Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Anim Reprod ; 17(3): e20200064, 2020 Aug 26.
Article in English | MEDLINE | ID: mdl-33029223

ABSTRACT

The global prevalence of diabetes mellitus and other metabolic diseases is rapidly increasing. Animal models play pivotal roles in unravelling disease mechanisms and developing and testing therapeutic strategies. Rodents are the most widely used animal models but may have limitations in their resemblance to human disease mechanisms and phenotypes. Findings in rodent models are consequently often difficult to extrapolate to human clinical trials. To overcome this 'translational gap', we and other groups are developing porcine disease models. Pigs share many anatomical and physiological traits with humans and thus hold great promise as translational animal models. Importantly, the toolbox for genetic engineering of pigs is rapidly expanding. Human disease mechanisms and targets can therefore be reproduced in pigs on a molecular level, resulting in precise and predictive porcine (PPP) models. In this short review, we summarize our work on the development of genetically (pre)diabetic pig models and how they have been used to study disease mechanisms and test therapeutic strategies. This includes the generation of reporter pigs for studying beta-cell maturation and physiology. Furthermore, genetically engineered pigs are promising donors of pancreatic islets for xenotransplantation. In summary, genetically tailored pig models have become an important link in the chain of translational diabetes and metabolic research.

2.
Dis Model Mech ; 12(8)2019 08 12.
Article in English | MEDLINE | ID: mdl-31308048

ABSTRACT

Alongside the obesity epidemic, the prevalence of maternal diabetes is rising worldwide, and adverse effects on fetal development and metabolic disturbances in the offspring's later life have been described. To clarify whether metabolic programming effects are due to mild maternal hyperglycemia without confounding obesity, we investigated wild-type offspring of INSC93S transgenic pigs, which are a novel genetically modified large-animal model expressing mutant insulin (INS) C93S in pancreatic ß-cells. This mutation results in impaired glucose tolerance, mild fasting hyperglycemia and insulin resistance during late pregnancy. Compared with offspring from wild-type sows, piglets from hyperglycemic mothers showed impaired glucose tolerance and insulin resistance (homeostatic model assessment of insulin resistance: +3-fold in males; +4.4-fold in females) prior to colostrum uptake. Targeted metabolomics in the fasting and insulin-stimulated state revealed distinct alterations in the plasma metabolic profile of piglets from hyperglycemic mothers. They showed increased levels of acylcarnitines, gluconeogenic precursors such as alanine, phospholipids (in particular lyso-phosphatidylcholines) and α-aminoadipic acid, a potential biomarker for type 2 diabetes. These observations indicate that mild gestational hyperglycemia can cause impaired glucose tolerance, insulin resistance and associated metabolic alterations in neonatal offspring of a large-animal model born at a developmental maturation status comparable to human babies.


Subject(s)
Glucose Intolerance/etiology , Hyperglycemia/etiology , Insulin/genetics , Prenatal Exposure Delayed Effects/pathology , Animals , Animals, Genetically Modified , Animals, Newborn , Female , Insulin Secretion , Insulin-Secreting Cells/metabolism , Pregnancy , Swine
3.
Mol Metab ; 6(8): 931-940, 2017 08.
Article in English | MEDLINE | ID: mdl-28752056

ABSTRACT

OBJECTIVE: The prevalence of diabetes mellitus and associated complications is steadily increasing. As a resource for studying systemic consequences of chronic insulin insufficiency and hyperglycemia, we established a comprehensive biobank of long-term diabetic INSC94Y transgenic pigs, a model of mutant INS gene-induced diabetes of youth (MIDY), and of wild-type (WT) littermates. METHODS: Female MIDY pigs (n = 4) were maintained with suboptimal insulin treatment for 2 years, together with female WT littermates (n = 5). Plasma insulin, C-peptide and glucagon levels were regularly determined using specific immunoassays. In addition, clinical chemical, targeted metabolomics, and lipidomics analyses were performed. At age 2 years, all pigs were euthanized, necropsied, and a broad spectrum of tissues was taken by systematic uniform random sampling procedures. Total beta cell volume was determined by stereological methods. A pilot proteome analysis of pancreas, liver, and kidney cortex was performed by label free proteomics. RESULTS: MIDY pigs had elevated fasting plasma glucose and fructosamine concentrations, C-peptide levels that decreased with age and were undetectable at 2 years, and an 82% reduced total beta cell volume compared to WT. Plasma glucagon and beta hydroxybutyrate levels of MIDY pigs were chronically elevated, reflecting hallmarks of poorly controlled diabetes in humans. In total, ∼1900 samples of different body fluids (blood, serum, plasma, urine, cerebrospinal fluid, and synovial fluid) as well as ∼17,000 samples from ∼50 different tissues and organs were preserved to facilitate a plethora of morphological and molecular analyses. Principal component analyses of plasma targeted metabolomics and lipidomics data and of proteome profiles from pancreas, liver, and kidney cortex clearly separated MIDY and WT samples. CONCLUSIONS: The broad spectrum of well-defined biosamples in the Munich MIDY Pig Biobank that will be available to the scientific community provides a unique resource for systematic studies of organ crosstalk in diabetes in a multi-organ, multi-omics dimension.


Subject(s)
Body Fluids , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Insulin/genetics , Swine/genetics , Tissue Banks , Animals , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/veterinary , Female , Germany
4.
Toxicol Pathol ; 44(3): 414-20, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26883152

ABSTRACT

This article provides guidelines for organ and tissue sampling adapted to porcine animal models in translational medical research. Detailed protocols for the determination of sampling locations and numbers as well as recommendations on the orientation, size, and trimming direction of samples from ∼50 different porcine organs and tissues are provided in the Supplementary Material. The proposed sampling protocols include the generation of samples suitable for subsequent qualitative and quantitative analyses, including cryohistology, paraffin, and plastic histology; immunohistochemistry;in situhybridization; electron microscopy; and quantitative stereology as well as molecular analyses of DNA, RNA, proteins, metabolites, and electrolytes. With regard to the planned extent of sampling efforts, time, and personnel expenses, and dependent upon the scheduled analyses, different protocols are provided. These protocols are adjusted for (I) routine screenings, as used in general toxicity studies or in analyses of gene expression patterns or histopathological organ alterations, (II) advanced analyses of single organs/tissues, and (III) large-scale sampling procedures to be applied in biobank projects. Providing a robust reference for studies of porcine models, the described protocols will ensure the efficiency of sampling, the systematic recovery of high-quality samples representing the entire organ or tissue as well as the intra-/interstudy comparability and reproducibility of results.


Subject(s)
Biomedical Research/methods , Histocytochemistry/methods , Specimen Handling/methods , Swine , Animals , Models, Animal , Random Allocation
5.
J Pathol ; 238(2): 345-58, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26455904

ABSTRACT

The two incretin hormones, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP1), were discovered 45 and 30 years ago. Initially, only their insulinotropic effect on pancreatic ß cells was known. Over the years, physiological and pharmacological effects of GIP and GLP1 in numerous extrapancreatic tissues were discovered which partially overlap, but may also be specific for GIP or GLP1 in certain target tissues. While the insulinotropic effect of GIP was found to be blunted in patients with type 2 diabetes, the function of GLP1 is preserved and GLP1 receptor agonists and dipeptidyl-peptidase 4 (DPP4) inhibitors, which prolong the half-life of incretins, are widely used in diabetes therapy. Wild-type and genetically modified rodent models have provided important mechanistic insights into the incretin system, but may have limitations in predicting the clinical efficacy and safety of incretin-based therapies. This review summarizes insights from rodent and non-rodent models (pig, non-human primate) into physiological and pharmacological incretin effects, with a focus on the pancreas. Similarities and differences between species are discussed and the increasing potential of genetically engineered pig models for translational incretin research is highlighted.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Hypoglycemic Agents/pharmacology , Incretins/physiology , Animals , Animals, Genetically Modified , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/physiology , Incretins/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/physiology , Liraglutide/pharmacology , Mice , Mice, Knockout , Primates , Receptors, Gastrointestinal Hormone/physiology , Rodentia , Swine
6.
J Transl Med ; 13: 73, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25890210

ABSTRACT

BACKGROUND: The glucagon-like peptide-1 receptor (GLP1R) agonist liraglutide improves glycemic control and reduces body weight of adult type 2 diabetic patients. However, efficacy and safety of liraglutide in adolescents has not been systematically investigated. Furthermore, possible pro-proliferative effects of GLP1R agonists on the endocrine and exocrine pancreas need to be further evaluated. We studied effects of liraglutide in adolescent pigs expressing a dominant-negative glucose-dependent insulinotropic polypeptide receptor (GIPR(dn)) in the beta-cells, leading to a pre-diabetic condition including disturbed glucose tolerance, reduced insulin secretion and progressive reduction of functional beta-cell mass. METHODS: Two-month-old GIPR(dn) transgenic pigs were treated daily with liraglutide (0.6-1.2 mg per day) or placebo for 90 days. Glucose homeostasis was evaluated prior to and at the end of the treatment period by performing mixed meal and intravenous glucose tolerance tests (MMGTT and IVGTT). Finally animals were subjected to necropsy and quantitative-stereological analyses were performed for evaluation of alpha- and beta-cell mass, beta-cell proliferation as well as acinus-cell proliferation. RESULTS: MMGTT at the end of the study revealed 23% smaller area under the curve (AUC) for glucose, a 36% smaller AUC insulin, and improved insulin sensitivity, while IVGTT showed a 15% smaller AUC glucose but unchanged AUC insulin in liraglutide- vs. placebo-treated animals. Liraglutide led to marked reductions in body weight gain (-31%) and food intake (-30%) compared to placebo treatment, associated with reduced phosphorylation of insulin receptor beta (INSRB)/insulin-like growth factor-1 receptor beta (IGF1RB) and protein kinase B (AKT) in skeletal muscle. Absolute alpha- and beta-cell mass was reduced in liraglutide-treated animals, but alpha- and beta-cell mass-to-body weight ratios were unchanged. Liraglutide neither stimulated beta-cell proliferation in the endocrine pancreas nor acinus-cell proliferation in the exocrine pancreas, excluding both beneficial and detrimental effects on the pig pancreas. CONCLUSIONS: Although plasma liraglutide levels of adolescent transgenic pigs treated in our study were higher compared to human trials, pro-proliferative effects on the endocrine or exocrine pancreas or other liraglutide-related side-effects were not observed.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Liraglutide/therapeutic use , Prediabetic State/drug therapy , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , Animals, Genetically Modified , Blood Glucose/metabolism , Cell Proliferation/drug effects , Cell Size/drug effects , Disease Models, Animal , Feeding Behavior/drug effects , Gastric Emptying/drug effects , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test , Insulin/metabolism , Insulin Secretion , Liraglutide/blood , Liraglutide/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Organ Size/drug effects , Prediabetic State/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Weight Gain/drug effects
7.
Transgenic Res ; 23(1): 27-38, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24065178

ABSTRACT

Diabetes mellitus (DM) has emerged into a steadily increasing health problem and the predicted future dimension of the global DM epidemic is alarming: an increase from currently 346 million to over 400 million affected people worldwide by the year 2030 was extrapolated. Thus concerted research efforts are imperative to gain insight into disease mechanisms and to expand the basis for development of preventive and therapeutic strategies. Diabetic rodent models have traditionally been used to follow these goals, but have limitations for translational research. The pig is another classical animal model for diabetes research. Genetic engineering now facilitates tailoring pig models which mimic human disease mechanisms at the molecular level. This article reviews the existing genetically engineered pig models for diabetes research and their current and future applications. Further, the potential role of the pig as donor of pancreatic islets for xenotransplantation or as host for growing human pancreas is outlined.


Subject(s)
Animals, Genetically Modified , Diabetes Mellitus/genetics , Swine/genetics , Animals , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Disease Models, Animal , Humans , Swine/metabolism
8.
Diabetes ; 62(5): 1505-11, 2013 May.
Article in English | MEDLINE | ID: mdl-23274907

ABSTRACT

Mutations in the insulin (INS) gene may cause permanent neonatal diabetes mellitus (PNDM). Ins2 mutant mouse models provided important insights into the disease mechanisms of PNDM but have limitations for translational research. To establish a large animal model of PNDM, we generated INS(C94Y) transgenic pigs. A line expressing high levels of INS(C94Y) mRNA (70-86% of wild-type INS transcripts) exhibited elevated blood glucose soon after birth but unaltered ß-cell mass at the age of 8 days. At 4.5 months, INS(C94Y) transgenic pigs exhibited 41% reduced body weight, 72% decreased ß-cell mass (-53% relative to body weight), and 60% lower fasting insulin levels compared with littermate controls. ß-cells of INS(C94Y) transgenic pigs showed a marked reduction of insulin secretory granules and severe dilation of the endoplasmic reticulum. Cataract development was already visible in 8-day-old INS(C94Y) transgenic pigs and became more severe with increasing age. Diabetes-associated pathological alterations of kidney and nervous tissue were not detected during the observation period of 1 year. The stable diabetic phenotype and its rescue by insulin treatment make the INS(C94Y) transgenic pig an attractive model for insulin supplementation and islet transplantation trials, and for studying developmental consequences of maternal diabetes mellitus.


Subject(s)
Diabetes Complications/pathology , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Disease Models, Animal , Insulin/genetics , Mutation , Amino Acid Substitution , Animals , Animals, Genetically Modified , Cataract/complications , Cataract/pathology , Cataract/physiopathology , Diabetes Complications/physiopathology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/physiopathology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Gene Expression Regulation , Hyperglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Insulin/therapeutic use , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , RNA, Messenger/metabolism , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Severity of Illness Index , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...