1
|
Saini T, Mazumder PM. Current advancement in the preclinical models used for the assessment of diabetic neuropathy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2727-2745. [PMID: 37987794 DOI: 10.1007/s00210-023-02802-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
Diabetic neuropathy is one of the prevalent and debilitating microvascular complications of diabetes mellitus, affecting a significant portion of the global population. Relational preclinical animal models are essential to understand its pathophysiology and develop effective treatments. This abstract provides an overview of current knowledge and advancements in such models. Various animal models have been developed to mimic the multifaceted aspects of human diabetic neuropathy, including both type 1 and type 2 diabetes. These models involve rodents (rats and mice) and larger animals like rabbits and dogs. Induction of diabetic neuropathy in these models is achieved through chemical, genetic, or dietary interventions, such as diabetogenic agents, genetic modifications, or high-fat diets. Preclinical animal models have greatly contributed to studying the intricate molecular and cellular mechanisms underlying diabetic neuropathy. They have shed light on hyperglycemia-induced oxidative stress, neuroinflammation, mitochondrial dysfunction, and altered neurotrophic factor signaling. Additionally, these models have allowed for the investigation of morphological changes, functional alterations, and behavioral manifestations associated with diabetic neuropathy. These models have also been crucial for evaluating the efficacy and safety of potential therapeutic interventions. Novel pharmacological agents, gene therapies, stem cell-based approaches, exercise, dietary modifications, and neurostimulation techniques have been tested using these models. However, limitations and challenges remain, including physiological differences between humans and animals, complex neuropathy phenotypes, and the need for translational validation. In conclusion, preclinical animal models have played a vital role in advancing our understanding and management of diabetic neuropathy. They have enhanced our knowledge of disease mechanisms, facilitated the development of novel treatments, and provided a platform for translational research. Ongoing efforts to refine and validate these models are crucial for future treatment developments for this debilitating condition.
Collapse
Affiliation(s)
- Tanishk Saini
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, 835215, Ranchi, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, 835215, Ranchi, India.
| |
Collapse
|
2
|
Singh R, Gholipourmalekabadi M, Shafikhani SH. Animal models for type 1 and type 2 diabetes: advantages and limitations. Front Endocrinol (Lausanne) 2024; 15:1359685. [PMID: 38444587 PMCID: PMC10912558 DOI: 10.3389/fendo.2024.1359685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
Diabetes mellitus, commonly referred to as diabetes, is a group of metabolic disorders characterized by chronic elevation in blood glucose levels, resulting from inadequate insulin production, defective cellular response to extracellular insulin, and/or impaired glucose metabolism. The two main types that account for most diabetics are type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM), each with their own pathophysiological features. T1D is an autoimmune condition where the body's immune system attacks and destroys the insulin-producing beta cells in the pancreas. This leads to lack of insulin, a vital hormone for regulating blood sugar levels and cellular glucose uptake. As a result, those with T1D depend on lifelong insulin therapy to control their blood glucose level. In contrast, T2DM is characterized by insulin resistance, where the body's cells do not respond effectively to insulin, coupled with a relative insulin deficiency. This form of diabetes is often associated with obesity, sedentary lifestyle, and/or genetic factors, and it is managed with lifestyle changes and oral medications. Animal models play a crucial role in diabetes research. However, given the distinct differences between T1DM and T2DM, it is imperative for researchers to employ specific animal models tailored to each condition for a better understanding of the impaired mechanisms underlying each condition, and for assessing the efficacy of new therapeutics. In this review, we discuss the distinct animal models used in type 1 and type 2 diabetes mellitus research and discuss their strengths and limitations.
Collapse
Affiliation(s)
- Raj Singh
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL, United States
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sasha H Shafikhani
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL, United States
- Cancer Center, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
3
|
Sadikan MZ, Abdul Nasir NA, Lambuk L, Mohamud R, Reshidan NH, Low E, Singar SA, Mohmad Sabere AS, Iezhitsa I, Agarwal R. Diabetic retinopathy: a comprehensive update on in vivo, in vitro and ex vivo experimental models. BMC Ophthalmol 2023; 23:421. [PMID: 37858128 PMCID: PMC10588156 DOI: 10.1186/s12886-023-03155-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023] Open
Abstract
Diabetic retinopathy (DR), one of the leading causes of visual impairment and blindness worldwide, is one of the major microvascular complications in diabetes mellitus (DM). Globally, DR prevalence among DM patients is 25%, and 6% have vision-threatening problems among them. With the higher incidence of DM globally, more DR cases are expected to be seen in the future. In order to comprehend the pathophysiological mechanism of DR in humans and discover potential novel substances for the treatment of DR, investigations are typically conducted using various experimental models. Among the experimental models, in vivo models have contributed significantly to understanding DR pathogenesis. There are several types of in vivo models for DR research, which include chemical-induced, surgical-induced, diet-induced, and genetic models. Similarly, for the in vitro models, there are several cell types that are utilised in DR research, such as retinal endothelial cells, Müller cells, and glial cells. With the advancement of DR research, it is essential to have a comprehensive update on the various experimental models utilised to mimic DR environment. This review provides the update on the in vitro, in vivo, and ex vivo models used in DR research, focusing on their features, advantages, and limitations.
Collapse
Affiliation(s)
- Muhammad Zulfiqah Sadikan
- Department of Pharmacology, Faculty of Medicine, Manipal University College Malaysia (MUCM), Bukit Baru, 75150, Melaka, Malaysia
| | - Nurul Alimah Abdul Nasir
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia.
| | - Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nur Hidayah Reshidan
- School of Biology, Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Evon Low
- Ageing Biology Centre, Newcastle University, NE1 7RU, Newcastle upon Tyne, UK
| | - Saiful Anuar Singar
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, 32306, Tallahassee, FL, USA
| | - Awis Sukarni Mohmad Sabere
- Kulliyyah of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Bandar Indera Mahkota, 25200, Kuantan, Pahang, Malaysia
| | - Igor Iezhitsa
- School of Medicine, International Medical University, 57000, Bukit Jalil, Kuala Lumpur, Malaysia
- Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Pavshikh Bortsov sq. 1, 400131 , Volgograd, Russian Federation
| | - Renu Agarwal
- School of Medicine, International Medical University, 57000, Bukit Jalil, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Rajlic S, Treede H, Münzel T, Daiber A, Duerr GD. Early Detection Is the Best Prevention-Characterization of Oxidative Stress in Diabetes Mellitus and Its Consequences on the Cardiovascular System. Cells 2023; 12:583. [PMID: 36831253 PMCID: PMC9954643 DOI: 10.3390/cells12040583] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Previous studies demonstrated an important role of oxidative stress in the pathogenesis of cardiovascular disease (CVD) in diabetic patients due to hyperglycemia. CVD remains the leading cause of premature death in the western world. Therefore, diabetes mellitus-associated oxidative stress and subsequent inflammation should be recognized at the earliest possible stage to start with the appropriate treatment before the onset of the cardiovascular sequelae such as arterial hypertension or coronary artery disease (CAD). The pathophysiology comprises increased reactive oxygen and nitrogen species (RONS) production by enzymatic and non-enzymatic sources, e.g., mitochondria, an uncoupled nitric oxide synthase, xanthine oxidase, and the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX). Considering that RONS originate from different cellular mechanisms in separate cellular compartments, adequate, sensitive, and compartment-specific methods for their quantification are crucial for early detection. In this review, we provide an overview of these methods with important information for early, appropriate, and effective treatment of these patients and their cardiovascular sequelae.
Collapse
Affiliation(s)
- Sanela Rajlic
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
| | - Hendrik Treede
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Department of Cardiology, Molecular Cardiology, University Medical Center, 55131 Mainz, Germany
| | - Andreas Daiber
- Center for Cardiology, Department of Cardiology, Molecular Cardiology, University Medical Center, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Georg Daniel Duerr
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
| |
Collapse
|
5
|
Nagao M, Asai A, Eliasson L, Oikawa S. Selectively bred rodent models for studying the etiology of type 2 diabetes: Goto-Kakizaki rats and Oikawa-Nagao mice. Endocr J 2023; 70:19-30. [PMID: 36477370 DOI: 10.1507/endocrj.ej22-0253] [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] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes (T2D) is a polygenic disease and studies to understand the etiology of the disease have required selectively bred animal models with polygenic background. In this review, we present two models; the Goto-Kakizaki (GK) rat and the Oikawa-Nagao Diabetes-Prone (ON-DP) and Diabetes-Resistant (ON-DR) mouse. The GK rat was developed by continuous selective breeding for glucose tolerance from the outbred Wistar rat around 50 years ago. The main cause of spontaneous hyperglycemia in this model is insulin secretion deficiency from pancreatic β-cells and mild insulin resistance in insulin target organs. A disadvantage of the GK rat is that environmental factors have not been considered in the selective breeding. Hence, the GK rat may not be suitable for elucidating predisposition to diabetes under certain environmental conditions, such as a high-fat diet. Therefore, we recently established two mouse lines with different susceptibilities to diet-induced diabetes, which are prone and resistant to the development of diabetes, designated as the ON-DP and ON-DR mouse, respectively. The two ON mouse lines were established by continuous selective breeding for inferior and superior glucose tolerance after high-fat diet feeding in hybrid mice of three inbred strains. Studies of phenotypic differences between ON-DP and ON-DR mice and their underlying molecular mechanisms will shed light on predisposing factors for the development of T2D in the modern obesogenic environment. This review summarizes the background and the phenotypic differences and similarities of GK rats and ON mice and highlights the advantages of using selectively bred rodent models in diabetes research.
Collapse
Affiliation(s)
- Mototsugu Nagao
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö 214 28, Sweden
- Clincal Research Centre (CRC), Skåne University Hospital(SUS), Malmö 214 28, Sweden
| | - Akira Asai
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö 214 28, Sweden
- Clincal Research Centre (CRC), Skåne University Hospital(SUS), Malmö 214 28, Sweden
| | - Shinichi Oikawa
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
6
|
Experimental animal models for diabetes and its related complications-a review. Lab Anim Res 2021; 37:23. [PMID: 34429169 PMCID: PMC8385906 DOI: 10.1186/s42826-021-00101-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetes mellitus, a very common and multifaceted metabolic disorder is considered as one of the fastest growing public health problems in the world. It is characterized by hyperglycemia, a condition with high glucose level in the blood plasma resulting from defects in insulin secretion or its action and in some cases both the impairment in secretion and also action of insulin coexist. Historically, animal models have played a critical role in exploring and describing malady pathophysiology and recognizable proof of targets and surveying new remedial specialists and in vivo medicines. In the present study, we reviewed the experimental models employed for diabetes and for its related complications. This paper reviews briefly the broad chemical induction of alloxan and streptozotocin and its mechanisms associated with type 1 and type 2 diabetes. Also we highlighted the different models in other species and other animals.
Collapse
|
7
|
Eckel RH, Bornfeldt KE, Goldberg IJ. Cardiovascular disease in diabetes, beyond glucose. Cell Metab 2021; 33:1519-1545. [PMID: 34289375 PMCID: PMC8411849 DOI: 10.1016/j.cmet.2021.07.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/21/2021] [Accepted: 07/01/2021] [Indexed: 02/06/2023]
Abstract
Despite the decades-old knowledge that diabetes mellitus is a major risk factor for cardiovascular disease, the reasons for this association are only partially understood. While this association is true for both type 1 and type 2 diabetes, different pathophysiological processes may be responsible. Lipids and other risk factors are indeed important, whereas the role of glucose is less clear. This lack of clarity stems from clinical trials that do not unambiguously show that intensive glycemic control reduces cardiovascular events. Animal models have provided mechanisms that link diabetes to increased atherosclerosis, and evidence consistent with the importance of factors beyond hyperglycemia has emerged. We review clinical, pathological, and animal studies exploring the pathogenesis of atherosclerosis in humans living with diabetes and in mouse models of diabetes. An increased effort to identify risk factors beyond glucose is now needed to prevent the increased cardiovascular disease risk associated with diabetes.
Collapse
Affiliation(s)
- Robert H Eckel
- Divisions of Endocrinology, Metabolism and Diabetes, and Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| | - Karin E Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, and Department of Laboratory Medicine and Pathology, University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
8
|
Zhao JD, Li Y, Sun M, Yu CJ, Li JY, Wang SH, Yang D, Guo CL, Du X, Zhang WJ, Cheng RD, Diao XC, Fang ZH. Effect of berberine on hyperglycaemia and gut microbiota composition in type 2 diabetic Goto-Kakizaki rats. World J Gastroenterol 2021; 27:708-724. [PMID: 33716449 PMCID: PMC7934002 DOI: 10.3748/wjg.v27.i8.708] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/17/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A recent investigation showed that the prevalence of type 2 diabetes mellitus (T2DM) is 12.8% among individuals of Han ethnicity. Gut microbiota has been reported to play a central role in T2DM. Goto-Kakizaki (GK) rats show differences in gut microbiota compared to non-diabetic rats. Previous studies have indicated that berberine could be successfully used to manage T2DM. We sought to understand its hypoglycaemic effect and role in the regulation of the gut microbiota.
AIM To determine whether berberine can regulate glucose metabolism in GK rats via the gut microbiota.
METHODS GK rats were acclimatized for 1 wk. The GK rats were randomly divided into three groups and administered saline (Mo), metformin (Me), or berberine (Be). The observation time was 8 wk, and weight, fasting blood glucose (FBG), insulin, and glucagon-like peptide-1 (GLP-1) were measured. Pancreatic tissue was observed for pathological changes. Additionally, we sequenced the 16S rRNA V3-V4 region of the gut microbiota and analysed the structure.
RESULTS Compared with the Mo group, the Me and Be groups displayed significant differences in FBG (P < 0.01) and GLP-1 (P < 0.05). A significant decrease in weight and homeostatic model assessment-insulin resistance was noted in the Be group compared with those in the Me group (P < 0.01). The pancreatic islets of the Me- and Be-treated rats showed improvement in number, shape, and necrosis compared with those of Mo-treated rats. A total of 580 operational taxonomic units were obtained in the three groups. Compared to the Mo group, the Me and Be groups showed a shift in the structure of the gut microbiota. Correlation analysis indicated that FBG was strongly positively correlated with Clostridia_UCG-014 (P < 0.01) and negatively correlated with Allobaculum (P < 0.01). Body weight showed a positive correlation with Desulfovibrionaceae (P < 0.01) and a negative correlation with Akkermansia (P < 0.01). Importantly, our results demonstrated that Me and Be could significantly decrease Bacteroidetes (P < 0.01) and the Bacteroidetes/Firmicutes ratio (P < 0.01). Furthermore, Muribaculaceae (P < 0.01; P < 0.05) was significantly decreased in the Me and Be groups, and Allobaculum (P < 0.01) was significantly increased.
CONCLUSION Berberine has a substantial effect in improving metabolic parameters and modulating the gut microbiota composition in T2DM rats.
Collapse
Affiliation(s)
- Jin-Dong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Yan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Min Sun
- School of Life Sciences, Anhui University, Hefei 230039, Anhui Province, China
| | - Chan-Juan Yu
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Jia-Yun Li
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Si-Hai Wang
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Di Yang
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Cheng-Lin Guo
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Xue Du
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Wen-Jin Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Ruo-Dong Cheng
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Xiao-Chuan Diao
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| | - Zhao-Hui Fang
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui Province, China
| |
Collapse
|
9
|
Effect of Postnatal Nutritional Environment Due to Maternal Diabetes on Beta Cell Mass Programming and Glucose Intolerance Risk in Male and Female Offspring. Biomolecules 2021; 11:biom11020179. [PMID: 33525575 PMCID: PMC7911592 DOI: 10.3390/biom11020179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 11/20/2022] Open
Abstract
Besides the fetal period, the suckling period is a critical time window in determining long-term metabolic health. We undertook the present study to elucidate the impact of a diabetic suckling environment alone or associated with an in utero diabetic environment on beta cell mass development and the risk of diabetes in the offspring in the long term. To that end, we have compared two experimental settings. In setting 1, we used Wistar (W) rat newborns resulting from W ovocytes (oW) transferred into diabetic GK rat mothers (pGK). These oW/pGK neonates were then suckled by diabetic GK foster mothers (oW/pGK/sGK model) and compared to oW/pW neonates suckled by normal W foster mothers (oW/pW/sW model). In setting 2, normal W rat newborns were suckled by diabetic GK rat foster mothers (nW/sGK model) or normal W foster mothers (nW/sW model). Our data revealed that the extent of metabolic disorders in term of glucose intolerance and beta cell mass are similar between rats which have been exposed to maternal diabetes both pre- and postnatally (oW/pGK/sGK model) and those which have been exposed only during postnatal life (nW/sW model). In other words, being nurtured by diabetic GK mothers from birth to weaning was sufficient to significantly alter the beta cell mass, glucose-induced insulin secretion and glucose homeostasis of offspring. No synergistic deleterious effects of pre-and postnatal exposure was observed in our setting.
Collapse
|
10
|
Nagao M, Esguerra JLS, Wendt A, Asai A, Sugihara H, Oikawa S, Eliasson L. Selectively Bred Diabetes Models: GK Rats, NSY Mice, and ON Mice. Methods Mol Biol 2020; 2128:25-54. [PMID: 32180184 DOI: 10.1007/978-1-0716-0385-7_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The polygenic background of selectively bred diabetes models mimics the etiology of type 2 diabetes. So far, three different rodent models (Goto-Kakizaki rats, Nagoya-Shibata-Yasuda mice, and Oikawa-Nagao mice) have been established in the diabetes research field by continuous selective breeding for glucose tolerance from outbred rodent stocks. The origin of hyperglycemia in these rodents is mainly insulin secretion deficiency from the pancreatic β-cells and mild insulin resistance in insulin target organs. In this chapter, we summarize backgrounds and phenotypes of these rodent models to highlight their importance in diabetes research. Then, we introduce experimental methodologies to evaluate β-cell exocytosis as a putative common defect observed in these rodent models.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/etiology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/etiology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Exocytosis
- Gene Expression Profiling/methods
- Glucose Intolerance
- Insulin Resistance/physiology
- Insulin Secretion/physiology
- Insulin-Secreting Cells/chemistry
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/physiology
- Mice
- Mice, Inbred C3H
- Patch-Clamp Techniques/methods
- Phenotype
- Rats
- Rats, Wistar
- Selective Breeding/genetics
Collapse
Affiliation(s)
- Mototsugu Nagao
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden.
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Jonathan Lou S Esguerra
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden
| | - Akira Asai
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hitoshi Sugihara
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shinichi Oikawa
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Diabetes and Lifestyle-related Disease Center, Japan Anti-Tuberculosis Association, Fukujuji Hospital, Tokyo, Japan
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
- Clinical Research Centre, Skåne University Hospital, Lund and Malmö, Sweden.
| |
Collapse
|
11
|
Movassat J, Delangre E, Liu J, Gu Y, Janel N. Hypothesis and Theory: Circulating Alzheimer's-Related Biomarkers in Type 2 Diabetes. Insight From the Goto-Kakizaki Rat. Front Neurol 2019; 10:649. [PMID: 31293498 PMCID: PMC6606723 DOI: 10.3389/fneur.2019.00649] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Epidemiological data suggest an increased risk of developing Alzheimer's disease (AD) in individuals with type 2 diabetes (T2D). AD is anatomically associated with an early progressive accumulation of Aβ leading to a gradual Tau hyperphosphorylation, which constitute the main characteristics of damaged brain in AD. Apart from these processes, mounting evidence suggests that specific features of diabetes, namely impaired glucose metabolism and insulin signaling in the brain, play a key role in AD. Moreover, several studies report a potential role of Aβ and Tau in peripheral tissues such as pancreatic β cells. Thus, it appears that several biological pathways associated with diabetes overlap with AD. The link between peripheral insulin resistance and brain insulin resistance with concomitant cognitive impairment may also potentially be mediated by a liver/pancreatic/brain axis, through the excessive trafficking of neurotoxic molecules across the blood-brain barrier. Insulin resistance incites inflammation and pro-inflammatory cytokine activation modulates the homocysteine cycle in T2D patients. Elevated plasma homocysteine level is a risk factor for AD pathology and is also closely associated with metabolic syndrome. We previously demonstrated a strong association between homocysteine metabolism and insulin via cystathionine beta synthase (CBS) activity, the enzyme implicated in the first step of the trans-sulfuration pathway, in Goto-Kakizaki (GK) rats, a spontaneous model of T2D, with close similarities with human T2D. CBS activity is also correlated with DYRK1A, a serine/threonine kinase regulating brain-derived neurotrophic factor (BDNF) levels, and Tau phosphorylation, which are implicated in a wide range of disease such as T2D and AD. We hypothesized that DYRK1A, BDNF, and Tau, could be among molecular factors linking T2D to AD. In this focused review, we briefly examine the main mechanisms linking AD to T2D and provide the first evidence that certain circulating AD biomarkers are found in diabetic GK rats. We propose that the spontaneous model of T2D in GK rat could be a suitable model to investigate molecular mechanisms linking T2D to AD.
Collapse
Affiliation(s)
- Jamileh Movassat
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - Etienne Delangre
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - Junjun Liu
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - YuChen Gu
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| | - Nathalie Janel
- Univ Paris Diderot-Sorbonne Paris Cité, Laboratoire Processus Dégénératifs, Stress et Vieillissement, Unité de Biologie Fonctionnelle et Adaptative (BFA), UMR 8251 CNRS, Paris, France
| |
Collapse
|
12
|
Camacho-Ramírez A, Almorza-Gomar D, Díaz-Gómez A, Mateo-Gavira I, Macias-Rodriguez M, Pérez-Arana GM, Prada-Oliveira JA. The histomorphometric parameters of endocrine pancreas after bariatric surgery in healthy animal models. Tissue Cell 2019; 57:78-83. [PMID: 30947967 DOI: 10.1016/j.tice.2019.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND The aim of this study is to describe in depth how different bariatric surgeries affect to the cellularity of β-cells in the pancreatic islet. There are much data regarding the possible physiological mechanisms involved in resolution of type 2 diabetes after bariatric surgery. But these data usually are controversial. We reported a direct influence of bariatric surgical technique on endocrine pancreas cellular turnover. Some surgeries increase proliferation processes of the β-cells. Our objective is to report the histomorphometric mechanism that these techniques stimulate over the cellularity of pancreatic islet. METHOD To this purpose, we used adult male Wistar rats to undergo the different techniques. We developed three surgical techniques (Sleeve gastrectomy and Y-Roux Gastric bypass as the most usual bariatric techniques, and a purely malabsorptive technique); moreover two control groups were performed (Sham and fasting controls). RESULTS We completed a sequence of morphometric studies to conclude the behaviour of endocrine pancreatic β-cell islet, correlating several histomorphometry parameters. CONCLUSION Our purpose was to show a comprehensive interpretation to the consequences that bariatric surgeries had on the pancreatic islets cellularity. Moreover, we included the main tests to report the cellularity in histological samples.
Collapse
Affiliation(s)
| | | | - Alfredo Díaz-Gómez
- Instituto para Investigación en Biomedicina (INIBICA). University of Cádiz, Avda. Ana de Viya, 21. Cádiz, 11009
| | - Isabel Mateo-Gavira
- Endocrinology and Nutrition Service, Universitary Hospital Puerta del Mar, Cádiz, 11010
| | | | - Gonzalo M Pérez-Arana
- Instituto para Investigación en Biomedicina (INIBICA). University of Cádiz, Avda. Ana de Viya, 21. Cádiz, 11009
| | - J Arturo Prada-Oliveira
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cádiz, Plaza Fragela s/n, Cádiz, 11003.
| |
Collapse
|
13
|
Kuwabara WMT, Panveloski-Costa AC, Yokota CNF, Pereira JNB, Filho JM, Torres RP, Hirabara SM, Curi R, Alba-Loureiro TC. Comparison of Goto-Kakizaki rats and high fat diet-induced obese rats: Are they reliable models to study Type 2 Diabetes mellitus? PLoS One 2017; 12:e0189622. [PMID: 29220408 PMCID: PMC5722336 DOI: 10.1371/journal.pone.0189622] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
Type 2 Diabetes mellitus (T2DM) is an evident growing disease that affects different cultures throughout the world. T2DM occurs under the influence of three main factors: the genetic background, environmental and behavioral components. Obesity is strongly associated to the development of T2DM in the occident, while in the orient most of the diabetic patients are considered lean. Genetics may be a key factor in the development of T2DM in societies where obesity is not a recurrent public health problem. Herein, two different models of rats were used to understand their differences and reliability as experimental models to study the pathophysiology of T2DM, in two different approaches: the genetic (GK rats) and the environmental (HFD-induced obese rats) influences. GK rats were resistant to weight gain even though food/energy consumption (relative to body weight) was higher in this group. HFD, on the other hand, induced obesity in Wistar rats. White adipose tissue (WAT) expansion in this group was accompanied by immune cells infiltration, inflammation and insulin resistance. GK rats also presented WAT inflammation and insulin resistance; however, no immune cells infiltration was observed in the WAT of this group. Liver of HFD group presented fat accumulation without differences in inflammatory cytokines content, while liver of GK rats didn't present fat accumulation, but showed an increase of IL-6 and IL-10 content and glycogen. Also, GK rats showed increased plasma GOT and GPT. Soleus muscle of HFD presented normal insulin signaling, contrary to GK rats, which presented higher content of basal phosphorylation of GSK-3β. Our results demonstrated that HFD developed a mild insulin resistance in Wistar rats, but was not sufficient to develop T2DM. In contrast, GK rats presented all the typical hallmarks of T2DM, such as insulin resistance, defective insulin production, fasting hyperglycemia/hyperinsulinemia and lipid plasma alteration. Thus, on the given time point of this study, we may conclude that only GK rats shown to be a reliable model to study T2DM.
Collapse
Affiliation(s)
| | - Ana Carolina Panveloski-Costa
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Joice Naiara Bertaglia Pereira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Cruzeiro do Sul University, São Paulo, Brazil
| | - Jorge Mancini Filho
- Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Cruzeiro do Sul University, São Paulo, Brazil
| | | |
Collapse
|
14
|
Špaček T, Pavluch V, Alán L, Capková N, Engstová H, Dlasková A, Berková Z, Saudek F, Ježek P. Nkx6.1 decline accompanies mitochondrial DNA reduction but subtle nucleoid size decrease in pancreatic islet β-cells of diabetic Goto Kakizaki rats. Sci Rep 2017; 7:15674. [PMID: 29142323 PMCID: PMC5688109 DOI: 10.1038/s41598-017-15958-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/04/2017] [Indexed: 01/12/2023] Open
Abstract
Hypertrophic pancreatic islets (PI) of Goto Kakizaki (GK) diabetic rats contain a lower number of β-cells vs. non-diabetic Wistar rat PI. Remaining β-cells contain reduced mitochondrial (mt) DNA per nucleus (copy number), probably due to declining mtDNA replication machinery, decreased mt biogenesis or enhanced mitophagy. We confirmed mtDNA copy number decrease down to <30% in PI of one-year-old GK rats. Studying relations to mt nucleoids sizes, we employed 3D superresolution fluorescent photoactivable localization microscopy (FPALM) with lentivirally transduced Eos conjugate of mt single-stranded-DNA-binding protein (mtSSB) or transcription factor TFAM; or by 3D immunocytochemistry. mtSSB (binding transcription or replication nucleoids) contoured "nucleoids" which were smaller by 25% (less diameters >150 nm) in GK β-cells. Eos-TFAM-visualized nucleoids, composed of 72% localized TFAM, were smaller by 10% (immunochemically by 3%). A theoretical ~70% decrease in cell nucleoid number (spatial density) was not observed, rejecting model of single mtDNA per nucleoid. The β-cell maintenance factor Nkx6.1 mRNA and protein were declining with age (>12-fold, 10 months) and decreasing with fasting hyperglycemia in GK rats, probably predetermining the impaired mtDNA replication (copy number decrease), while spatial expansion of mtDNA kept nucleoids with only smaller sizes than those containing much higher mtDNA in non-diabetic β-cells.
Collapse
Affiliation(s)
- Tomáš Špaček
- Department of Mitochondrial Physiology, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vojtěch Pavluch
- Department of Mitochondrial Physiology, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Lukáš Alán
- Department of Mitochondrial Physiology, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Nikola Capková
- Department of Mitochondrial Physiology, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hana Engstová
- Department of Mitochondrial Physiology, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Andrea Dlasková
- Department of Mitochondrial Physiology, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Zuzana Berková
- Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | - František Saudek
- Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
15
|
Candeias E, Duarte AI, Sebastião I, Fernandes MA, Plácido AI, Carvalho C, Correia S, Santos RX, Seiça R, Santos MS, Oliveira CR, Moreira PI. Middle-Aged Diabetic Females and Males Present Distinct Susceptibility to Alzheimer Disease-like Pathology. Mol Neurobiol 2016; 54:6471-6489. [PMID: 27730513 DOI: 10.1007/s12035-016-0155-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/22/2016] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is a highly concerning public health problem of the twenty-first century. Currently, it is estimated that T2D affects 422 million people worldwide with a rapidly increasing prevalence. During the past two decades, T2D has been widely shown to have a major impact in the brain. This, together with the cognitive decline and increased risk for dementia upon T2D, may arise from the complex interaction between normal brain aging and central insulin signaling dysfunction. Among the several features shared between T2D and some neurodegenerative disorders (e.g., Alzheimer disease (AD)), the impairment of insulin signaling may be a key link. However, these may also involve changes in sex hormones' function and metabolism, ultimately contributing to the different susceptibilities between females and males to some pathologies. For example, female sex has been pointed as a risk factor for AD, particularly after menopause. However, less is known on the underlying molecular mechanisms or even if these changes start during middle-age (perimenopause). From the above, we hypothesized that sex differentially affects hormone-mediated intracellular signaling pathways in T2D brain, ultimately modulating the risk for neurodegenerative conditions. We aimed to evaluate sex-associated alterations in estrogen/insulin-like growth factor-1 (IGF-1)/insulin-related signaling, oxidative stress markers, and AD-like hallmarks in middle-aged control and T2D rat brain cortices. We used brain cortices homogenates obtained from middle-aged (8-month-old) control Wistar and non-obese, spontaneously T2D Goto-Kakizaki (GK) male and female rats. Peripheral characterization of the animal models was done by standard biochemical analyses of blood, plasma, or serum. Steroid sex hormones, oxidative stress markers, and AD-like hallmarks were given by specific ELISA kits and colorimetric techniques, whereas the levels of intracellular signaling proteins were determined by Western blotting. Albeit the high levels of plasma estradiol and progesterone observed in middle-aged control females suggested that they were still under their reproductive phase, some gonadal dysfunction might be already occurring in T2D ones, hence, anticipating their menopause. Moreover, the higher blood and lower brain cholesterol levels in female rats suggested that its dysfunctional uptake into the brain cortex may also hamper peripheral estrogen uptake and/or its local brain steroidogenic metabolism. Despite the massive drop in IGF-1 levels in females' brains, particularly upon T2D, they might have developed some compensatory mechanisms towards the maintenance of estrogen, IGF-1, and insulin receptors function and of the subsequent Akt- and ERK1/2-mediated signaling. These may ultimately delay the deleterious AD-like brain changes (including oxidative damage to lipids and DNA, amyloidogenic processing of amyloid precursor protein and increased tau protein phosphorylation) associated with T2D and/or age (reproductive senescence) in female rats. By demonstrating that differential sex steroid hormone profiles/action may play a pivotal role in brain over T2D progression, the present study reinforces the need to establish sex-specific preventive and/or therapeutic approaches and an appropriate time window for the efficient treatment against T2D and AD.
Collapse
Affiliation(s)
- E Candeias
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - A I Duarte
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal.
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal.
| | - I Sebastião
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
| | - M A Fernandes
- Life Sciences Department, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal
- Instituto do Mar, Life Sciences Department, University of Coimbra, 3004-517, Coimbra, Portugal
| | - A I Plácido
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - C Carvalho
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - S Correia
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - R X Santos
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Life Sciences Department, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal
| | - R Seiça
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - M S Santos
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Instituto do Mar, Life Sciences Department, University of Coimbra, 3004-517, Coimbra, Portugal
| | - C R Oliveira
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - P I Moreira
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal.
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal.
| |
Collapse
|
16
|
Sárközy M, Szűcs G, Fekete V, Pipicz M, Éder K, Gáspár R, Sója A, Pipis J, Ferdinandy P, Csonka C, Csont T. Transcriptomic alterations in the heart of non-obese type 2 diabetic Goto-Kakizaki rats. Cardiovasc Diabetol 2016; 15:110. [PMID: 27496100 PMCID: PMC4975916 DOI: 10.1186/s12933-016-0424-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/14/2016] [Indexed: 12/22/2022] Open
Abstract
Background There is a spectacular rise in the global prevalence of type 2 diabetes mellitus (T2DM) due to the worldwide obesity epidemic. However, a significant proportion of T2DM patients are non-obese and they also have an increased risk of cardiovascular diseases. As the Goto-Kakizaki (GK) rat is a well-known model of non-obese T2DM, the goal of this study was to investigate the effect of non-obese T2DM on cardiac alterations of the transcriptome in GK rats. Methods Fasting blood glucose, serum insulin and cholesterol levels were measured at 7, 11, and 15 weeks of age in male GK and control rats. Oral glucose tolerance test and pancreatic insulin level measurements were performed at 11 weeks of age. At week 15, total RNA was isolated from the myocardium and assayed by rat oligonucleotide microarray for 41,012 genes, and then expression of selected genes was confirmed by qRT-PCR. Gene ontology and protein–protein network analyses were performed to demonstrate potentially characteristic gene alterations and key genes in non-obese T2DM. Results Fasting blood glucose, serum insulin and cholesterol levels were significantly increased, glucose tolerance and insulin sensitivity were significantly impaired in GK rats as compared to controls. In hearts of GK rats, 204 genes showed significant up-regulation and 303 genes showed down-regulation as compared to controls according to microarray analysis. Genes with significantly altered expression in the heart due to non-obese T2DM includes functional clusters of metabolism (e.g. Cyp2e1, Akr1b10), signal transduction (e.g. Dpp4, Stat3), receptors and ion channels (e.g. Sln, Chrng), membrane and structural proteins (e.g. Tnni1, Mylk2, Col8a1, Adam33), cell growth and differentiation (e.g. Gpc3, Jund), immune response (e.g. C3, C4a), and others (e.g. Lrp8, Msln, Klkc1, Epn3). Gene ontology analysis revealed several significantly enriched functional inter-relationships between genes influenced by non-obese T2DM. Protein–protein interaction analysis demonstrated that Stat is a potential key gene influenced by non-obese T2DM. Conclusions Non-obese T2DM alters cardiac gene expression profile. The altered genes may be involved in the development of cardiac pathologies and could be potential therapeutic targets in non-obese T2DM. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0424-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Márta Sárközy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Gergő Szűcs
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary.,Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Veronika Fekete
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Márton Pipicz
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Katalin Éder
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Renáta Gáspár
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Andrea Sója
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | | | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csaba Csonka
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Tamás Csont
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary.
| |
Collapse
|
17
|
Erchegyi J, Wang L, Gulyas J, Samant M, Perrin MH, Lewis K, Miller C, Vaughan J, Donaldson C, Fischer W, Low W, Yakabi S, Karasawa H, Taché Y, Rivier C, Rivier J. Characterization of Multisubstituted Corticotropin Releasing Factor (CRF) Peptide Antagonists (Astressins). J Med Chem 2016; 59:854-66. [PMID: 26789203 DOI: 10.1021/acs.jmedchem.5b00926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CRF mediates numerous stress-related endocrine, autonomic, metabolic, and behavioral responses. We present the synthesis and chemical and biological properties of astressin B analogues {cyclo(30-33)[D-Phe(12),Nle(21,38),C(α)MeLeu(27,40),Glu(30),Lys(33)]-acetyl-h/r-CRF(9-41)}. Out of 37 novel peptides, 17 (2, 4, 6-8, 10, 11, 16, 17, 27, 29, 30, 32-36) and 16 (3, 5, 9, 12-15, 18, 19, 22-26, 28, 31) had k(i) to CRF receptors in the high picomolar and low nanomole ranges, respectively. Peptides 1, 2, and 11 inhibited h/rCRF and urocortin 1-induced cAMP release from AtT20 and A7r5 cells. When Astressin C 2 was administered to adrenalectomized rats at 1.0 mg subcutaneously, it inhibited ACTH release for >7 d. Additional rat data based on the inhibitory effect of (2) on h/rCRF-induced stimulation of colonic secretory motor activity and urocortin 2-induced delayed gastric emptying also indicate a safe and long-lasting antagonistic effect. The overall properties of selected analogues may fulfill the criteria expected from clinical candidates.
Collapse
Affiliation(s)
- Judit Erchegyi
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Lixin Wang
- Department of Medicine, CURE/Digestive Diseases Center, Digestive Diseases Division, University of California at Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, United States
| | - Jozsef Gulyas
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Manoj Samant
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Marilyn H Perrin
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Kathy Lewis
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Charleen Miller
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Joan Vaughan
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Cynthia Donaldson
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Wolfgang Fischer
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - William Low
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Seiichi Yakabi
- Department of Medicine, CURE/Digestive Diseases Center, Digestive Diseases Division, University of California at Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, United States
| | - Hiroshi Karasawa
- Department of Medicine, CURE/Digestive Diseases Center, Digestive Diseases Division, University of California at Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, United States
| | - Yvette Taché
- Department of Medicine, CURE/Digestive Diseases Center, Digestive Diseases Division, University of California at Los Angeles, and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California 90073, United States
| | - Catherine Rivier
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Jean Rivier
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies , 10010 N. Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
18
|
Comparative Genome of GK and Wistar Rats Reveals Genetic Basis of Type 2 Diabetes. PLoS One 2015; 10:e0141859. [PMID: 26529237 PMCID: PMC4631338 DOI: 10.1371/journal.pone.0141859] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022] Open
Abstract
The Goto-Kakizaki (GK) rat, which has been developed by repeated inbreeding of glucose-intolerant Wistar rats, is the most widely studied rat model for Type 2 diabetes (T2D). However, the detailed genetic background of T2D phenotype in GK rats is still largely unknown. We report a survey of T2D susceptible variations based on high-quality whole genome sequencing of GK and Wistar rats, which have generated a list of GK-specific variations (228 structural variations, 2660 CNV amplification and 2834 CNV deletion, 1796 protein affecting SNVs or indels) by comparative genome analysis and identified 192 potential T2D-associated genes. The genes with variants are further refined with prior knowledge and public resource including variant polymorphism of rat strains, protein-protein interactions and differential gene expression. Finally we have identified 15 genetic mutant genes which include seven known T2D related genes (Tnfrsf1b, Scg5, Fgb, Sell, Dpp4, Icam1, and Pkd2l1) and eight high-confidence new candidate genes (Ldlr, Ccl2, Erbb3, Akr1b1, Pik3c2a, Cd5, Eef2k, and Cpd). Our result reveals that the T2D phenotype may be caused by the accumulation of multiple variations in GK rat, and that the mutated genes may affect biological functions including adipocytokine signaling, glycerolipid metabolism, PPAR signaling, T cell receptor signaling and insulin signaling pathways. We present the genomic difference between two closely related rat strains (GK and Wistar) and narrow down the scope of susceptible loci. It also requires further experimental study to understand and validate the relationship between our candidate variants and T2D phenotype. Our findings highlight the importance of sequenced-based comparative genomics for investigating disease susceptibility loci in inbreeding animal models.
Collapse
|
19
|
Radosinska J, Kurahara LH, Hiraishi K, Viczenczova C, Egan Benova T, Szeiffova Bacova B, Dosenko V, Navarova J, Obsitnik B, Imanaga I, Soukup T, Tribulova N. Modulation of cardiac connexin-43 by omega-3 fatty acid ethyl-ester supplementation demonstrated in spontaneously diabetic rats. Physiol Res 2015; 64:795-806. [PMID: 26447526 DOI: 10.33549/physiolres.933075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Previous data suggest that type 1 diabetes mellitus leads to the deterioration of myocardial intercellular communication mediated by connexin-43 (Cx43) channels. We therefore aimed to explore Cx43, PKC signaling and ultrastructure in non-treated and omega-3 fatty acid (omega-3) treated spontaneously diabetic Goto-Kakizaki (GK) rats considered as type 2 diabetes model. Four-week-old GK and non-diabetic Wistar-Clea rats were fed omega-3 (200 mg/kg/day) for 2 months and compared with untreated rats. Real-time PCR and immunoblotting were performed to determine Cx43, PKC-epsilon and PKC-delta expression. In situ Cx43 was examined by immunohistochemistry and subcellular alterations by electron microscopy. Omega-3 intake reduced blood glucose, triglycerides, and cholesterol in diabetic rats and this was associated with improved integrity of cardiomyocytes and capillaries in the heart. Myocardial Cx43 mRNA and protein levels were higher in diabetic versus non-diabetic rats and were further enhanced by omega-3. The ratio of phosphorylated (functional) to non-phosphorylated Cx43 was lower in diabetic compared to non-diabetic rats but was increased by omega-3, in part due to up-regulation of PKC-epsilon. In addition, pro-apoptotic PKC-delta expression was decreased. In conclusion, spontaneously diabetic rats at an early stage of disease benefit from omega-3 intake due to its hypoglycemic effect, upregulation of myocardial Cx43, and preservation of cardiovascular ultrastructure. These findings indicates that supplementation of omega-3 may be beneficial also in the management of diabetes in humans.
Collapse
Affiliation(s)
- J Radosinska
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovakia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Sleeve gastrectomy, but not duodenojejunostomy, preserves total beta-cell mass in Goto-Kakizaki rats evaluated by three-dimensional optical projection tomography. Surg Endosc 2015; 30:532-542. [DOI: 10.1007/s00464-015-4236-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 04/21/2015] [Indexed: 12/31/2022]
|
21
|
Alán L, Olejár T, Cahová M, Zelenka J, Berková Z, Smětáková M, Saudek F, Matěj R, Ježek P. Delta Cell Hyperplasia in Adult Goto-Kakizaki (GK/MolTac) Diabetic Rats. J Diabetes Res 2015; 2015:385395. [PMID: 26236746 PMCID: PMC4506919 DOI: 10.1155/2015/385395] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 05/22/2015] [Accepted: 05/31/2015] [Indexed: 01/23/2023] Open
Abstract
Reduced beta cell mass in pancreatic islets (PI) of Goto-Kakizaki (GK) rats is frequently observed in this diabetic model, but knowledge on delta cells is scarce. Aiming to compare delta cell physiology/pathology of GK to Wistar rats, we found that delta cell number increased over time as did somatostatin mRNA and delta cells distribution in PI is different in GK rats. Subtle changes in 6-week-old GK rats were found. With maturation and aging of GK rats, disturbed cytoarchitecture occurred with irregular beta cells accompanied by delta cell hyperplasia and loss of pancreatic polypeptide (PPY) positivity. Unlike the constant glucose-stimulation index for insulin PI release in Wistar rats, this index declined with GK age, whereas for somatostatin it increased with age. A decrease of GK rat PPY serum levels was found. GK rat body weight decreased with increasing hyperglycemia. Somatostatin analog octreotide completely blocked insulin secretion, impaired proliferation at low autocrine insulin, and decreased PPY secretion and mitochondrial DNA in INS-1E cells. In conclusion, in GK rats PI, significant local delta cell hyperplasia and suspected paracrine effect of somatostatin diminish beta cell viability and contribute to the deterioration of beta cell mass. Altered PPY-secreting cells distribution amends another component of GK PI's pathophysiology.
Collapse
Affiliation(s)
- Lukáš Alán
- Department No. 75, Institute of Physiology, Academy of Sciences, 14220 Prague, Czech Republic
| | - Tomáš Olejár
- Department No. 75, Institute of Physiology, Academy of Sciences, 14220 Prague, Czech Republic
| | - Monika Cahová
- Institute of Clinical & Experimental Medicine, 14021 Prague, Czech Republic
| | - Jaroslav Zelenka
- Department No. 75, Institute of Physiology, Academy of Sciences, 14220 Prague, Czech Republic
| | - Zuzana Berková
- Institute of Clinical & Experimental Medicine, 14021 Prague, Czech Republic
| | - Magdalena Smětáková
- Teaching Thomayer Hospital and Third Medical School, Charles University, 14059 Prague, Czech Republic
| | - František Saudek
- Institute of Clinical & Experimental Medicine, 14021 Prague, Czech Republic
| | - Radoslav Matěj
- Teaching Thomayer Hospital and Third Medical School, Charles University, 14059 Prague, Czech Republic
| | - Petr Ježek
- Department No. 75, Institute of Physiology, Academy of Sciences, 14220 Prague, Czech Republic
- *Petr Ježek:
| |
Collapse
|
22
|
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
|
23
|
Portha B, Giroix MH, Tourrel-Cuzin C, Le-Stunff H, Movassat J. The GK rat: a prototype for the study of non-overweight type 2 diabetes. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2012; 933:125-59. [PMID: 22893405 DOI: 10.1007/978-1-62703-068-7_9] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus (T2D) arises when the endocrine pancreas fails to secrete sufficient insulin to cope with the metabolic demand because of β-cell secretory dysfunction and/or decreased β-cell mass. Defining the nature of the pancreatic islet defects present in T2D has been difficult, in part because human islets are inaccessible for direct study. This review is aimed to illustrate to what extent the Goto Kakizaki rat, one of the best characterized animal models of spontaneous T2D, has proved to be a valuable tool offering sufficient commonalities to study this aspect. A comprehensive compendium of the multiple functional GK abnormalities so far identified is proposed in this perspective, together with their time-course and interactions. A special focus is given toward the pathogenesis of defective β-cell number and function in the GK model. It is proposed that the development of T2D in the GK model results from the complex interaction of multiple events: (1) several susceptibility loci containing genes responsible for some diabetic traits; (2) gestational metabolic impairment inducing an epigenetic programming of the offspring pancreas and the major insulin target tissues; and (3) environmentally induced loss of β-cell differentiation due to chronic exposure to hyperglycemia/hyperlipidemia, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Bernard Portha
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), Université Paris-Diderot, CNRS EAC 4413, Paris, France.
| | | | | | | | | |
Collapse
|
24
|
Effects of Roux-en-Y operations on glucose homeostasis in obese GK rats. Surg Endosc 2011; 25:3493-8. [PMID: 21853396 DOI: 10.1007/s00464-011-1717-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Accepted: 02/20/2011] [Indexed: 10/17/2022]
Abstract
AIM To investigate the therapeutic effects of different styles of gastric bypass surgery on type 2 diabetes mellitus (T2DM) GK rats. METHODS Twenty 6-8-week-old male GK rats were randomly divided into four groups: group A was operated by Roux-en-Y gastrojejunostomy with duodenum exclusion and stomach capacity maintenance, group B was operated by loop-type gastrojejunostomy with duodenum exclusion and stomach capacity maintenance, group C was operated by Roux-en-Y gastrojejunostomy with partial gastrectomy, and group D was operated by loop-type gastrojejunostomy with partial gastrectomy. Changes of fasting blood glucose, oral glucose tolerance test (OGTT), and insulin tolerance test (ITT) in different operations were detected. RESULTS The operations exerted good effects on controlling blood glucose in groups A, B, C, and D. There was no significant difference between groups A and C (P > 0.05) or between groups B and D (P > 0.05), while operations in groups A and C were more effective than groups B and D (P < 0.05). On the 21st day after surgery, OGTT in animals of groups A and C was significantly improved, as indicated by a 34% reduction in the area under the curve (AUC) for blood glucose (P < 0.05 versus groups B and D); pregavage insulin levels (ng/ml) were significantly decreased in groups A and C (P < 0.05 versus groups B and D). The insulin tolerance test (ITT) confirmed impaired insulin sensitivity in groups B and D, compared with groups A and C. CONCLUSIONS Gastric bypass surgery might be effective to treat type 2 diabetes mellitus (T2DM), and Roux-en-Y gastrojejunostomy might be more effective than other operative styles.
Collapse
|
25
|
Whole grain consumption has a modest effect on the development of diabetes in the Goto-Kakisaki rat. Br J Nutr 2011; 107:192-201. [PMID: 21733309 DOI: 10.1017/s0007114511002741] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Epidemiological evidence suggests that whole grain intake is associated with reduced risk of type 2 diabetes. However, studies of individual whole grains on the prevention of type 2 diabetes are lacking. The objective of the present study was to examine the effect of different whole grains on type 2 diabetes in an animal model of type 2 diabetes, the Goto-Kakisaki (GK) rat. GK rats were fed either a basal diet or a whole grain-containing diet for 5 months. Whole grain diets contained 65 % whole grain flours of wheat, barley, oats or maize. After 2 months of feeding, fasting plasma glucose concentrations were lower in the wheat, barley and oats groups, compared with the basal group, whereas glycated Hb was significantly greater in the wheat group compared with other groups. Feeding of whole barley and maize increased plasma C-peptide concentrations compared with whole wheat at 2 months. There was a trend in the improvement of insulin resistance with a consumption of barley and oats diets at 2 months (P = 0·06) compared with the basal diet. Oxidative stress markers, urinary thiobarbituric acid-reactive substances and 8-isoprostane, did not improve with whole grain intake at 2 months. At 5 months, whole grain diets did not differ from the basal diet in glycaemic control, insulin secretion, oxidative stress and preservation of pancreatic β-cell mass. These results suggest that the consumption of whole grains may offer modest benefit early in the development of type 2 diabetes, but this benefit is lost with further development of the disease.
Collapse
|
26
|
Shen L, Keenan MJ, Raggio A, Williams C, Martin RJ. Dietary-resistant starch improves maternal glycemic control in Goto-Kakizaki rat. Mol Nutr Food Res 2011; 55:1499-508. [PMID: 21638778 DOI: 10.1002/mnfr.201000605] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 02/23/2011] [Accepted: 03/17/2011] [Indexed: 12/27/2022]
Abstract
SCOPE Dietary prebiotics show potential in anti-diabetes. Dietary resistant starch (RS) has a favorable impact on gut hormone profiles, including glucagon-like peptide-1 (GLP-1) consistently released, a potent anti-diabetic incretin. Also RS reduced body fat and improved glucose tolerance in rats and mice. In the current project, we hypothesize that dietary-resistant starch can improve insulin sensitivity and pancreatic β cell mass in a type 2 diabetic rat model. Altered gut fermentation and microbiota are the initial mechanisms, and enhancement in serum GLP-1 is the secondary mechanism. METHODS AND RESULTS In this study, GK rats were fed an RS diet with 30% RS and an energy control diet. After 10 wk, these rats were mated and went through pregnancy and lactation. At the end of the study, pancreatic β cell mass, insulin sensitivity, pancreatic insulin content, total GLP-1 levels, cecal short-chain fatty acid concentrations and butyrate producing bacteria in cecal contents were greatly improved by RS feeding. The offspring of RS-fed dams showed improved fasting glucose levels and normal growth curves. CONCLUSION Dietary RS is potentially of great therapeutic importance in the treatment of diabetes and improvement in outcomes of pregnancy complicated by diabetes.
Collapse
Affiliation(s)
- Li Shen
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| | | | | | | | | |
Collapse
|
27
|
Gao W, Bihorel S, DuBois DC, Almon RR, Jusko WJ. Mechanism-based disease progression modeling of type 2 diabetes in Goto-Kakizaki rats. J Pharmacokinet Pharmacodyn 2010; 38:143-62. [PMID: 21127951 DOI: 10.1007/s10928-010-9182-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 11/04/2010] [Indexed: 11/25/2022]
Abstract
The dynamics of aging and type 2 diabetes (T2D) disease progression were investigated in normal [Wistar-Kyoto (WKY)] and diabetic [Goto-Kakizaki (GK)] rats and a mechanistic disease progression model was developed for glucose, insulin, and glycosylated hemoglobin (HbA1c) changes over time. The study included 30 WKY and 30 GK rats. Plasma glucose and insulin, blood glucose and HbA1c concentrations and hematological measurements were taken at ages 4, 8, 12, 16 and 20 weeks. A mathematical model described the development of insulin resistance (IR) and β-cell function with age/growth and diabetes progression. The model utilized transit compartments and an indirect response model to quantitate biomarker changes over time. Glucose, insulin and HbA1c concentrations in WKY rats increased to a steady-state at 8 weeks due to developmental changes. Glucose concentrations at 4 weeks in GK rats were almost twice those of controls, and increased to a steady-state after 8 weeks. Insulin concentrations at 4 weeks in GK rats were similar to controls, and then hyperinsulinemia occurred until 12-16 weeks of age indicating IR. Subsequently, insulin concentrations in GK rats declined to slightly below WKY controls due to β-cell failure. HbA1c showed a delayed increase relative to glucose. Modeling of HbA1c was complicated by age-related changes in hematology in rats. The diabetes model quantitatively described the glucose/insulin inter-regulation and HbA1c production and reflected the underlying pathogenic factors of T2D--IR and β-cell dysfunction. The model could be extended to incorporate other biomarkers and effects of various anti-diabetic drugs.
Collapse
Affiliation(s)
- Wei Gao
- Department of Pharmaceutical Sciences, State University of New York, 565 Hochstetter Hall, Buffalo, NY, USA
| | | | | | | | | |
Collapse
|
28
|
Analyses of copy number variation of GK rat reveal new putative type 2 diabetes susceptibility loci. PLoS One 2010; 5:e14077. [PMID: 21124896 PMCID: PMC2990713 DOI: 10.1371/journal.pone.0014077] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 10/31/2010] [Indexed: 01/15/2023] Open
Abstract
Large efforts have been taken to search for genes responsible for type 2 diabetes (T2D), but have resulted in only about 20 in humans due to its complexity and heterogeneity. The GK rat, a spontanous T2D model, offers us a superior opportunity to search for more diabetic genes. Utilizing array comparative genome hybridization (aCGH) technology, we identifed 137 non-redundant copy number variation (CNV) regions from the GK rats when using normal Wistar rats as control. These CNV regions (CNVRs) covered approximately 36 Mb nucleotides, accounting for about 1% of the whole genome. By integrating information from gene annotations and disease knowledge, we investigated the CNVRs comprehensively for mining new T2D genes. As a result, we prioritized 16 putative protein-coding genes and two microRNA genes (rno-mir-30b and rno-mir-30d) as good candidates. The catalogue of CNVRs between GK and Wistar rats identified in this work served as a repository for mining genes that might play roles in the pathogenesis of T2D. Moreover, our efforts in utilizing bioinformatics methods to prioritize good candidate genes provided a more specific set of putative candidates. These findings would contribute to the research into the genetic basis of T2D, and thus shed light on its pathogenesis.
Collapse
|
29
|
Lacraz G, Figeac F, Movassat J, Kassis N, Portha B. Diabetic GK/Par rat beta-cells are spontaneously protected against H2O2-triggered apoptosis. A cAMP-dependent adaptive response. Am J Physiol Endocrinol Metab 2010; 298:E17-27. [PMID: 19843875 DOI: 10.1152/ajpendo.90871.2008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The alteration of the beta-cell population in the Goto-Kakizaki rat (GK/Par line), a model of spontaneous type 2 diabetes, has been ascribed to significantly decreased beta-cell replication and neogenesis, while beta-cell apoptosis is surprisingly not enhanced and remains in the normal range. To gain insight into the mechanisms by which those beta-cells are protected from death, we studied ex vivo the apoptotic activity and the expression of a large set of pro/antiapoptotic and pro/antioxidant genes in GK/Par islet cells. This was done in vitro in freshly isolated islets as well as in response to culture conditions and calibrated reactive oxygen species (ROS) exposure (i.e., H2O2). We also investigated the intracellular mechanisms of the diabetic beta-cell response to ROS, the role if any of the intracellular cAMP metabolism, and finally the kinetic of ROS response, taking advantage of the GK/Par rat normoglycemia until weaning. Our results show that the peculiar GK/Par beta-cell phenotype was correlated with an increased expression of a large panel of antioxidant genes as well as pro/antiapoptotic genes. We demonstrate that such combination confers resistance to cytotoxic H2O2 exposure in vitro, raising the possibility that at least some of the activated stress/defense genes have protective effects against H2O2-triggered beta-cell death. We also present some evidence that the GK/Par beta-cell resistance to H2O2 is at least partly cAMP dependent. Finally, we show that such a phenotype is not innate but is spontaneously acquired after diabetes onset as the result of an adaptive response to the diabetic environment.
Collapse
MESH Headings
- Adaptation, Physiological/physiology
- Animals
- Apoptosis/drug effects
- Apoptosis/physiology
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Cell Division/physiology
- Cells, Cultured
- Cyclic AMP/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Disease Models, Animal
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Hydrogen Peroxide/pharmacology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Male
- Oxidants/pharmacology
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Mutant Strains
- Rats, Wistar
- Reactive Oxygen Species/metabolism
Collapse
Affiliation(s)
- Grégory Lacraz
- Laboratoire Biologie et Pathologie du Pancréas Endocrine, Unité Biologie Fonctionnelle et Adaptive, Equipe 1, Université Paris-Diderot et CNRS EAC-4413, Paris, France
| | | | | | | | | |
Collapse
|
30
|
Portha B, Lacraz G, Chavey A, Figeac F, Fradet M, Tourrel-Cuzin C, Homo-Delarche F, Giroix MH, Bailbé D, Gangnerau MN, Movassat J. Islet structure and function in the GK rat. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:479-500. [PMID: 20217511 DOI: 10.1007/978-90-481-3271-3_21] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Type 2 diabetes mellitus (T2D) arises when the endocrine pancreas fails to secrete sufficient insulin to cope with the metabolic demand because of beta-cell secretory dysfunction and/or decreased beta-cell mass. Defining the nature of the pancreatic islet defects present in T2D has been difficult, in part because human islets are inaccessible for direct study. This review is aimed to illustrate to what extent the Goto-Kakizaki rat, one of the best characterized animal models of spontaneous T2D, has proved to be a valuable tool offering sufficient commonalities to study this aspect. A comprehensive compendium of the multiple functional GK islet abnormalities so far identified is proposed in this perspective. The pathogenesis of defective beta-cell number and function in the GK model is also discussed. It is proposed that the development of T2D in the GK model results from the complex interaction of multiple events: (i) several susceptibility loci containing genes responsible for some diabetic traits (distinct loci encoding impairment of beta-cell metabolism and insulin exocytosis, but no quantitative trait locus for decreased beta-cell mass); (ii) gestational metabolic impairment inducing an epigenetic programming of the offspring pancreas (decreased beta-cell neogenesis and proliferation) transmitted over generations; and (iii) loss of beta-cell differentiation related to chronic exposure to hyperglycaemia/hyperlipidaemia, islet inflammation, islet oxidative stress, islet fibrosis and perturbed islet vasculature.
Collapse
Affiliation(s)
- Bernard Portha
- Laboratoire B2PE, Unité BFA, Université Paris-Diderot et CNRS EAC4413, F - 75205 Paris Cedex13, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fernández-Millán E, Gangnerau MN, De Miguel-Santos L, Calderari S, Serradas P, Escrivá F, Portha B, Alvarez C. Undernutrition of the GK rat during gestation improves pancreatic IGF-2 and beta-cell mass in the fetuses. Growth Factors 2009; 27:409-18. [PMID: 19919529 DOI: 10.3109/08977190903199074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Goto-Kakizaki (GK) rat is a type 2 diabetes model with a defective beta-cell mass detectable in late fetal development. Diminished IGF-2 production seems to be involved in this effect. Herein, we analyzed the effect of maternal food-restriction on the beta-cell mass of GK fetuses and the involvement of the IGF system, highly responsive to nutritional status in this process. To this end, in undernourished GK fetuses (U-GK), we measured serum GH/IGF levels, beta-cell mass, replication and differentiation, and IGF-1/-2 protein content in liver and pancreas tissue. Pregnant GK females were food restricted (65% restriction) during the last week of gestation. Our results show that maternal malnutrition ameliorates beta-cell mass in U-GK fetuses and a specific pancreatic IGF-2 increase may be instrumental in this effect. Further studies are needed to determine whether maternal undernutrition is sufficient to delay or decrease the risk of the GK rat for developing diabetes.
Collapse
|
32
|
The utility of [(11)C] dihydrotetrabenazine positron emission tomography scanning in assessing beta-cell performance after sleeve gastrectomy and duodenal-jejunal bypass. Surgery 2009; 147:303-9. [PMID: 19828168 DOI: 10.1016/j.surg.2009.08.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 08/20/2009] [Indexed: 01/06/2023]
Abstract
BACKGROUND The aim of this study was to evaluate the effect of sleeve gastrectomy (SG) and duodenal-jejunal bypass (DJB) on glucose homeostasis and to evaluate the utility of positron emission tomography (PET) scanning for assessing beta-cell mass. METHODS Goto-Kakizaki rats were divided into 4 groups: control, sham, SG, or DJB. Oral glucose tolerance, insulin, and glucagon-like peptide-1 (GLP-1) were measured before and after surgery. Before and 90 days after treatment, [(11)C] DTBZ micro PET scanning was performed. RESULTS The control and sham animals gained more weight compared with SG and DJB animals (P < or = .05). Compared with control animals, the glucose area under the curve was lower in DJB animals 30 and 45 days after operations (P < or = .05). At killing, GLP-1 levels were greater in the DJB group compared with sham and SG (P < or = .05), whereas insulin levels were greater in both DJB and SG compared with sham (P < or = .05). With PET scanning, the 90-day posttreatment mean vesicular monoamine transporter type 2 binding index was greatest in the DJB animals (2.45) compared with SG (1.17), both of which were greater than baseline control animals (0.81). CONCLUSION In type 2 diabetic rodents, DJB leads to improved glucose homeostasis and an increase in VMAT2 density as measured by PET scanning.
Collapse
|
33
|
Duodenal-jejunal exclusion improves glucose tolerance in the diabetic, Goto-Kakizaki rat by a GLP-1 receptor-mediated mechanism. J Gastrointest Surg 2009; 13:1762-72. [PMID: 19488823 DOI: 10.1007/s11605-009-0912-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 04/15/2009] [Indexed: 01/31/2023]
Abstract
BACKGROUND Gastric bypass results in the rapid resolution of type 2 diabetes. No causal evidence exists to link specific gut hormone changes with improvements in glucose homeostasis post-operatively. We hypothesized that surgical augmentation of the glucoregulatory factor GLP-1 would improve glucose tolerance in diabetic GK rats. We compared two procedures that increase distal small bowel stimulation, ileal interposition (IT), and duodenal-jejunal exclusion (DJE). METHODS DJE, IT, DJE Sham, or IT Sham were performed in GK rats. Glucose tolerance was tested at 4 and 6 weeks, the latter with and without Exendin-[9-39], a GLP-1 receptor antagonist. Small bowel segments were harvested for GLP-1 protein content 2 weeks after DJE or Sham surgery. RESULTS Despite similar weight profiles, a significant improvement in the OGTT was noted at 4 weeks after DJE and IT. Plasma GLP-1 levels were significantly elevated after DJE and IT. Intestinal GLP-1 was increased in the mid-jejunum and ileum after DJE. Exendin-[9-39] abolished the improvement in glucose tolerance after DJE. CONCLUSIONS DJE increased GLP-1 secretion and improved glucose tolerance, an effect that was reversed by GLP-1 receptor antagonism. This study provides direct evidence that improvement of glucose tolerance following a gastric bypass-like surgery is mediated by enhanced GLP-1 action.
Collapse
|
34
|
Kindel TL, Yang Q, Yoder SM, Tso P. Nutrient-driven incretin secretion into intestinal lymph is different between diabetic Goto-Kakizaki rats and Wistar rats. Am J Physiol Gastrointest Liver Physiol 2009; 296:G168-74. [PMID: 19056762 PMCID: PMC2643916 DOI: 10.1152/ajpgi.90506.2008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The incretin hormones gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) augment postprandial glucose-mediated insulin release from pancreatic beta-cells. The Goto-Kakizaki (GK) rat is a widely used, lean rodent model of Type 2 diabetes; however, little is known regarding the incretin secretion profile to different nutrients in these rats. We have recently shown that lymph is a sensitive medium to measure incretin secretion in rodents and probably the preferred compartment for GLP-1 monitoring. To characterize the meal-induced incretin profile, we compared lymphatic incretin concentrations in the GK and Wistar rat after enteral macronutrient administration. After cannulation of the major mesenteric lymphatic duct and duodenum, each animal received an intraduodenal bolus of either a fat emulsion, dextrin, a mixed meal, or saline. Lymph was collected for 3 h and analyzed for triglyceride, glucose, GLP-1, and GIP content. There was no statistical difference in GIP or GLP-1 secretion after a lipid bolus between GK and Wistar rats. Dextrin and a mixed meal both increased incretin concentration area under the curve, however, significantly less in GK rats compared with Wistar rats (dextrin GIP: 707 +/- 106 vs. 1,373 +/- 114 pg ml(-1) h, respectively, P < 0.001; dextrin GLP-1: 82.7 +/- 24.3 vs. 208.3 +/- 26.3 pM/h, respectively, P = 0.001). After administration of a carbohydrate-containing meal, GK rats were unable to mount as robust a response of both GIP and GLP-1 compared with Wistar rats, a phenomenon not seen after a lipid meal. We propose a similar, glucose-mediated incretin secretion pathway defect of both K and L cells in GK rats.
Collapse
Affiliation(s)
- Tammy L. Kindel
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Qing Yang
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Stephanie M. Yoder
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
35
|
Abstract
The ability of pancreatic beta-cell mass to vary according to insulin requirements is an important component of optimal long-term control of glucose homeostasis. It is generally assumed that alteration of this property largely contributes to the impairment of insulin secretion in type 2 diabetes. However, data in humans are scarce and it is impossible to correlate beta-cell mass and function with the various stages of the disease. Thus, the importance of animal models is obvious. In rodents, increased beta-cell mass associated with an increase in the function of individual beta-cells contributes to the adaptation of the insulin response to insulin resistance in late pregnancy and in obesity. A reduction in beta-cell mass always corresponds to an alteration in insulin secretory capacity of islet tissue (Zucker diabetic fatty and Goto-Kakisaki rats, db/db mice). During regenerative processes following experimental reduction of beta-cell mass [partial pancreatectomy, streptozocin (STZ) injection], beta-cell mass increase is not associated with a corresponding improvement of beta-cell function, thus indicating that regenerative beta-cells did not achieve functional maturity. The main lesson from experimental diabetes is therefore that beta-cell mass cannot always predict functional capacity of the beta-cell tissue and that the functional beta-cell mass rather than the anatomical beta-cell mass must be taken into account at all times.
Collapse
Affiliation(s)
- C Kargar
- Diabetes and Metabolic Diseases Research Department, Institut de Recherches Servier, Suresnes, France.
| | | |
Collapse
|