory will be made to examine spinal mechanisms involved in NP-induced hyperexcitability and underlying the current pharmacological approaches used in clinics with the possibility to present new avenues for future treatment. The success of pre-clinical studies in this area of research depends on the choice of the relevant model and the appropriate test based on the objectives of the study.
Collapse
Affiliation(s)
- Rabia Bouali-Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.
| | - Marc Landry
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Abdelhamid Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Pascal Fossat
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
6
|
Pandey S, Dvorakova MC. Future Perspective of Diabetic Animal Models. Endocr Metab Immune Disord Drug Targets 2020; 20:25-38. [PMID: 31241444 PMCID: PMC7360914 DOI: 10.2174/1871530319666190626143832] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/06/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
Abstract
Objective The need of today’s research is to develop successful and reliable diabetic animal models for understanding the disease susceptibility and pathogenesis. Enormous success of animal models had already been acclaimed for identifying key genetic and environmental factors like Idd loci and effects of microorganisms including the gut microbiota. Furthermore, animal models had also helped in identifying many therapeutic targets and strategies for immune-intervention. In spite of a quite success, we have acknowledged that many of the discovered immunotherapies are working on animals and did not have a significant impact on human. Number of animal models were developed in the past to accelerate drug discovery pipeline. However, due to poor initial screening and assessment on inequivalent animal models, the percentage of drug candidates who succeeded during clinical trials was very low. Therefore, it is essential to bridge this gap between pre-clinical research and clinical trial by validating the existing animal models for consistency. Results and Conclusion In this review, we have discussed and evaluated the significance of animal models on behalf of published data on PUBMED. Amongst the most popular diabetic animal models, we have selected six animal models (e.g. BioBreeding rat, “LEW IDDM rat”, “Nonobese Diabetic (NOD) mouse”, “STZ RAT”, “LEPR Mouse” and “Zucker Diabetic Fatty (ZDF) rat” and ranked them as per their published literature on PUBMED. Moreover, the vision and brief imagination for developing an advanced and robust diabetic model of 21st century was discussed with the theme of one mice-one human concept including organs-on-chips.
Collapse
Affiliation(s)
- Shashank Pandey
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Magdalena C Dvorakova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.,Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
7
|
Abboud C, Duveau A, Bouali-Benazzouz R, Massé K, Mattar J, Brochoire L, Fossat P, Boué-Grabot E, Hleihel W, Landry M. Animal models of pain: Diversity and benefits. J Neurosci Methods 2020; 348:108997. [PMID: 33188801 DOI: 10.1016/j.jneumeth.2020.108997] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Chronic pain is a maladaptive neurological disease that remains a major health problem. A deepening of our knowledge on mechanisms that cause pain is a prerequisite to developing novel treatments. A large variety of animal models of pain has been developed that recapitulate the diverse symptoms of different pain pathologies. These models reproduce different pain phenotypes and remain necessary to examine the multidimensional aspects of pain and understand the cellular and molecular basis underlying pain conditions. In this review, we propose an overview of animal models, from simple organisms to rodents and non-human primates and the specific traits of pain pathologies they model. We present the main behavioral tests for assessing pain and investing the underpinning mechanisms of chronic pathological pain. The validity of animal models is analysed based on their ability to mimic human clinical diseases and to predict treatment outcomes. Refine characterization of pathological phenotypes also requires to consider pain globally using specific procedures dedicated to study emotional comorbidities of pain. We discuss the limitations of pain models when research findings fail to be translated from animal models to human clinics. But we also point to some recent successes in analgesic drug development that highlight strategies for improving the predictive validity of animal models of pain. Finally, we emphasize the importance of using assortments of preclinical pain models to identify pain subtype mechanisms, and to foster the development of better analgesics.
Collapse
Affiliation(s)
- Cynthia Abboud
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France; Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Alexia Duveau
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Rabia Bouali-Benazzouz
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Karine Massé
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Joseph Mattar
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Louison Brochoire
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Pascal Fossat
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Eric Boué-Grabot
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Walid Hleihel
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Marc Landry
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France.
| |
Collapse
|
8
|
Noshahr ZS, Salmani H, Khajavi Rad A, Sahebkar A. Animal Models of Diabetes-Associated Renal Injury. J Diabetes Res 2020; 2020:9416419. [PMID: 32566684 PMCID: PMC7256713 DOI: 10.1155/2020/9416419] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/28/2020] [Indexed: 12/29/2022] Open
Abstract
Diabetic nephropathy (DN) is the main factor leading to end-stage renal disease (ESRD) and subsequent morbidity and mortality. Importantly, the prevalence of DN is continuously increasing in developed countries. Many rodent models of type 1 and type 2 diabetes have been established to elucidate the pathogenesis of diabetes and examine novel therapies against DN. These models are developed by chemical, surgical, genetic, drug, and diet/nutrition interventions or combination of two or more methods. The main characteristics of DN including a decrease in renal function, albuminuria and mesangiolysis, mesangial expansion, and nodular glomerulosclerosis should be exhibited by an animal model of DN. However, a rodent model possessing all of the abovementioned features of human DN has not yet been developed. Furthermore, mice of different genetic backgrounds and strains show different levels of susceptibility to DN with respect to albuminuria and development of glomerular and tubulointerstitial lesions. Therefore, the type of diabetes, development of nephropathy, duration of the study, cost of maintaining and breeding, and animals' mortality rate are important factors that might be affected by the type of DN model. In this review, we discuss the pros and cons of different rodent models of diabetes that are being used to study DN.
Collapse
Affiliation(s)
- Zahra Samadi Noshahr
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Salmani
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Khajavi Rad
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Argaev Frenkel L, Rozenfeld H, Rozenberg K, Sampson SR, Rosenzweig T. N-Acetyl-l-Cysteine Supplement in Early Life or Adulthood Reduces Progression of Diabetes in Nonobese Diabetic Mice. Curr Dev Nutr 2019; 3:nzy097. [PMID: 30993256 PMCID: PMC6459986 DOI: 10.1093/cdn/nzy097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Oxidative stress contributes to the pathologic process leading to the development, progression, and complications of type 1 diabetes (T1D). OBJECTIVE The aim of this study was to investigate the effect of the antioxidant N-acetyl-l-cysteine (NAC), supplemented during early life or adulthood on the development of T1D. METHODS NAC was administered to nonobese diabetic (NOD) female mice during pregnancy and lactation, and the development of diabetes was followed in offspring. In an additional set of experiments, offspring of untreated mice were given NAC during adulthood, and the development of T1D was followed. Morbidity rate, insulitis and serum cytokines were measured in the 2 sets of experiments. In addition, markers of oxidative stress, glutathione, lipid peroxidation, total antioxidant capacity and activity of antioxidant enzymes, were followed. RESULTS Morbidity rate was reduced in both treatment protocols. A decrease in interferon γ, tumor necrosis factor α, interleukin 1α, and other type 1 diabetes-associated proinflammatory cytokines was found in mice supplemented with NAC in adulthood or during early life compared with control NOD mice. The severity of insulitis was higher in control NOD mice than in treated groups. NAC administration significantly reduced oxidative stress, as determined by reduced lipid peroxidation and increased total antioxidant capacity in serum and pancreas of mice treated in early life or in adulthood and increased pancreatic glutathione when administrated in adulthood. The activity of antioxidant enzymes was not affected in mice given NAC in adulthood, whereas an increase in the activity of superoxide dismutase and catalase was demonstrated in the pancreas of their offspring. CONCLUSION NAC decreased morbidity of NOD mice by attenuating the immune response, presumably by eliminating oxidative stress, and might be beneficial in reducing morbidity rates of T1D in high-risk individuals.
Collapse
Affiliation(s)
- Lital Argaev Frenkel
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| | - Hava Rozenfeld
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| | - Konstantin Rozenberg
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| | - Sanford R Sampson
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tovit Rosenzweig
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
10
|
Diallo K, Oppong AK, Lim GE. Can 14-3-3 proteins serve as therapeutic targets for the treatment of metabolic diseases? Pharmacol Res 2019; 139:199-206. [DOI: 10.1016/j.phrs.2018.11.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022]
|
11
|
Priyadarsini S, Rowsey TG, Ma JX, Karamichos D. Unravelling the stromal-nerve interactions in the human diabetic cornea. Exp Eye Res 2017; 164:22-30. [PMID: 28827027 DOI: 10.1016/j.exer.2017.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/14/2017] [Accepted: 08/02/2017] [Indexed: 01/18/2023]
Abstract
Corneal defects due to diabetes mellitus (DM) may cause severe vision impairments. Current studies focus on the corneal epithelium and nerve defects neglecting the corneal stroma. The aim of this study was to develop a 3D in vitro model to examine the interactions between corneal stroma and nerves in the context of DM. Primary human corneal stromal fibroblasts isolated from healthy (HCFs), Type 1 (T1DM) and Type 2 (T2DM) patients were stimulated with stable ascorbic acid to secrete and assemble an extracellular matrix (ECM). Human neuronal cells were then seeded on top and differentiated to create the 3D co-cultures. Our data revealed successful co-culture of stromal fibroblasts and neuronal cells with large elongated neuron extensions. T2DM showed significant upregulation of Collagen III and IGF1 when compared to T1DM. Interestingly, upon nerve addition, those markers returned to HCF levels. Neuronal markers were also differentially modulated with T2DM co-cultures expressing high levels of βIII tubulin where T1DM co-cultures expressed Substance P. . Overall, our unique 3D co-culture model provides us with a tool that can be utilized for both molecular and therapeutic studies for diabetic keratopathy.
Collapse
Affiliation(s)
- Shrestha Priyadarsini
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Tyler G Rowsey
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jian-Xing Ma
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dimitrios Karamichos
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
12
|
Lee S, Elaskandrany M, Ahad A, Chaqour B. Analysis of CCN Protein Expression and Activities in Vasoproliferative Retinopathies. Methods Mol Biol 2017; 1489:543-556. [PMID: 27734405 DOI: 10.1007/978-1-4939-6430-7_46] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The retina is a complex neurovascular structure that conveys light/visual image through the optic nerve to the visual cortex of the brain. Neuronal and vascular activities in the retina are physically and functionally intertwined, and vascular alterations are consequential to the proper function of the entire visual system. In particular, alteration of the structure and barrier function of the retinal vasculature is commonly associated with the development of vasoproliferative ischemic retinopathy, a set of clinically well-defined chronic ocular microvascular complications causing blindness in all age groups. Experimentally, the retinal tissue provides researchers with a convenient, easily accessible, and directly observable model suitable to investigate whether and how newly identified genes regulate vascular development and regeneration. The six mammalian CCN gene-encoded proteins are part of an extracellular network of bioactive molecules that regulate various aspects of organ system development and diseases. Whether and how these molecules regulate the fundamental aspects of blood vessel development and pathology and subsequently the neurovascular link in the retina are open-ended questions. Sophisticated methods have been developed to gain insight into the pathogenesis of retinal vasculopathy. This chapter describes several useful methodologies and animal models to investigate the regulation and potential relevance of the CCN proteins in vasoproliferative diseases of the retina.
Collapse
Affiliation(s)
- Sangmi Lee
- Department of Cell Biology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, Brooklyn, NY, USA
- Department of Ophthalmology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Menna Elaskandrany
- Department of Cell Biology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, Brooklyn, NY, USA
- Department of Ophthalmology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Afruja Ahad
- Department of Cell Biology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, Brooklyn, NY, USA
- Department of Ophthalmology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Brahim Chaqour
- Department of Cell Biology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, Brooklyn, NY, USA.
- Department of Ophthalmology, SUNY Downstate Medical Center, College of Medicine and SUNY Eye Institute, State University of New York, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
13
|
Priyadarsini S, Sarker-Nag A, Rowsey TG, Ma JX, Karamichos D. Establishment of a 3D In Vitro Model to Accelerate the Development of Human Therapies against Corneal Diabetes. PLoS One 2016; 11:e0168845. [PMID: 28005998 PMCID: PMC5179241 DOI: 10.1371/journal.pone.0168845] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/07/2016] [Indexed: 11/18/2022] Open
Abstract
PURPOSE To establish an in vitro model that would mirror the in vivo corneal stromal environment in diabetes (DM) patients. METHODS Human corneal fibroblasts from Healthy (HCFs), Type 1DM (T1DM) and Type 2DM (T2DM) donors were isolated and cultured for 4 weeks with Vitamin C stimulation in order to allow for extracellular matrix (ECM) secretion and assembly. RESULTS Our data indicated altered cellular morphology, increased cellular migration, increased ECM assembly, and severe mitochondrial damage in both T1DM and T2DMs when compared to HCFs. Furthermore, we found significant downregulation of Collagen I and Collagen V expression in both T1DM and T2DMs. Furthermore, a significant up regulation of fibrotic markers was seen, including α-smooth muscle actin in T2DM and Collagen III in both T1DM and T2DMs. Metabolic analysis suggested impaired Glycolysis and Tricarboxylic acid cycle (TCA) pathway. CONCLUSION DM has significant effects on physiological and clinical aspects of the human cornea. The benefits in developing and fully characterizing our 3D in vitro model are enormous and might provide clues for the development of novel therapeutics.
Collapse
Affiliation(s)
- Shrestha Priyadarsini
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Akhee Sarker-Nag
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Tyler G. Rowsey
- Department of Biology and Chemistry, East Central University, Ada, Oklahoma, United States of America
| | - Jian-Xing Ma
- Department of Physiology Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Dimitrios Karamichos
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
14
|
Trans-presentation of interleukin-15 by interleukin-15 receptor alpha is dispensable for the pathogenesis of autoimmune type 1 diabetes. Cell Mol Immunol 2016; 14:590-596. [PMID: 26853723 DOI: 10.1038/cmi.2015.102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/15/2015] [Accepted: 11/15/2015] [Indexed: 02/01/2023] Open
Abstract
Interleukin-15 (IL-15) is a pro-inflammatory cytokine that is required for the survival and activation of memory CD8+T cells, natural killer (NK) cells, innate lymphoid cells, macrophages and dendritic cells. IL-15 is implicated in the pathogenesis of various autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease, psoriasis and autoimmune type 1 diabetes (T1D). IL-15 receptor (IL-15R) consists of a specific α chain, the β chain that is shared with IL-2R and the common γ chain. IL-15 is unique in the manner in which it binds and signals through its receptor subunits. IL-15 that is complexed with IL-15Rα binds to the βγ receptor complex present on the responding cell to mediate its biological effects through a process referred to as trans-presentation. The trans-presented IL-15 is essential to mediate the biological effects on T lymphocytes and NK cells. Here we show that IL-15, but not IL-15Rα, is required for the development of spontaneous and virus-induced T1D, viral clearance and for antigen cross-presentation to CD8+ T lymphocytes. Our findings provide insight into the complexities of IL-15 signalling in the initiation and maintenance of CD8+ T cell-mediated immune responses.
Collapse
|
15
|
Al-awar A, Kupai K, Veszelka M, Szűcs G, Attieh Z, Murlasits Z, Török S, Pósa A, Varga C. Experimental Diabetes Mellitus in Different Animal Models. J Diabetes Res 2016; 2016:9051426. [PMID: 27595114 PMCID: PMC4993915 DOI: 10.1155/2016/9051426] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/16/2022] Open
Abstract
Animal models have historically played a critical role in the exploration and characterization of disease pathophysiology and target identification and in the evaluation of novel therapeutic agents and treatments in vivo. Diabetes mellitus disease, commonly known as diabetes, is a group of metabolic disorders characterized by high blood glucose levels for a prolonged time. To avoid late complications of diabetes and related costs, primary prevention and early treatment are therefore necessary. Due to its chronic symptoms, new treatment strategies need to be developed, because of the limited effectiveness of the current therapies. We overviewed the pathophysiological features of diabetes in relation to its complications in type 1 and type 2 mice along with rat models, including Zucker Diabetic Fatty (ZDF) rats, BB rats, LEW 1AR1/-iddm rats, Goto-Kakizaki rats, chemically induced diabetic models, and Nonobese Diabetic mouse, and Akita mice model. The advantages and disadvantages that these models comprise were also addressed in this review. This paper briefly reviews the wide pathophysiological and molecular mechanisms associated with type 1 and type 2 diabetes, particularly focusing on the challenges associated with the evaluation and predictive validation of these models as ideal animal models for preclinical assessments and discovering new drugs and therapeutic agents for translational application in humans.
Collapse
Affiliation(s)
- Amin Al-awar
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Kozep Fasor 52, 6726 Szeged, Hungary
| | - Krisztina Kupai
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Kozep Fasor 52, 6726 Szeged, Hungary
- *Krisztina Kupai:
| | - Médea Veszelka
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Kozep Fasor 52, 6726 Szeged, Hungary
| | - Gergő Szűcs
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Kozep Fasor 52, 6726 Szeged, Hungary
| | - Zouhair Attieh
- Department of Laboratory Science and Technology, Faculty of Health Sciences, American University of Science and Technology, Alfred Naccache Avenue, Beirut 1100, Lebanon
| | | | - Szilvia Török
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Kozep Fasor 52, 6726 Szeged, Hungary
| | - Anikó Pósa
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Kozep Fasor 52, 6726 Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Kozep Fasor 52, 6726 Szeged, Hungary
| |
Collapse
|
16
|
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: 50] [Impact Index Per Article: 5.6] [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
|
17
|
The Cnes2 locus on mouse chromosome 17 regulates host defense against cryptococcal infection through pleiotropic effects on host immunity. Infect Immun 2015; 83:4541-54. [PMID: 26371125 DOI: 10.1128/iai.00697-15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/01/2015] [Indexed: 02/06/2023] Open
Abstract
The genetic basis of natural susceptibility to progressive Cryptococcus neoformans infection is not well understood. Using C57BL/6 and CBA/J inbred mice, we previously identified three chromosomal regions associated with C. neoformans susceptibility (Cnes1, Cnes2, and Cnes3). To validate and characterize the role of Cnes2 during the host response, we constructed a congenic strain on the C57BL/6 background (B6.CBA-Cnes2). Phenotypic analysis of B6.CBA-Cnes2 mice 35 days after C. neoformans infection showed a significant reduction of fungal burden in the lungs and spleen with higher pulmonary expression of gamma interferon (IFN-γ) and interleukin-12 (IL-12), lower expression of IL-4, IL-5, and IL-13, and an absence of airway epithelial mucus production compared to that in C57BL/6 mice. Multiparameter flow cytometry of infected lungs also showed a significantly higher number of neutrophils, exudate macrophages, CD11b(+) dendritic cells, and CD4(+) cells in B6.CBA-Cnes2 than in C57BL/6 mice. The activation state of recruited macrophages and dendritic cells was also significantly increased in B6.CBA-Cnes2 mice. Taken together, these findings demonstrate that the Cnes2 interval is a potent regulator of host defense, immune responsiveness, and differential Th1/Th2 polarization following C. neoformans infection.
Collapse
|
18
|
Biessels GJ, Bril V, Calcutt NA, Cameron NE, Cotter MA, Dobrowsky R, Feldman EL, Fernyhough P, Jakobsen J, Malik RA, Mizisin AP, Oates PJ, Obrosova IG, Pop-Busui R, Russell JW, Sima AA, Stevens MJ, Schmidt RE, Tesfaye S, Veves A, Vinik AI, Wright DE, Yagihashi S, Yorek MA, Ziegler D, Zochodne DW. Phenotyping animal models of diabetic neuropathy: a consensus statement of the diabetic neuropathy study group of the EASD (Neurodiab). J Peripher Nerv Syst 2015; 19:77-87. [PMID: 24934510 DOI: 10.1111/jns5.12072] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/27/2014] [Accepted: 04/07/2014] [Indexed: 11/28/2022]
Abstract
NIDDK, JDRF, and the Diabetic Neuropathy Study Group of EASD sponsored a meeting to explore the current status of animal models of diabetic peripheral neuropathy. The goal of the workshop was to develop a set of consensus criteria for the phenotyping of rodent models of diabetic neuropathy. The discussion was divided into five areas: (1) status of commonly used rodent models of diabetes, (2) nerve structure, (3) electrophysiological assessments of nerve function, (4) behavioral assessments of nerve function, and (5) the role of biomarkers in disease phenotyping. Participants discussed the current understanding of each area, gold standards (if applicable) for assessments of function, improvements of existing techniques, and utility of known and exploratory biomarkers. The research opportunities in each area were outlined, providing a possible roadmap for future studies. The meeting concluded with a discussion on the merits and limitations of a unified approach to phenotyping rodent models of diabetic neuropathy and a consensus formed on the definition of the minimum criteria required for establishing the presence of the disease. A neuropathy phenotype in rodents was defined as the presence of statistically different values between diabetic and control animals in 2 of 3 assessments (nocifensive behavior, nerve conduction velocities, or nerve structure). The participants propose that this framework would allow different research groups to compare and share data, with an emphasis on data targeted toward the therapeutic efficacy of drug interventions.
Collapse
|
19
|
Yi SS. Effects of exercise on brain functions in diabetic animal models. World J Diabetes 2015; 6:583-597. [PMID: 25987956 PMCID: PMC4434079 DOI: 10.4239/wjd.v6.i4.583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/16/2015] [Accepted: 02/09/2015] [Indexed: 02/05/2023] Open
Abstract
Human life span has dramatically increased over several decades, and the quality of life has been considered to be equally important. However, diabetes mellitus (DM) characterized by problems related to insulin secretion and recognition has become a serious health problem in recent years that threatens human health by causing decline in brain functions and finally leading to neurodegenerative diseases. Exercise is recognized as an effective therapy for DM without medication administration. Exercise studies using experimental animals are a suitable option to overcome this drawback, and animal studies have improved continuously according to the needs of the experimenters. Since brain health is the most significant factor in human life, it is very important to assess brain functions according to the different exercise conditions using experimental animal models. Generally, there are two types of DM; insulin-dependent type 1 DM and an insulin-independent type 2 DM (T2DM); however, the author will mostly discuss brain functions in T2DM animal models in this review. Additionally, many physiopathologic alterations are caused in the brain by DM such as increased adiposity, inflammation, hormonal dysregulation, uncontrolled hyperphagia, insulin and leptin resistance, and dysregulation of neurotransmitters and declined neurogenesis in the hippocampus and we describe how exercise corrects these alterations in animal models. The results of changes in the brain environment differ according to voluntary, involuntary running exercises and resistance exercise, and gender in the animal studies. These factors have been mentioned in this review, and this review will be a good reference for studying how exercise can be used with therapy for treating DM.
Collapse
|
20
|
Kuczma M, Wang CY, Ignatowicz L, Gourdie R, Kraj P. Altered connexin 43 expression underlies age-dependent decrease of regulatory T cell suppressor function in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2015; 194:5261-71. [PMID: 25911751 DOI: 10.4049/jimmunol.1400887] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 03/24/2015] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes is one of the most extensively studied autoimmune diseases, but the cellular and molecular mechanisms leading to T cell-mediated destruction of insulin-producing β cells are still not well understood. In this study, we show that regulatory T cells (T(regs)) in NOD mice undergo age-dependent loss of suppressor functions exacerbated by the decreased ability of activated effector T cells to upregulate Foxp3 and generate T(regs) in the peripheral organs. This age-dependent loss is associated with reduced intercellular communication mediated by gap junctions, which is caused by impaired upregulation and decreased expression of connexin 43. Regulatory functions can be corrected, even in T cells isolated from aged, diabetic mice, by a synergistic activity of retinoic acid, TGF-β, and IL-2, which enhance connexin 43 and Foxp3 expression in T(regs) and restore the ability of conventional CD4(+) T cells to upregulate Foxp3 and generate peripherally derived T(regs). Moreover, we demonstrate that suppression mediated by T(regs) from diabetic mice is enhanced by a novel reagent, which facilitates gap junction aggregation. In summary, our report identifies gap junction-mediated intercellular communication as an important component of the T(reg) suppression mechanism compromised in NOD mice and suggests how T(reg) mediated immune regulation can be improved.
Collapse
Affiliation(s)
- Michal Kuczma
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912
| | - Cong-Yi Wang
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912; The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; and
| | - Leszek Ignatowicz
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912
| | - Robert Gourdie
- Virginia Tech Carilion Research Institute, Roanoke, VA 24015
| | - Piotr Kraj
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, GA 30912;
| |
Collapse
|
21
|
Racine JJ, Zhang M, Wang M, Morales W, Shen C, Zeng D. MHC-mismatched mixed chimerism mediates thymic deletion of cross-reactive autoreactive T cells and prevents insulitis in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2014; 194:407-17. [PMID: 25429069 DOI: 10.4049/jimmunol.1401584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Type 1 diabetic NOD mice have defects in both thymic negative selection and peripheral regulation of autoreactive T cells, and induction of mixed chimerism can effectively reverse these defects. Our recent studies suggest that MHC-mismatched mixed chimerism mediates negative selection of autoreactive thymocytes in wild-type NOD and TCR-transgenic NOD.Rag1(+/+).BDC2.5 mice. However, it remains unknown how mismatched I-A(b) MHC class II can mediate deletion of autoreactive T cells positively selected by I-A(g7). In the present study, we directly tested the hypothesis that mismatched MHC class II in mixed chimeras mediates deletion of cross-reactive autoreactive thymocytes. We first identify that transgenic BDC2.5 T cells from NOD.Rag1(+/+).BDC2.5 but not NOD.Rag1(-/-).BDC2.5 mice possess cross-reactive TCRs with endogenous TCRα-chains; MHC-mismatched H-2(b) but not matched H-2(g7) mixed chimerism mediates thymic deletion of the cross-reactive transgenic T cells in NOD.Rag1(+/+).BDC2.5 mice. Second, by transplanting T cell-depleted (TCD) bone marrow (BM) cells from NOD.Rag1(+/+).BDC2.5 or NOD.Rag1(-/-).BDC2.5 mice into lethally irradiated MHC-mismatched H-2(b) C57BL/6 or MHC-matched congenic B6.H-2(g7) recipients, we demonstrate that NOD.Rag1(+/+).BDC2.5 BM-derived cross-reactive transgenic T cells, but not NOD.Rag1(-/-).BDC2.5 BM-derived non-cross-reactive transgenic T cells, can be positively selected in MHC-mismatched H-2(b) thymus. Third, by cotransplanting NOD.Rag1(+/+).BDC2.5 TCD BM cells with BM cells from MHC-mismatched T cell-deficient C57BL/6 mice into lethally irradiated MHC-matched B6.H-2(g7) recipients, we demonstrate that thymic deletion of the cross-reactive transgenic T cells is dependent on MHC-mismatched donor BM-derived APCs but not on donor BM-derived T cells. Taken together, our studies indicate that MHC-mismatched mixed chimerism can mediate thymic deletion of cross-reactive autoreactive T cells that express more than one TCR.
Collapse
Affiliation(s)
- Jeremy J Racine
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010; Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - Mingfeng Zhang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - Miao Wang
- Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| | - William Morales
- Eugene and Ruth Roberts Summer Student Academy, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Christine Shen
- Eugene and Ruth Roberts Summer Student Academy, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | - Defu Zeng
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010; Department of Diabetes Research, Beckman Research Institute, City of Hope, Duarte, CA 91010; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010; and
| |
Collapse
|
22
|
Abstract
Inflammation-induced inhibition of the insulin signalling pathway can lead to insulin resistance and contribute to the development of type 2 diabetes mellitus (T2DM). Obesity and insulin resistance are associated with a chronic but subclinical inflammatory process that impairs insulin action in most tissues and could also hamper pancreatic β-cell function. The involvement of monocytic cells and the profiles of the chemokines and cytokines induced by this inflammation suggest an innate immune response. However, emerging data indicate that elements of the adaptive immune system could also be involved. As activation of an adaptive response requires antigen specificity, some researchers have hypothesized that T2DM evolves from an innate immune response to an autoimmune condition. In this Perspectives article, we present the arguments for and against this hypothesis and discuss which mechanisms could be involved in a putative switch from innate immunity to autoimmunity.
Collapse
Affiliation(s)
- Lício A Velloso
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Centre, University of Campinas, DCM-FCM UNICAMP, 13,084-970 Campinas, São Paulo, Brazil
| | | | | |
Collapse
|
23
|
da Rosa LC, Chiuso-Minicucci F, Zorzella-Pezavento SFG, França TGD, Ishikawa LLW, Colavite PM, Balbino B, Tavares LCB, Silva CL, Marques C, Ikoma MRV, Sartori A. Bacille Calmette-Guérin/DNAhsp65 prime-boost is protective against diabetes in non-obese diabetic mice but not in the streptozotocin model of type 1 diabetes. Clin Exp Immunol 2013; 173:430-7. [PMID: 23692306 DOI: 10.1111/cei.12140] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2013] [Indexed: 12/14/2022] Open
Abstract
Type I diabetes is a disease caused by autoimmune destruction of the beta cells in the pancreas that leads to a deficiency in insulin production. The aim of this study was to evaluate the prophylactic potential of a prime-boost strategy involving bacille Calmette-Guérin (BCG) and the pVAXhsp65 vaccine (BCG/DNAhsp65) in diabetes induced by streptozotocin (STZ) in C57BL/6 mice and also in spontaneous type 1 diabetes in non-obese diabetic (NOD) mice. BCG/DNAhsp65 vaccination in NOD mice determined weight gain, protection against hyperglycaemia, decreased islet inflammation, higher levels of cytokine production by the spleen and a reduced number of regulatory T cells in the spleen compared with non-immunized NOD mice. In the STZ model, however, there was no significant difference in the clinical parameters. Although this vaccination strategy did not protect mice in the STZ model, it was very effective in NOD mice. This is the first report demonstrating that a prime-boost strategy could be explored as an immunomodulatory procedure in autoimmune diseases.
Collapse
Affiliation(s)
- L C da Rosa
- Department of Microbiology and Immunology, Biosciences Institute, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mitchell T, Johnson MS, Ouyang X, Chacko BK, Mitra K, Lei X, Gai Y, Moore DR, Barnes S, Zhang J, Koizumi A, Ramanadham S, Darley-Usmar VM. Dysfunctional mitochondrial bioenergetics and oxidative stress in Akita(+/Ins2)-derived β-cells. Am J Physiol Endocrinol Metab 2013; 305:E585-99. [PMID: 23820623 PMCID: PMC3761167 DOI: 10.1152/ajpendo.00093.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Insulin release from pancreatic β-cells plays a critical role in blood glucose homeostasis, and β-cell dysfunction leads to the development of diabetes mellitus. In cases of monogenic type 1 diabetes mellitus (T1DM) that involve mutations in the insulin gene, we hypothesized that misfolding of insulin could result in endoplasmic reticulum (ER) stress, oxidant production, and mitochondrial damage. To address this, we used the Akita(+/Ins2) T1DM model in which misfolding of the insulin 2 gene leads to ER stress-mediated β-cell death and thapsigargin to induce ER stress in two different β-cell lines and in intact mouse islets. Using transformed pancreatic β-cell lines generated from wild-type Ins2(+/+) (WT) and Akita(+/Ins2) mice, we evaluated cellular bioenergetics, oxidative stress, mitochondrial protein levels, and autophagic flux to determine whether changes in these processes contribute to β-cell dysfunction. In addition, we induced ER stress pharmacologically using thapsigargin in WT β-cells, INS-1 cells, and intact mouse islets to examine the effects of ER stress on mitochondrial function. Our data reveal that Akita(+/Ins2)-derived β-cells have increased mitochondrial dysfunction, oxidant production, mtDNA damage, and alterations in mitochondrial protein levels that are not corrected by autophagy. Together, these findings suggest that deterioration in mitochondrial function due to an oxidative environment and ER stress contributes to β-cell dysfunction and could contribute to T1DM in which mutations in insulin occur.
Collapse
|
25
|
Smith CJ, Lawrence CB, Rodriguez-Grande B, Kovacs KJ, Pradillo JM, Denes A. The immune system in stroke: clinical challenges and their translation to experimental research. J Neuroimmune Pharmacol 2013; 8:867-87. [PMID: 23673977 DOI: 10.1007/s11481-013-9469-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/28/2013] [Indexed: 12/27/2022]
Abstract
Stroke represents an unresolved challenge for both developed and developing countries and has a huge socio-economic impact. Although considerable effort has been made to limit stroke incidence and improve outcome, strategies aimed at protecting injured neurons in the brain have all failed. This failure is likely to be due to both the incompleteness of modelling the disease and its causes in experimental research, and also the lack of understanding of how systemic mechanisms lead to an acute cerebrovascular event or contribute to outcome. Inflammation has been implicated in all forms of brain injury and it is now clear that immune mechanisms profoundly influence (and are responsible for the development of) risk and causation of stroke, and the outcome following the onset of cerebral ischemia. Until very recently, systemic inflammatory mechanisms, with respect to common comorbidities in stroke, have largely been ignored in experimental studies. The main aim is therefore to understand interactions between the immune system and brain injury in order to develop novel therapeutic approaches. Recent data from clinical and experimental research clearly show that systemic inflammatory diseases -such as atherosclerosis, obesity, diabetes or infection - similar to stress and advanced age, are associated with dysregulated immune responses which can profoundly contribute to cerebrovascular inflammation and injury in the central nervous system. In this review, we summarize recent advances in the field of inflammation and stroke, focusing on the challenges of translation between pre-clinical and clinical studies, and potential anti-inflammatory/immunomodulatory therapeutic approaches.
Collapse
Affiliation(s)
- Craig J Smith
- Stroke and Vascular Research Centre, Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Science Centre, Salford Royal Foundation Trust, Salford M6 8HD, UK.
| | | | | | | | | | | |
Collapse
|
26
|
Wang P, Moore A. Theranostic magnetic resonance imaging of type 1 diabetes and pancreatic islet transplantation. Quant Imaging Med Surg 2012; 2:151-62. [PMID: 23256077 DOI: 10.3978/j.issn.2223-4292.2012.08.04] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/27/2012] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes mellitus results in impaired insulin production by pancreatic islets due to autoimmunity. Islet transplantation has recently emerged as a promising treatment for this disease. To visualize and monitor endogenous and transplanted islets, non-invasive strategies are currently being developed. These include strategies for in vivo magnetic resonance imaging of microvascular changes during diabetes development, tracking the recruitment of diabetogenic T cells to the pancreas, and imaging of endogenous and transplanted islet mass. The combination of MR imaging agents with therapy is a novel state-of-the-art theranostic approach that has a tremendous potential for type 1 diabetes management. Though still in its infancy, theranostic MR imaging has shown certain encouraging progress. Here we provide an overview of the latest accomplishments in this area as it applies to changes in islet vasculature during diabetes development, monitoring autoimmune attack mediated by T cells, and imaging of transplanted islets. Future challenges and opportunities in the area of theranostic MRI are discussed as well.
Collapse
Affiliation(s)
- Ping Wang
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, USA
| | | |
Collapse
|
27
|
Abstract
Type 1 diabetes (T1D) represents 10 to 15% of all forms of diabetes. Its incidence shows a consistent rise in all countries under survey. Evidence for autoimmunity in human T1D relies on the detection of insulitis, of islet cell antibodies, of activated β-cell-specific T lymphocytes and on the association of T1D with a restricted set of class II major histocompatibility complex (MHC) alleles. However, mechanisms that initiate the failure of immune tolerance to β-cell autoantigens remain elusive in common forms of T1D. T1D commonly develop as a multifactorial disease in which environmental factors concur with a highly multigenic background. The disease is driven by the activation of T-lymphocytes against pancreatic β-cells. Several years elapse between initial triggering of the autoimmune response to β cells, as evidenced by the appearance or islet cell autoantibodies, and the onset of clinical diabetes, defining a prediabetes stage. Active mechanisms hold back autoreactive effector T-cells in prediabetes, in particular a subset of CD4+ T-cells (T(reg)) and other regulatory T-cells, such as invariant NKT cells. There is evidence in experimental models that systemic or local infections can trigger autoimmune reactions to β-cells. However, epidemiological observations that have accumulated over years have failed to identify undisputable environmental factors that trigger T1D. Moreover, multiple environmental factors may intervene in the disease evolution and protective as weel as triggering environmental factors may be involved. Available models also indicate that local signals within the islets are required for full-blown diabetes to develop. Many autoantigens that are expressed by β-cells but also by the other endocrine islet cells and by neurons are recognized by lymphocytes along the development of T1D. The immune image of β-cells is that of native components of the β-cell membrane, as seen by B-lymphocytes, and of fragments of intracellular β-cell proteins in the form of peptides loaded onto class I MHC molecules on the β-cell surface and class I and class II molecules onto professional antigen presenting cells. Given the key role of T lymphocytes in T1D, the cartography of autoantigen-derived peptides that are presented to class I-restricted CD8(+) T-cells and class II-restricted CD4(+) T-cells is of outmost importance and is a necessary step in the development of diagnostic T-cell assays and of immunotherapy of T1D.
Collapse
|
28
|
Abstract
Diabetes is a disease characterized by a relative or absolute lack of insulin, leading to hyperglycaemia. There are two main types of diabetes: type 1 diabetes and type 2 diabetes. Type 1 diabetes is due to an autoimmune destruction of the insulin-producing pancreatic beta cells, and type 2 diabetes is caused by insulin resistance coupled by a failure of the beta cell to compensate. Animal models for type 1 diabetes range from animals with spontaneously developing autoimmune diabetes to chemical ablation of the pancreatic beta cells. Type 2 diabetes is modelled in both obese and non-obese animal models with varying degrees of insulin resistance and beta cell failure. This review outlines some of the models currently used in diabetes research. In addition, the use of transgenic and knock-out mouse models is discussed. Ideally, more than one animal model should be used to represent the diversity seen in human diabetic patients.
Collapse
|
29
|
Gene expression profile in bone of diabetes-prone BB/OK rats fed a high-fat diet. GENES AND NUTRITION 2012; 8:99-104. [PMID: 22639302 DOI: 10.1007/s12263-012-0299-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 05/02/2012] [Indexed: 12/13/2022]
Abstract
A high-fat diet (HFD) has been recognized as a risk factor for diseases such as dyslipidemia, atherosclerosis, obesity, and osteoporosis. However, studies analyzing gene expression after HFD in bone are rare. That prompted us to analyze the expression of selected genes in bone of 4-week-old diabetes-prone B(io)B(reeding) rats. Two breeding pairs were fed a HFD (+10 % tallow) or were fed a normal diet (ND; Ssniff R-Z) before mating and afterward during pregnancy. After the birth of progeny, parents continued to be given HFD or ND until the progeny was weaned (3 weeks). Thereafter, offspring were weaned and were fed the same food as their parents up to an age of 4 weeks. Body weight was measured at an age of 4 weeks, and subsequently 13 HFD rats and 13 ND rats were killed and the tibial bone was harvested to analyze the expression of 53 genes in bone. All rats fed HFD were significantly heavier than rats fed ND after 3 and 4 weeks. The diet also influenced the expression of genes in bone. There were significant differences in 20 out of 53 genes studied between rats fed HFD compared with rats fed ND. Four out of 20 had a lower and 17 out of 20 genes a higher expression in HFD rats, but differences in gene expression showed obvious differences between males and females. There were only two genes that were similarly different between males and females: Bmp4 and Atf4. Two genes, Foxg1 and Npy, were inversely expressed in males and females. It seems that the gene expression is differently regulated by diet during pregnancy and later in life between males and females. Nevertheless, it cannot be excluded that HFD also acts as an epigenetic factor in the development of offspring in utero.
Collapse
|
30
|
Kiekhaefer CM, Weber B, Huggins M, Gorichanaz C, Nehring JA, DeLuca HF. 2α-Methyl-19-nor-(20S)-1,25-dihydroxyvitamin D(3) protects the insulin 2 knockout non-obese diabetic mouse from developing type 1 diabetes without hypercalcaemia. Clin Exp Immunol 2012; 166:325-32. [PMID: 22059989 DOI: 10.1111/j.1365-2249.2011.04481.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that destroys the insulin-producing beta-islet cells of the pancreas. Currently, there are no treatment modalities for prevention of T1D, and the mechanisms influencing disease inception and early progression are not well understood. We have used the insulin 2(-/-) non-obese diabetic (Ins2(-/-) NOD) model to study stages of T1D and to examine the protective effects of a potent analogue of 1α,25-dihydroxyvitamin D(3), 2α-methyl-19-nor-(20S)-1α,25-dihydroxyvitamin D(3) (2AMD). Pancreatic tissues from control and 2AMD-treated Ins2(-/-) NOD mice were obtained weekly from 5 to 16 weeks of age. Using immunohistochemical (IHC) analysis, samples were analysed for changes in beta cell survival, islet structure and T cell invasion. Weekly intraperitoneal glucose tolerance tests (IPGTT) were performed to assess comparative beta cell function in control and treated animals. IHC demonstrated progressive beta cell destruction in control mice. In contrast, 2AMD treatment preserved islet cell architecture, arrested intra-islet T cell invasion and prevented the transition from insulitis to diabetes. IPGTT results revealed progressive impairment of beta cell function with increasing age in control mice, while 2AMD treatment resulted in normal beta function throughout the study. These results demonstrate that the Ins2(-/-) NOD model provides a rapid and effective method for studying T1D and for assessing efficacy of anti-diabetic agents.
Collapse
Affiliation(s)
- C M Kiekhaefer
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706-1544, USA
| | | | | | | | | | | |
Collapse
|
31
|
Polychronakos C, Li Q. Understanding type 1 diabetes through genetics: advances and prospects. Nat Rev Genet 2011; 12:781-92. [PMID: 22005987 DOI: 10.1038/nrg3069] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Starting with early crucial discoveries of the role of the major histocompatibility complex, genetic studies have long had a role in understanding the biology of type 1 diabetes (T1D), which is one of the most heritable common diseases. Recent genome-wide association studies (GWASs) have given us a clearer picture of the allelic architecture of genetic susceptibility to T1D. Fine mapping and functional studies are gradually revealing the complex mechanisms whereby immune self-tolerance is lost, involving multiple aspects of adaptive immunity. The triggering of these events by dysregulation of the innate immune system has also been implicated by genetic evidence. Finally, genetic prediction of T1D risk is showing promise of use for preventive strategies.
Collapse
Affiliation(s)
- Constantin Polychronakos
- Departments of Pediatrics and Human Genetics, McGill University, Montreal, Québec, Canada H3H 1P3. Constantin.
| | | |
Collapse
|
32
|
Bahr J, Klöting N, Wilke B, Klöting I, Follak N. High-fat diet protects BB/OK rats from developing type 1 diabetes. Diabetes Metab Res Rev 2011; 27:552-6. [PMID: 21523881 DOI: 10.1002/dmrr.1207] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND It is well known that lipid metabolism plays an important role in the early stages of type 1 diabetes (T1D). For that reason, we examined factors that influence lipid metabolism of BioBreeding/Ottawa Kalsburg (BB/OK) rats that spontaneously develop an insulin-dependent T1D. METHODS BB/OK female rats were fed a high-fat diet during pregnancy (Ssniff R-Z + 10% tallow) and their progeny were also given this diet up to an age of 30 weeks (n = 55) or 4 weeks (n = 14) to study gene expression of Pparg, Fasn, Lep, Adipoq, Repin1, Rarres 2, and Glut4 in adipose tissue. Forty-two BB/OK rats fed the normal diet (Ssniff R-Z) during pregnancy and the observation period served as controls. RESULTS The high-fat diet significantly decreased diabetes frequency in BB/OK rats when compared with control rats (71 versus 95%, p = 0.002). Although this difference was also reflected in the male rats (68 versus 100%, p = 0.003), no significant variation was observed in female rats (73 versus 90%, p = 0.23). The high-fat diet resulted in significantly reduced mRNA expression of examined genes in subcutaneous adipose tissue, but not in visceral adipose tissue, except for Fasn and Repin1 expression. CONCLUSIONS A high-fat diet seems to protect BB/OK rats from T1D in a sex-specific manner. The data suggest that a high-fat diet might influence fat accumulation and/or fat metabolism and prevent T1D development in male rats, which is supported by changes in adipose tissue gene expression.
Collapse
Affiliation(s)
- Jeanette Bahr
- Department of Laboratory Animal Science, Medical Faculty, University of Greifswald, Karlsburg, Germany
| | | | | | | | | |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW To update on the clinical trials using antigen-specific therapies in autoimmune diabetes. RECENT FINDINGS Type 1 diabetes is now a predictable disease with the measurement of islet autoantibodies, and the incidence is increasing dramatically. Well tolerated and effective interventions are needed to stop the underlying autoimmune destruction of insulin-producing beta cells. Beta-cell antigens, insulin and glutamic acid decarboxylase, are being used to preserve endogenous insulin production in individuals with new-onset diabetes and to prevent diabetes. The results of antigen-specific immune intervention trials are reviewed and consideration is given to future directions for inducing tolerance in type 1 diabetes. SUMMARY Antigen-specific immune therapies act by enhancing regulatory T cell function, in animal models often locally and selectively in islets or pancreatic lymph nodes while inhibiting effector T cells. This therapeutic pathway provides a safe treatment to preserve beta cell function in new-onset diabetic individuals with the GAD-Alum vaccine being the most extensively studied therapy. Insulin is being used in many forms to prevent diabetes and stop the underlying autoimmune process. For the future, combination immune therapies targeting different pathways in the immune system will be needed to effectively induce sustained tolerance in type 1 diabetes.
Collapse
Affiliation(s)
- Aaron W. Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Court, MS B140, PO BOX 6511, Aurora, CO 80045, , Phone: 303-724-6812, Fax: 303-724-6839
| | - Matthias von Herrath
- Center for Type 1 Diabetes, La Jolla Institute for Allergy and Immunology, La Jolla, California,
| |
Collapse
|
34
|
Ghaemi Oskouie F, Shameli A, Yang A, Desrosiers MD, Mucsi AD, Blackburn MR, Yang Y, Santamaria P, Shi Y. High Levels of Adenosine Deaminase on Dendritic Cells Promote Autoreactive T Cell Activation and Diabetes in Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 2011; 186:6798-806. [DOI: 10.4049/jimmunol.1004222] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
35
|
Bahr J, Klöting N, Klöting I, Follak N. Transplantation of adipose tissue protects BB/OK rats from type 1 diabetes development. Transpl Immunol 2011; 24:238-40. [PMID: 21277980 DOI: 10.1016/j.trim.2011.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 01/18/2011] [Accepted: 01/19/2011] [Indexed: 01/08/2023]
Abstract
B(io) B(reedding)/O(ttawa) K(alsburg) rats spontaneously develop insulin-dependent type 1 diabetes. Days before BB/OK rats become diabetic, their body seems to be flabby which may be attributed to loss of subcutaneous fat. However, the rats are normoglycemic and manifest 3-4 days later. This observation prompted us to search for possibilities to avoid the loss of adipose tissue. BB/OK rats were subcutaneously grafted with visceral adipose tissue. In total, 34 (71%) out of 48 male and 23 (49%) out of 47 female BB/OK rats grafted with adipose tissue developed type 1 diabetes so that significantly more females than males were protected from diabetes development (p=0.03). In the control group, 17 (85%) out of 20 male and 20 (95%) out of 21 female BB/OK rats were diabetic. Adipose tissue transplantation can protect BB/OK rats from type 1 diabetes development in a sex specific manner. One could conclude that the manipulations have influenced fat accumulation and/or fat metabolism which prevent type 1 diabetes development in about 50% of BB/OK rats. This idea is supported by the finding that a mutation in the leptin receptor of NOD mice suppresses type 1 diabetes progression.
Collapse
Affiliation(s)
- Jeanette Bahr
- Department of Laboratory Animal Science, Medical Faculty, University of Greifswald, Germany
| | | | | | | |
Collapse
|
36
|
van den Brandt J, Fischer HJ, Walter L, Hünig T, Klöting I, Reichardt HM. Type 1 diabetes in BioBreeding rats is critically linked to an imbalance between Th17 and regulatory T cells and an altered TCR repertoire. THE JOURNAL OF IMMUNOLOGY 2010; 185:2285-94. [PMID: 20644174 DOI: 10.4049/jimmunol.1000462] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Diabetes-prone BioBreeding (DP-BB) rats spontaneously develop type 1 diabetes mellitus (T1DM) on grounds of their MHC haplotype RT1(u) and a point mutation in the Gimap5 gene. In this study, we report that DP-BB rats exhibit an increasingly severe imbalance, in particular between Th17 and regulatory T (T(reg)) cells, within the first months of age. This can be assigned to an excess in effector T cells because neither the percentage nor the function of the T(reg) cells is compromised. Flow cytometric analysis of Vbeta segment usage and CDR3 spectratyping further suggest that the disturbed repertoire of peripheral T cells may also contribute to the development of T1DM in DP-BB rats. Importantly, expansion of T(reg) cells in vivo by means of a CD28 superagonistic Ab as well as adoptive transfer of T(reg) cells efficiently interferes with the development of T1DM in DP-BB rats, whereas treatment with conventional Th cells does not afford protection. Using a newly generated strain of enhanced GFP transgenic rats, we could further demonstrate that the transferred T(reg) cells persist in the recipient rats for several months and partially correct the imbalance between Th17 and T(reg) cells. Thus, our data support the hypothesis that unchecked effector T cell action and a disturbed T cell repertoire contribute to the development of T1DM in DP-BB rats, which may also have implications for a better understanding of the human disease.
Collapse
Affiliation(s)
- Jens van den Brandt
- Department of Cellular and Molecular Immunology, University of Göttingen Medical School, Germany
| | | | | | | | | | | |
Collapse
|
37
|
La Torre D, Lernmark A. Immunology of beta-cell destruction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:537-83. [PMID: 20217514 DOI: 10.1007/978-90-481-3271-3_24] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pancreatic islet beta-cells are the target for an autoimmune process that eventually results in an inability to control blood glucose due to the lack of insulin. The different steps that eventually lead to the complete loss of the beta-cells are reviewed to include the very first step of a triggering event that initiates the development of beta-cell autoimmunity to the last step of appearance of islet-cell autoantibodies, which may mark that insulitis is about to form. The observations that the initial beta-cell destruction by virus or other environmental factors triggers islet autoimmunity not in the islets but in the draining pancreatic lymph nodes are reviewed along with possible basic mechanisms of loss of tolerance to islet autoantigens. Once islet autoimmunity is established the question is how beta-cells are progressively killed by autoreactive lymphocytes which eventually results in chronic insulitis. Many of these series of events have been dissected in spontaneously diabetic mice or rats, but controlled clinical trials have shown that rodent observations are not always translated into mechanisms in humans. Attempts are therefore needed to clarify the step 1 triggering mechanisms and the step to chronic autoimmune insulitis to develop evidence-based treatment approaches to prevent type 1 diabetes.
Collapse
Affiliation(s)
- Daria La Torre
- Lund University, CRC, Department of Clinical Sciences, University Hospital MAS, SE-205 02, Malmö, Sweden.
| | | |
Collapse
|
38
|
Miwako I, Shudo K. Oral administration of synthetic retinoid Am80 inhibits the development of type 1 diabetes in non-obese diabetic (NOD) mice. Biol Pharm Bull 2009; 32:157-9. [PMID: 19122301 DOI: 10.1248/bpb.32.157] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Synthetic retinoid Am80 is a potent modulator of the immune system. Am80 is effective in various experimentally induced autoimmune disorders. The purpose of this study is to confirm its effect on non-obese diabetic (NOD) mice, which spontaneously develop autoimmune type 1 diabetes. Am80 was orally administered in feed to 6 NOD mice per group at a dose of 0 (control), 0.1 (low) or 1 (high) mg/kg/d for 19 weeks. During the experiment period, the high urine glucose levels were observed in 33% mice of the control and low Am80 groups, whereas any mouse in the high Am80 group did not show abnormal urine glucose level. Histological examination showed that the average score of insulitis severity in the low Am80 group was similar to that in the control; however in the high Am80 group, the score was significantly reduced compared to that in the control group. Similarly, the severity of lymphocyte infiltration in the submandibular glands showed a tendency to decrease in the high Am80 group, but not in the low Am80 group, compared to the control. These data strongly suggest that the development of type 1 diabetes in NOD mice can be inhibited by oral administration of Am80.
Collapse
|
39
|
Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol 2009; 155:173-81. [PMID: 19128359 DOI: 10.1111/j.1365-2249.2008.03860.x] [Citation(s) in RCA: 490] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The immunopathology of type 1 diabetes (T1D) has proved difficult to study in man because of the limited availability of appropriate samples, but we now report a detailed study charting the evolution of insulitis in human T1D. Pancreas samples removed post-mortem from 29 patients (mean age 11.7 years) with recent-onset T1D were analysed by immunohistochemistry. The cell types constituting the inflammatory infiltrate within islets (insulitis) were determined in parallel with islet insulin content. CD8(+) cytotoxic T cells were the most abundant population during insulitis. Macrophages (CD68(+)) were also present during both early and later insulitis, although in fewer numbers. CD20(+) cells were present in only small numbers in early insulitis but were recruited to islets as beta cell death progressed. CD138(+) plasma cells were infrequent at all stages of insulitis. CD4(+) cells were present in the islet infiltrate in all patients but were less abundant than CD8(+) or CD68(+) cells. Forkhead box protein P3(+) regulatory T cells were detected in the islets of only a single patient. Natural killer cells were detected rarely, even in heavily inflamed islets. The results suggest a defined sequence of immune cell recruitment in human T1D. They imply that both CD8(+) cytotoxic cells and macrophages may contribute to beta cell death during early insulitis. CD20(+) cells are recruited in greatest numbers during late insulitis, suggesting an increasing role for these cells as insulitis develops. Natural killer cells and forkhead box protein P3(+) T cells do not appear to be required for beta cell death.
Collapse
Affiliation(s)
- A Willcox
- Institute of Biomedical and Clinical Sciences, Peninsula Medical School, Tamar Science Park, Plymouth, UK
| | | | | | | | | |
Collapse
|
40
|
Russell JC. Evaluating micro- and macro-vascular disease, the end stage of atherosclerosis, in rat models. Methods Mol Biol 2009; 573:17-44. [PMID: 19763920 DOI: 10.1007/978-1-60761-247-6_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Development of effective treatment or, more critically, preventative measures against atherosclerosis and cardiovascular disease will require animal models that mimic the disease processes seen in humans and permit identification of the genetic and physiological factors. The Rat is normally resistant to cardiovascular disease, but a number of genetic mutations make affected strains of rats highly susceptible to atherosclerosis and micro- and macro-vascular disease that is highly analogous to human disease. These models of obesity develop the metabolic syndrome and type 2 diabetes, hyperinsulinemia, hyperlipidemia, vascular and myocardial dysfunction, and end-stage lesions in the heart and kidney. The models offer the prospect of both genetic and molecular biology studies that are linked directly to spontaneous cardiovascular disease and exploration of putative preventative or treatment approaches, including pharmaceutical agents. Use of small animal models of cardiovascular disease is dependent on appropriate experimental design and techniques that take account of the complex nature of the disease processes. Detailed experimental procedures for the use of rat models, including handling and treatment of animals, choice of experimental variables and endpoints, assay methods, and histological and electron microscopy techniques are covered in this chapter.
Collapse
Affiliation(s)
- James C Russell
- Alberta Institute for Human Nutrition, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
41
|
Abstract
Apoptotic β-cell death appears to be central to the pathogenesis of Type 1 diabetes mellitus and in islet graft rejection. The β-cell destruction is partially mediated by cytokines, such as IL-1β (interleukin 1β), TNFα (tumour necrosis factor α) and IFN-γ (interferon γ). IL-1β and TNFα mediate activation of the transcription factor NF-κB (nuclear factor κB) pathway. Use of a degradation-resistant NF-κB protein inhibitor (ΔNIκBα), specifically expressed in β-cells, significantly reduced IL-1β+IFN-γ-induced apoptosis. Moreover, in vivo, it protected against multiple low-dose streptozocin-induced diabetes, with reduced intra-islet lymphocytic infiltration. Thus β-cell-specific activation of NF-κB is a key event in the progressive loss of β-cells in diabetes. Inhibition of this process could be a potential effective strategy for β-cell protection.
Collapse
|
42
|
Ounissi-Benkalha H, Polychronakos C. The molecular genetics of type 1 diabetes: new genes and emerging mechanisms. Trends Mol Med 2008; 14:268-75. [PMID: 18482868 DOI: 10.1016/j.molmed.2008.04.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 04/08/2008] [Accepted: 04/08/2008] [Indexed: 12/12/2022]
Abstract
Susceptibility to type 1 diabetes (T1D) is determined by complex interactions between several genetic loci and environmental factors. Alleles at the human leukocyte antigen (HLA) locus explain up to 50% of the familial clustering of T1D, and the remainder is contributed to by multiple loci, of which only four were known until recently. First-stage results of genome-wide association (GWA) studies performed with high-density genotyping arrays have already produced four novel loci and the promise that, with the completion of the second stage of the GWA studies, most of the genetic basis of T1D will be known. We will review what is known to date about the mechanisms of genetic susceptibility to T1D, with special emphasis on possible diagnostic and therapeutic applications of these recent genetic findings.
Collapse
Affiliation(s)
- Houria Ounissi-Benkalha
- Endocrine Genetics Laboratory, McGill University Health Center, Montreal Children's Hospital Research Institute, 2300 Tupper, Montréal, Québec H3H 1P3, Canada
| | | |
Collapse
|
43
|
Abstract
About 10% of patients with the clinical presentation of type 2 diabetes suffer from an autoimmune form of diabetes associated with a rapid decline of residual beta-cell mass and subsequent development of insulin dependency. In this condition, called latent autoimmune diabetes in adults (LADA), there are clinical and metabolic features intermediate between type 1 and type 2 diabetes. Recent studies provide novel information on the immune markers associated with progressive beta-cell loss in LADA patients. However, LADA pathogenesis is still poorly understood; further studies are needed to establish general recommendation for preventing and treating this subtype of autoimmune diabetes.
Collapse
Affiliation(s)
- Jochen Seissler
- Diabetes Center, Medical Clinic Innenstadt, Ludwig-Maximilians-University Munich, Ziemssenstrasse 1, München 80336, Germany.
| |
Collapse
|
44
|
Pirot P, Cardozo AK, Eizirik DL. Mediators and mechanisms of pancreatic beta-cell death in type 1 diabetes. ACTA ACUST UNITED AC 2008; 52:156-65. [DOI: 10.1590/s0004-27302008000200003] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Accepted: 12/03/2007] [Indexed: 12/27/2022]
Abstract
Type 1 diabetes mellitus (T1D) is characterized by severe insulin deficiency resulting from chronic and progressive destruction of pancreatic beta-cells by the immune system. The triggering of autoimmunity against the beta-cells is probably caused by environmental agent(s) acting in the context of a predisposing genetic background. Once activated, the immune cells invade the islets and mediate their deleterious effects on beta-cells via mechanisms such as Fas/FasL, perforin/granzyme, reactive oxygen and nitrogen species and pro-inflammatory cytokines. Binding of cytokines to their receptors on the beta-cells activates MAP-kinases and the transcription factors STAT-1 and NFkappa-B, provoking functional impairment, endoplasmic reticulum stress and ultimately apoptosis. This review discusses the potential mediators and mechanisms leading to beta-cell destruction in T1D.
Collapse
|
45
|
Eisenbarth GS, Jeffrey J. The natural history of type 1A diabetes. ACTA ACUST UNITED AC 2008; 52:146-55. [DOI: 10.1590/s0004-27302008000200002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Accepted: 02/18/2008] [Indexed: 01/16/2023]
Abstract
We can now predict the development of Type 1A (Immune Mediated) diabetes primarily through the determination of four biochemically characterized islet autoantibodies [insulin, GAD65, IA-2 (ICA512) and (Znt8)]. Prediction is possible because beta-cell destruction is chronically progressive and very slow in most, but not all individuals. We can also prevent type 1A diabetes in animal models and a major goal is the prevention of type 1A diabetes in man with multiple clinical trials underway.
Collapse
Affiliation(s)
| | - Joy Jeffrey
- University of Colorado at Denver and Health Sciences Center, USA
| |
Collapse
|
46
|
Matteucci E, Giampietro O. Proposal open for discussion: defining agreed diagnostic procedures in experimental diabetes research. JOURNAL OF ETHNOPHARMACOLOGY 2008; 115:163-172. [PMID: 17961942 DOI: 10.1016/j.jep.2007.08.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 08/21/2007] [Accepted: 08/23/2007] [Indexed: 05/25/2023]
Abstract
BACKGROUND Animal experimentation has a long tradition in diabetes research and has provided invaluable benefits with regard to insulin discovery and treatment assessment. METHODS The review focuses on chemical-induced diabetes in rats and surveys the protocols of diabetes induction, diabetes diagnosis, and glucose tolerance evaluation in a selection of recent research. RESULTS This brief review of techniques in experimental diabetes highlights that there is no uniformity, whereas standardisation of procedures is desirable so that comparability will exist among experiments carried out in different settings. CONCLUSIONS On this basis, questions are put and standards are proposed. It would be a platform to promote the exchange of ideas through expert consultation about practical issues related to animal research and a basis on which standards can be set according to user requirements and animal respect.
Collapse
|
47
|
Choi MS, Jung UJ, Yeo J, Kim MJ, Lee MK. Genistein and daidzein prevent diabetes onset by elevating insulin level and altering hepatic gluconeogenic and lipogenic enzyme activities in non-obese diabetic (NOD) mice. Diabetes Metab Res Rev 2008; 24:74-81. [PMID: 17932873 DOI: 10.1002/dmrr.780] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Non-obese diabetic (NOD) mice are regarded as being excellent animal models of human type 1 diabetes or insulin dependent diabetes (IDDM). This study investigated the beneficial effects of genistein and daidzein on IDDM, an autoimmune disease. METHODS Female NOD mice were divided into control, genistein (0.02%, w/w) and daidzein (0.02%, w/w) groups. Blood glucose level, plasma biomarkers, hepatic glucose and lipid regulating enzyme activities and pancreas immunohistochemistry analysis were examined after a 9-week experimental period. RESULTS Blood glucose levels of genistein and daidzein groups were 40 and 36% of control value at the end of study (9th week). The genistein and daidzein supplements increased insulin/glucagon ratio and C-peptide level with preservation of insulin staining beta-cell of pancreas in the NOD mice. In the liver, genistein and daidzein supplements resulted in lowering glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) activities, while increasing two lipogenic enzymes activities, malic enzyme and glucose-6-phosphate dehydrogenase (G6PD), compared to the control group. Significantly, genistein and daidzein supplementation lowered the activities of fatty acid beta-oxidation and carnitine palmitoyltransferase (CPT) in these mice. Genistein and daidzein also improved plasma triglyceride and free fatty acid (FFA) concentrations compared to the control group. CONCLUSIONS These results suggest that genistein and daidzein play important roles in regulation of glucose homeostasis in type 1 diabetic mice by down-regulating G6Pase, PEPCK, fatty acid beta-oxidation and CPT activities, while up-regulating malic enzyme and G6PD activities in liver with preservation of pancreatic beta-cells. The supplementation of genistein and daidzein are seemingly helpful for preventing IDDM onset.
Collapse
Affiliation(s)
- M S Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, 702-701, Republic of Korea
| | | | | | | | | |
Collapse
|
48
|
Pontes Andersen CC, Flyvbjerg A, Buschard K, Holmstrup P. Relationship Between Periodontitis and Diabetes: Lessons From Rodent Studies. J Periodontol 2007; 78:1264-75. [PMID: 17608582 DOI: 10.1902/jop.2007.060491] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND A great amount of investigations have provided evidence that both type 1 and type 2 diabetes increase the risk and severity of periodontitis; several alterations in the diabetic periodontium are likely to be involved. Conversely, periodontitis has been shown to have an impact on diabetes, although less evidence is available on the underlying mechanisms. The association between periodontitis and diabetes has been discussed in several reviews over the past years; however, none has focused on the use and contribution of rodent models. METHODS This review describes the most commonly used rodent models of diabetes, periodontitis, and the association between the two diseases. Further, we summarize the influence of diabetes in the periodontium as well as the effect of periodontitis on diabetes status with special focus on evidence from experimental studies. RESULTS Rodent studies have confirmed human findings and further increased our knowledge on the alterations in the diabetic periodontium. On the other hand, few rodent investigations have explored the consequences of periodontitis for diabetes. Their results clearly indicate that periodontitis can become a health hazard not only for diabetes but also for prediabetes; the exact mechanisms are still to be unraveled. CONCLUSIONS Findings from rodent studies have been useful in increasing our understanding of periodontitis, diabetes, and their association and hold great promise for future investigations given the wide variety of possibilities for testing biologic hypotheses.
Collapse
Affiliation(s)
- Carla C Pontes Andersen
- Department of Periodontology, School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
49
|
Schäffer M, Bongartz M, Fischer S, Proksch B, Viebahn R. Nitric oxide restores impaired healing in normoglycaemic diabetic rats. J Wound Care 2007; 16:311-6. [PMID: 17708383 DOI: 10.12968/jowc.2007.16.7.27057] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Hyperglycaemia impairs wound healing. However, little is known about the underlying cellular mechanisms that lead to diminished wound repair in insulin-controlled and non-insulin-controlled diabetes. This study investigated the role of endogenous and exogenous nitric oxide on incisional wound healing in diabetic rats. METHOD Groups of 10 wild-typeWistar control rats - 10 genetically diabetic BioBreeding rats and 10 genetically diabetic BioBreeding rats treated with subcutaneous insulin implants to render them normoglycaemic - underwent dorsal skin incision followed by subcutaneous insertion of polyvinyl alcohol sponges. The rats were sacrificed 10 days later to determine the wound-breaking strength and reparative collagen deposition. Nitric oxide, an important mediator in diabetic wound healing and collagen synthesis, was measured in wound fluid. Wound-derived fibroblasts were tested for ex vivo synthesis of nitric oxide and collagen. Exogenous nitric oxide was used for the therapeutic interventions. RESULTS Wound-breaking strength and wound collagen deposition were significantly impaired in the hyperglycaemic diabetic animals (p<0.01). Wound nitric-oxide synthesis and ex vivo wound fibroblast nitric-oxide production were reduced in the hyperglycaemic rats (p<0.01). Insulin treatment partially reversed some of the effects of hyperglycaemia on wound repair (p<0.05). Exogenous nitric oxide further restored wound mechanical strength, collagen deposition and fibroblast collagen synthesis (p<0.01) in insulin-treated (normoglycaemic) diabetic animals. CONCLUSION Wound healing is impaired in hyperglycaemic and normoglycaemic diabetic rats. This is reflected in impaired wound fibroblast nitric-oxide synthesis. Used in combination with insulin, exogenous nitric oxide further improves healing outcomes, making it a potential target for therapeutic intervention in insulin-treated normoglycaemic diabetes.
Collapse
Affiliation(s)
- M Schäffer
- I Department of Surgery, University Hospital, Knappschaftskrankenhaus Bochum-Langendreer, Germany.
| | | | | | | | | |
Collapse
|
50
|
Wiwanitkit V. Type I diabetes mellitus in human and chimpanzee: a comparison of kyoto encyclopedia of genes and genomes pathway. Diabetes Technol Ther 2007; 9:145-8. [PMID: 17425439 DOI: 10.1089/dia.2006.0022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Diabetes is a worldwide medical problem and is a significant cause of morbidity and mortality. Type 1 diabetes results from the autoimmune destruction of insulin-producing beta cells in the pancreas. The identification of causative genes for the autoimmune disease type 1 diabetes in humans has made significant progress in recent years. Studies of pathways for type 1 diabetes in other living things can give useful information on the nature of type 1 diabetes. METHODS Here, the author used a new pathway technology to compare type 1 diabetes mellitus in the human and the chimpanzee. RESULTS According to the comparison, the mainframes of pathways are similar for both the human and the chimpanzee. CONCLUSIONS These results can imply a close relation between the human and the chimpanzee. They also confirm usage of the chimpanzee model for studies of type 1 diabetes pathophysiology.
Collapse
Affiliation(s)
- Viroj Wiwanitkit
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|