1
|
Paniagua-Sancho M, Casanova AG, Rodríguez-Estévez L, Cruz-González I, López-Hernández FJ, Martínez-Salgado C. Pathophysiological characterization of the ApoE -/-;db/db mouse: A model of diabetes and atherosclerosis. Methods 2025; 234:223-232. [PMID: 39761864 DOI: 10.1016/j.ymeth.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
The high prevalence of type 2 diabetes and atherosclerosis makes essential the availability of in vivo experimental models that accurately replicate the pathophysiological mechanisms of these diseases. Apolipoprotein E knockout mice (ApoE-/-) have been used in atherosclerosis studies, and the db/db mice show hyperphagia and obesity. Mice harbouring both alterations (i.e., ApoE-/-;db/db) are expected to develop combined features of type 2 diabetes, obesity and accelerated atherosclerosis. To deepen into their pathophysiological profile and further assess their potential as an experimental model, we studied their mortality and their pancreatic, cardiac, and renal phenotype. We analysed during 6 months the glycemic and lipid profile, pancreatic, cardiac and renal structure and function and atherosclerosis in ApoE-/-;db/db mice. ApoE-/-;db/db mice show increases in plasma glucose (although without statistical significance) and glucagon levels, total cholesterol, triglycerides and HDL-cholesterol and in both insulin-producing β and glucagon producing α cells, and in the tissue expression of both hormones with respect to control (C57BL/6) mice; they show a remarkably high degree of atherosclerosis, higher left ventricular ejection fraction. Although renal function is normal, glucose, sodium and albumin excretion and urinary flow are increased with respect to control mice. Summarizing, ApoE-/-;db/db mice constitute a suitable experimental model for the study of type 2 diabetes associated with atherosclerosis.
Collapse
Affiliation(s)
- María Paniagua-Sancho
- Translational Research On Renal and Cardiovascular Diseases (TRECARD), Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.
| | - Alfredo G Casanova
- Translational Research On Renal and Cardiovascular Diseases (TRECARD), Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.
| | | | | | - Francisco J López-Hernández
- Translational Research On Renal and Cardiovascular Diseases (TRECARD), Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.
| | - Carlos Martínez-Salgado
- Translational Research On Renal and Cardiovascular Diseases (TRECARD), Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.
| |
Collapse
|
2
|
Mrosewski I, Mantel V, Urbank M, Schulze-Tanzil G, Werner C, Gögele C, Kokozidou M, Bertsch T. Menaquinone-7 and its therapeutic potential in type 2 diabetes mellitus based on a Zucker diabetic fatty rat model. Heliyon 2024; 10:e40826. [PMID: 39719993 PMCID: PMC11666950 DOI: 10.1016/j.heliyon.2024.e40826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/26/2024] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is marked by insulin resistance, low grade chronic inflammation, and endothelial dysfunction. Vitamin K2, especially menaquinone-7 (MK-7), might delay T2DM progression and alleviate its consequences. Hence, this study evaluated the effects of MK-7 on serum and urine markers of diabetes in an animal model of T2DM. Methods Hetero- (fa/+, control) and homozygous (fa/fa, diabetic) male Zucker diabetic fatty (ZDF) rats were supplemented or not with MK-7 for 12 weeks. After euthanasia, vitamin K1, menaquinone-4 and MK-7 serum concentrations were analyzed via reversed phase high pressure liquid chromatography. Glucose (serum), fructosamine (serum) and creatinine (serum and urine) levels were assessed photometrically, serum cystatin C and urinary total protein were turbidimetrically determined. Serum transforming growth factor beta 1 (TGF-β1) and procollagen type III N-terminal peptide (PIIINP) were quantified with enzyme-linked immunosorbent assay. Urinary marker proteins were analyzed via sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Nephropathy was assessed histologically. Results Supplementation led to significantly elevated MK-7 serum levels and a significant reduction of PIIINP serum levels in both hetero- and homozygous ZDF rats. Additionally, not statistically significant reductions of TGF-β1 serum levels, proteinuria as well as the nephropathy score were observed. In vivo body mass, serum fructosamine, glucose, cystatin C and creatinine levels were unaffected. Conclusion MK-7 reduced serum markers of fibrosis, histological features of nephropathy and urinary protein excretion, but failed to affect serum markers of T2DM. The therapeutic potential of MK-7 in T2DM and its mode of action should be further investigated in more detail.
Collapse
Affiliation(s)
- Ingo Mrosewski
- MDI Limbach Berlin GmbH, Aroser Allee 84, 13407, Berlin, Germany
| | - Valeriya Mantel
- MDI Limbach Berlin GmbH, Aroser Allee 84, 13407, Berlin, Germany
- Berlin University of Applied Sciences and Technology, Luxemburger Str. 10, 13353, Berlin, Germany
| | - Matthias Urbank
- MDI Limbach Berlin GmbH, Aroser Allee 84, 13407, Berlin, Germany
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany
| | - Christian Werner
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany
| | - Clemens Gögele
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany
| | - Maria Kokozidou
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany
| | - Thomas Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany
| |
Collapse
|
3
|
Utrilla Ramos VA, Mendoza Valero MD, Robles Soto R, Domínguez Juárez L, Ojeda Nani V, Sandoval Romero MC, Silva Gómez AB. Obese male zucker rats show basilar dendritic retraction in the medial prefrontal cortex. Heliyon 2024; 10:e40210. [PMID: 39584109 PMCID: PMC11583711 DOI: 10.1016/j.heliyon.2024.e40210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
Obesity, a prevalent disorder, predisposes individuals to metabolic syndrome, type 2 diabetes mellitus, and high blood pressure. Obesity has been investigated in various organisms that display genetic, high-fat, and high-carbohydrate diet (HFCD)-induced obesity. Recent studies have found that both male and female Zucker rats, which are genetically obese, exhibit alterations in dendritic arborization of neurons in certain structures of the central nervous system. Therefore, the present study aimed to analyze dendritic arborization and dendritic spine density of pyramidal neurons in the medial prefrontal cortex (mPFC) of obese adult male Zucker rats using the Golgi-Cox method and Sholl analysis. Obese male Zucker rats exhibit increased body weight and high concentrations of glucose, triglycerides, and cholesterol. Analysis of mPFC pyramidal neurons in these rats revealed basilar dendritic retraction at a medium distance from the soma, in addition to a reduction in total basilar dendritic length, without any changes in dendritic spine density. These findings are consistent with previous reports, indicating that changes in dendritic retraction may occur as a result of the leptin receptor mutation itself, in addition to the reduction in dendritic arborization observed in other regions of the central nervous system in rats. Furthermore, we suggest the possibility that biological processes modulated by the mPFC, such as foraging and social behavior, are also affected.
Collapse
Affiliation(s)
- Vanessa Abigail Utrilla Ramos
- Laboratorio de Neurofisiología Experimental, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
| | - Martha Denice Mendoza Valero
- Maestría en Ciencias Biológicas, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
| | - Ricardo Robles Soto
- Maestría en Ciencias Biológicas, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
| | - Lesly Domínguez Juárez
- Laboratorio de Neurofisiología Experimental, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
| | - Valentina Ojeda Nani
- Laboratorio de Neurofisiología Experimental, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
| | - María Constelación Sandoval Romero
- Maestría en Ciencias Biológicas, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
| | - Adriana Berenice Silva Gómez
- Laboratorio de Neurofisiología Experimental, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
- Maestría en Ciencias Biológicas, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Edificio BIO1, Ciudad Universitaria, CP 72570, Puebla, Puebla, Mexico
| |
Collapse
|
4
|
Daniels Gatward LF, King AJF. Matching model with mechanism: Appropriate rodent models for studying various aspects of diabetes pathophysiology. Methods Cell Biol 2024; 192:39-68. [PMID: 39863393 DOI: 10.1016/bs.mcb.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Abstract
Many rodent models are available for preclinical diabetes research making it a challenge for researchers to choose the most appropriate one for their experimental question. To aid in this, models have classically been categorized according to which type of diabetes they represent, and further into whether the model is induced, spontaneous or the result of genetic manipulation. This fails to capture the complexity of pathogenesis seen in diabetes in humans. This includes pathogenesis specifically involving the beta cell, which is no longer considered to be innocuous in the development and progression of diabetes. In this chapter we explore rodent models that incorporate the initiating factors believed to be involved in type 1 diabetes (autoimmunity) and type 2 diabetes (insulin resistance), before further discussing rodents that can be used to model specific mechanisms involved in a failure of functional beta cell mass (impaired beta cell function and beta cell apoptosis). We segregate models of beta cell pathogenesis based on the beta cell stressor predominantly associated with phenotype, but it is important to consider that most rodent models will exhibit more than one beta cell stressor. Similarly, many models exhibit more than one pathogenic mechanism, for example the same model may show insulin resistance, impaired beta cell function as well as beta cell loss. This can complicate interpretation of results and should be considered, and the model thoroughly researched, during the experimental planning stage.
Collapse
Affiliation(s)
- Lydia F Daniels Gatward
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom
| | - Aileen J F King
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
5
|
Tovar R, de Ceglia M, Rodríguez-Pozo M, Vargas A, Gavito A, Suárez J, Boronat A, de la Torre R, de Fonseca FR, Baixeras E, Decara J. Hydroxytyrosol Linoleoyl Ether Ameliorates Metabolic-Associated Fatty Liver Disease Symptoms in Obese Zucker Rats. ACS Pharmacol Transl Sci 2024; 7:1571-1583. [PMID: 38751648 PMCID: PMC11092116 DOI: 10.1021/acsptsci.4c00105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 05/18/2024]
Abstract
A main hepatic consequence of obesity is metabolic-associated fatty liver disease (MAFLD), currently treated by improving eating habits and administrating fibrates yet often yielding suboptimal outcomes. Searching for a new therapeutic approach, we aimed to evaluate the efficacy of hydroxytyrosol linoleoyl ether (HTLE), a dual Ppar-α agonist/Cb1 antagonist with inherent antioxidant properties, as an antisteatotic agent. Using lean and obese Zucker rats, they were administrated daily doses of HTLE (3 mg/kg) over a 15-day period, evaluating its safety profile, pharmacokinetics, impact on body weight, hepatic fat content, expression of key enzymes involved in lipogenesis/fatty acid oxidation, and antioxidant capacity. HTLE decreased the body weight and food intake in both rat genotypes. Biochemical analysis demonstrated a favorable safety profile for HTLE along with decreased concentrations of urea, total cholesterol, and aspartate aminotransferase AST transaminases in plasma. Notably, HTLE exhibited potent antisteatotic effects in obese rats, evidenced by a decrease in liver fat content and downregulation of lipogenesis-related enzymes, alongside increased expression of proteins controlling lipid oxidation. Moreover, HTLE successfully counteracted the redox imbalance associated with MAFLD in obese rats, attenuating lipid peroxidation and replenishing both glutathione levels and the overall antioxidant. Our findings highlight the effectiveness of triple-action strategies in managing MAFLD effectively. Based on our results in the Zucker rat model, HTLE emerges as a promising candidate with triple functionality as an anorexigenic, antisteatotic, and antioxidant agent, offering potential relief from MAFLD symptoms associated with obesity while exhibiting minimal side effects. In conclusion, our study positions HTLE as a highly promising compound for therapeutic intervention in MAFLD treatment, warranting further exploration in clinical trials.
Collapse
Affiliation(s)
- Rubén Tovar
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
- Facultad
de Ciencias, Universidad de Málaga,
Campus Universitario de Teatinos s/n, Málaga 29010, Spain
| | - Marialuisa de Ceglia
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
| | - Miguel Rodríguez-Pozo
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
| | - Antonio Vargas
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
| | - Ana Gavito
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
| | - Juan Suárez
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
- Departamento
de Anatomía Humana, Medicina Legal e Historia de la Ciencia,
Facultad de Medicina, Universidad de Málaga,
Campus Universitario de Teatinos s/n, Málaga 29010, Spain
| | - Anna Boronat
- Grupo
de Farmacología Integrada y Neurociencia de Sistemas, Programa
de investigación en Neurociencias, Instituto de Investigaciones Médicas Hospital del Mar (IMIM), C/del Dr. Aiguader 88, Barcelona 08003, Spain
| | - Rafael de la Torre
- Grupo
de Farmacología Integrada y Neurociencia de Sistemas, Programa
de investigación en Neurociencias, Instituto de Investigaciones Médicas Hospital del Mar (IMIM), C/del Dr. Aiguader 88, Barcelona 08003, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
| | - Elena Baixeras
- Departamento
de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Málaga, Campus Universitario
de Teatinos s/n, Málaga 29010, Spain
| | - Juan Decara
- Instituto
de Investigación Biomédica de Málaga y Plataforma
en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, UGC Salud Mental, Av. Carlos Haya 82, Málaga 29010, Spain
- Departamento
de Anatomía Humana, Medicina Legal e Historia de la Ciencia,
Facultad de Medicina, Universidad de Málaga,
Campus Universitario de Teatinos s/n, Málaga 29010, Spain
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Chandrasekaran P, Weiskirchen R. The Role of Obesity in Type 2 Diabetes Mellitus-An Overview. Int J Mol Sci 2024; 25:1882. [PMID: 38339160 PMCID: PMC10855901 DOI: 10.3390/ijms25031882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity or excessive weight gain is identified as the most important and significant risk factor in the development and progression of type 2 diabetes mellitus (DM) in all age groups. It has reached pandemic dimensions, making the treatment of obesity crucial in the prevention and management of type 2 DM worldwide. Multiple clinical studies have demonstrated that moderate and sustained weight loss can improve blood glucose levels, insulin action and reduce the need for diabetic medications. A combined approach of diet, exercise and lifestyle modifications can successfully reduce obesity and subsequently ameliorate the ill effects and deadly complications of DM. This approach also helps largely in the prevention, control and remission of DM. Obesity and DM are chronic diseases that are increasing globally, requiring new approaches to manage and prevent diabetes in obese individuals. Therefore, it is essential to understand the mechanistic link between the two and design a comprehensive approach to increase life expectancy and improve the quality of life in patients with type 2 DM and obesity. This literature review provides explicit information on the clinical definitions of obesity and type 2 DM, the incidence and prevalence of type 2 DM in obese individuals, the indispensable role of obesity in the pathophysiology of type 2 DM and their mechanistic link. It also discusses clinical studies and outlines the recent management approaches for the treatment of these associated conditions. Additionally, in vivo studies on obesity and type 2 DM are discussed here as they pave the way for more rigorous development of therapeutic approaches.
Collapse
Affiliation(s)
- Preethi Chandrasekaran
- UT Southwestern Medical Center Dallas, 5323 Harry Hines Blvd. ND10.504, Dallas, TX 75390-9014, USA
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH), University Hospital Aachen, D-52074 Aachen, Germany
| |
Collapse
|
9
|
Sun Z, Liu Y, Zhao Y, Xu Y. Animal Models of Type 2 Diabetes Complications: A Review. Endocr Res 2024; 49:46-58. [PMID: 37950485 DOI: 10.1080/07435800.2023.2278049] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Diabetes mellitus is a multifactorial metabolic disease, of which type 2 diabetes (T2D) is one of the most common. The complications of diabetes are far more harmful than diabetes itself. Type 2 diabetes complications include diabetic nephropathy (DN), diabetic heart disease, diabetic foot ulcers (DFU), diabetic peripheral neuropathy (DPN), and diabetic retinopathy (DR) et al. Many animal models have been developed to study the pathogenesis of T2D and discover an effective strategy to treat its consequences. In this sense, it is crucial to choose the right animal model for the corresponding diabetic complication. This paper summarizes and classifies the animal modeling approaches to T2D complications and provides a comprehensive review of their advantages and disadvantages. It is hopeful that this paper will provide theoretical support for animal trials of diabetic complications.
Collapse
Affiliation(s)
- Zhongyan Sun
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao SAR, Taipa, PR China
| | - Yadi Liu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao SAR, Taipa, PR China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, Taipa, PR China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao SAR, Taipa, PR China
- Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine,Macau University of Science and Technology, Zhuhai, PR China
- Macau University of Science and Technology, Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, PR China
| |
Collapse
|
10
|
Patel NA, Lui A, Trujillo AN, Motawe ZY, Bader D, Schuster J, Burgess A, Alves NG, Jo M, Breslin JW. Female and male obese Zucker rats display differential inflammatory mediator and long non-coding RNA profiles. Life Sci 2023; 335:122285. [PMID: 37995934 PMCID: PMC10760426 DOI: 10.1016/j.lfs.2023.122285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023]
Abstract
AIMS The goal of this study was to identify mediators in peri-lymphatic adipose tissue (PLAT) that are altered in obese versus lean Zucker rats, with focus on potential sex differences MAIN METHODS: Mesenteric PLAT was analyzed with protein and lncRNA arrays. Additional RT-PCR confirmation was performed with epididymal/ovarian fat. KEY FINDINGS MCP-1, TCK-1, Galectin-1, Galectin-3, and neuropilin-1 were elevated in PLAT from obese rats of both sexes. However, 11 additional proteins were elevated only in obese males while 24 different proteins were elevated in obese females. Profiling of lncRNAs revealed lean males have elevated levels of NEAT1, MALAT1 and GAS5 compared to lean females. NEAT1, MALAT1, and GAS5 were significantly reduced with obesity in males but not in females. Another lncRNA, HOTAIR, was higher in lean females compared to males, and its levels in females were reduced with obesity. Obese rats of both sexes had similar histologic findings of mesenteric macrophage crown-like structures and hepatocyte fat accumulation. SIGNIFICANCE While obese male and female Zucker rats both have increased inflammation, they have distinct signals. Future studies of the proteome and lncRNA landscape of obese males vs. females in various animal models and in human subjects are warranted to better guide development of therapeutics for obesity-induced inflammation.
Collapse
Affiliation(s)
- Niketa A Patel
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, United States of America; James A. Haley Veteran's Hospital, United States of America
| | - Ashley Lui
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, United States of America; James A. Haley Veteran's Hospital, United States of America
| | - Andrea N Trujillo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Zeinab Y Motawe
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Deena Bader
- James A. Haley Veteran's Hospital, United States of America
| | - Jane Schuster
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Andrea Burgess
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Natascha G Alves
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Michiko Jo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America; Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Japan
| | - Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America.
| |
Collapse
|
11
|
Athmuri DN, Shiekh PA. Experimental diabetic animal models to study diabetes and diabetic complications. MethodsX 2023; 11:102474. [PMID: 38023309 PMCID: PMC10661736 DOI: 10.1016/j.mex.2023.102474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023] Open
Abstract
Diabetes is an endocrine illness involving numerous physiological systems. To understand the intricated pathophysiology and disease progression in diabetes, small animals are still the most relevant model systems, despite the availability and progression in numerous invitro and insilico research methods in recent years. In general, experimental diabetes is instigated mainly due to the effectiveness of animal models in illuminating disease etiology. Most diabetes trials are conducted on rodents, while some research is conducted on larger animals. This review will discuss the methodology and mechanisms in detail for preparing diabetic animal models, considering the following important points. The exact pathophysiology of the disease may or may not be replicated in animal models, the correct induction doses must be given and the combination of different approaches for the models is recommended to get desired results.•Animal models are essential to understand diabetes etiology and pathophysiology.•Diabetic models can be developed in both rodents and non-rodents.•Chemically induced and genetic models of diabetes are widely used to study diabetes and diabetic complications.
Collapse
Affiliation(s)
- Durga Nandini Athmuri
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Parvaiz Ahmad Shiekh
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
12
|
Xu X, Hu X, Ma G, Wang T, Wu J, Zhu X, Chen G, Zhao L, Chen J. Detecting fa leptin receptor mutation in Zucker rats with tetra-primer amplification-refractory mutation system (ARMS)-PCR. Heliyon 2023; 9:e20159. [PMID: 37809507 PMCID: PMC10559934 DOI: 10.1016/j.heliyon.2023.e20159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the genetic mutation (fa) in the gene encoding for leptin receptor, homozygous Zucker rats (fa-/-) develop excessive adiposity and become an experimental animal model in obesity and metabolic-related diseases research. Based on tetra-primer amplification refractory mutation system-polymerase chain reaction (ARMS-PCR), we developed a method to quickly genotype Zucker rats with a mutated fa allele from their wildtype littermates. The three genotypes are clearly discriminated on 2.0% agarose gel. Our method can be used as a reliable tool to set up and maintain the breeding colony in animal facilities as well as assign animals to control and treatment groups based on their genotypes for animal studies.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Xinge Hu
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Guodong Ma
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Tiannan Wang
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Jayne Wu
- Department of Electrical Engineering and Computer Science, The University of Tennessee Knoxville, TN, 37996, United States
| | - Xiaojuan Zhu
- Office of Information Technology, The University of Tennessee Knoxville, TN, 37996, United States
| | - Guoxun Chen
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee Knoxville, TN, 37996, United States
| |
Collapse
|
13
|
Bordet S, Luaces JP, Herrera MI, Gonzalez LM, Kobiec T, Perez-Lloret S, Otero-Losada M, Capani F. Neuroprotection from protein misfolding in cerebral hypoperfusion concurrent with metabolic syndrome. A translational perspective. Front Neurosci 2023; 17:1215041. [PMID: 37650104 PMCID: PMC10463751 DOI: 10.3389/fnins.2023.1215041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/17/2023] [Indexed: 09/01/2023] Open
Abstract
Based on clinical and experimental evidence, metabolic syndrome (MetS) and type 2 diabetes (T2D) are considered risk factors for chronic cerebral hypoperfusion (CCH) and neurodegeneration. Scientific evidence suggests that protein misfolding is a potential mechanism that explains how CCH can lead to either Alzheimer's disease (AD) or vascular cognitive impairment and dementia (VCID). Over the last decade, there has been a significant increase in the number of experimental studies regarding this issue. Using several animal paradigms and different markers of CCH, scientists have discussed the extent to which MetSor T2D causes a decrease in cerebral blood flow (CBF). In addition, different models of CCH have explored how long-term reductions in oxygen and energy supply can trigger AD or VCID via protein misfolding and aggregation. Research that combines two or three animal models could broaden knowledge of the links between these pathological conditions. Recent experimental studies suggest novel neuroprotective properties of protein-remodeling factors. In this review, we present a summarized updated revision of preclinical findings, discussing clinical implications and proposing new experimental approaches from a translational perspective. We are confident that research studies, both clinical and experimental, may find new diagnostic and therapeutic tools to prevent neurodegeneration associated with MetS, diabetes, and any other chronic non-communicable disease (NCD) associated with diet and lifestyle risk factors.
Collapse
Affiliation(s)
- Sofía Bordet
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Juan Pablo Luaces
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
| | - Maria Ines Herrera
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Liliana Mirta Gonzalez
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
| | - Tamara Kobiec
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Santiago Perez-Lloret
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Observatorio de Salud Pública, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
14
|
Martín-Carro B, Donate-Correa J, Fernández-Villabrille S, Martín-Vírgala J, Panizo S, Carrillo-López N, Martínez-Arias L, Navarro-González JF, Naves-Díaz M, Fernández-Martín JL, Alonso-Montes C, Cannata-Andía JB. Experimental Models to Study Diabetes Mellitus and Its Complications: Limitations and New Opportunities. Int J Mol Sci 2023; 24:10309. [PMID: 37373455 DOI: 10.3390/ijms241210309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Preclinical biomedical models are a fundamental tool to improve the knowledge and management of diseases, particularly in diabetes mellitus (DM) since, currently, the pathophysiological and molecular mechanisms involved in its development are not fully clarified, and there is no treatment to cure DM. This review will focus on the features, advantages and limitations of some of the most used DM models in rats, such as the spontaneous models: Bio-Breeding Diabetes-Prone (BB-DP) and LEW.1AR1-iddm, as representative models of type 1 DM (DM-1); the Zucker diabetic fatty (ZDF) and Goto-kakizaki (GK) rats, as representative models of type 2 DM (DM-2); and other models induced by surgical, dietary and pharmacological-alloxan and streptozotocin-procedures. Given the variety of DM models in rats, as well as the non-uniformity in the protocols and the absence of all the manifestation of the long-term multifactorial complications of DM in humans, the researchers must choose the one that best suits the final objectives of the study. These circumstances, added to the fact that most of the experimental research in the literature is focused on the study of the early phase of DM, makes it necessary to develop long-term studies closer to DM in humans. In this review, a recently published rat DM model induced by streptozotocin injection with chronic exogenous administration of insulin to reduce hyperglycaemia has also been included in an attempt to mimic the chronic phase of DM in humans.
Collapse
Affiliation(s)
- Beatriz Martín-Carro
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Donate-Correa
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Sara Fernández-Villabrille
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Julia Martín-Vírgala
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sara Panizo
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan F Navarro-González
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- Nephrology Service, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José L Fernández-Martín
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Alonso-Montes
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
15
|
Larsen AT, Melander SA, Sonne N, Bredtoft E, Al-Rubai M, Karsdal MA, Henriksen K. Dual amylin and calcitonin receptor agonist treatment improves insulin sensitivity and increases muscle-specific glucose uptake independent of weight loss. Biomed Pharmacother 2023; 164:114969. [PMID: 37269811 DOI: 10.1016/j.biopha.2023.114969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023] Open
Abstract
Dual amylin and calcitonin receptor agonists (DACRAs) are known to induce significant weight loss as well as improve glucose tolerance, glucose control, and insulin action in rats. However, to what extent DACRAs affect insulin sensitivity beyond that induced by weight loss and if DACRAs affect glucose turnover including tissue-specific glucose uptake is still unknown. Hyperinsulinemic glucose clamp studies were carried out in pre-diabetic ZDSD and diabetic ZDF rats treated with either the DACRA KBP or the long-acting DACRA KBP-A for 12 days. The glucose rate of disappearance was assessed using 3-3H glucose and tissue-specific glucose uptake was evaluated using 14C-2-deoxy-D-glucose (14C-2DG). In diabetic ZDF rats, KBP treatment significantly reduced fasting blood glucose and improved insulin sensitivity independent of weight loss. Furthermore, KBP increased the rate of glucose clearance, likely by increasing glucose storage, but without altering the endogenous glucose production. This was confirmed in pre-diabetic ZDSD rats. Direct assessment of tissue-specific glucose uptake showed, that both KBP and KBP-A significantly increased glucose uptake in muscles. In summary, KBP treatment significantly improved insulin sensitivity in diabetic rats and markedly increased glucose uptake in muscles. Importantly, in addition to their well-established weight loss potential, the KBPs have an insulin-sensitizing effect independent of weight loss, highlighting DACRAs as promising agents for the treatment of type 2 diabetes and obesity.
Collapse
Affiliation(s)
| | | | | | | | | | - Morten A Karsdal
- Nordic Bioscience, 2730 Herlev, Denmark; KeyBioscience AG, Stans, Switzerland
| | - Kim Henriksen
- Nordic Bioscience, 2730 Herlev, Denmark; KeyBioscience AG, Stans, Switzerland
| |
Collapse
|
16
|
Weninger SN, Ding A, Browne EN, Frost ML, Schiro G, Laubitz D, Duca FA. Longitudinal Characterization of the Gut Microbiota in the Diabetic ZDSD Rat Model and Therapeutic Potential of Oligofructose. Metabolites 2023; 13:660. [PMID: 37233701 PMCID: PMC10220957 DOI: 10.3390/metabo13050660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
The complex development of type 2 diabetes (T2D) creates challenges for studying the progression and treatment of the disease in animal models. A newly developed rat model of diabetes, the Zucker Diabetic Sprague Dawley (ZDSD) rat, closely parallels the progression of T2D in humans. Here, we examine the progression of T2D and associated changes in the gut microbiota in male ZDSD rats and test whether the model can be used to examine the efficacy of potential therapeutics such as prebiotics, specifically oligofructose, that target the gut microbiota. Bodyweight, adiposity, and fed/fasting blood glucose and insulin were recorded over the course of the study. Glucose and insulin tolerance tests were performed, and feces collected at 8, 16, and 24 weeks of age for short-chain fatty acids and microbiota analysis using 16s rRNA gene sequencing. At the end of 24 weeks of age, half of the rats were supplemented with 10% oligofructose and tests were repeated. We observed a transition from healthy/nondiabetic to prediabetic and overtly diabetic states, via worsened insulin and glucose tolerance and significant increases in fed/fasted glucose, followed by a significant decrease in circulating insulin. Acetate and propionate levels were significantly increased in the overt diabetic state compared to healthy and prediabetic. Microbiota analysis demonstrated alterations in the gut microbiota with shifts in alpha and beta diversity as well as alterations in specific bacterial genera in healthy compared to prediabetic and diabetic states. Oligofructose treatment improved glucose tolerance and shifted the cecal microbiota of the ZDSD rats during late-stage diabetes. These findings underscore the translational potential of ZDSD rats as a model of T2D and highlight potential gut bacteria that could impact the development of the disease or serve as a biomarker for T2D. Additionally, oligofructose treatment was able to moderately improve glucose homeostasis.
Collapse
Affiliation(s)
- Savanna N. Weninger
- Department of Physiological Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Angela Ding
- Department of Physiological Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Elizabeth N. Browne
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Morgan L. Frost
- School of Animal and Comparative Biomedical Sciences, College of Agricultural and Life Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Gabriele Schiro
- The PANDA Core for Genomics and Microbiome Research, Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
| | - Daniel Laubitz
- The PANDA Core for Genomics and Microbiome Research, Department of Pediatrics, University of Arizona, Tucson, AZ 85721, USA
| | - Frank A. Duca
- School of Animal and Comparative Biomedical Sciences, College of Agricultural and Life Sciences, University of Arizona, Tucson, AZ 85721, USA
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
17
|
Rodent Models of Diabetic Retinopathy as a Useful Research Tool to Study Neurovascular Cross-Talk. BIOLOGY 2023; 12:biology12020262. [PMID: 36829539 PMCID: PMC9952991 DOI: 10.3390/biology12020262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023]
Abstract
Diabetes is a group of metabolic diseases leading to dysfunction of various organs, including ocular complications such as diabetic retinopathy (DR). Nowadays, DR treatments involve invasive options and are applied at the sight-threatening stages of DR. It is important to investigate noninvasive or pharmacological methods enabling the disease to be controlled at the early stage or to prevent ocular complications. Animal models are useful in DR laboratory practice, and this review is dedicated to them. The first part describes the characteristics of the most commonly used genetic rodent models in DR research. The second part focuses on the main chemically induced models. The authors pay particular attention to the streptozotocin model. Moreover, this section is enriched with practical aspects and contains the current protocols used in research in the last three years. Both parts include suggestions on which aspect of DR can be tested using a given model and the disadvantages of each model. Although animal models show huge variability, they are still an important and irreplaceable research tool. Note that the choice of a research model should be thoroughly considered and dependent on the aspect of the disease to be analyzed.
Collapse
|
18
|
Liu B, Xu J, Lu L, Gao L, Zhu S, Sui Y, Cao T, Yang T. Metformin induces pyroptosis in leptin receptor-defective hepatocytes via overactivation of the AMPK axis. Cell Death Dis 2023; 14:82. [PMID: 36737598 PMCID: PMC9898507 DOI: 10.1038/s41419-023-05623-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023]
Abstract
Metformin is the biguanide of hepatic insulin sensitizer for patients with non-alcohol fatty liver disease (NAFLD). Findings regarding its efficacy in restoring blood lipids and liver histology have been contradictory. In this study, we explore metformin's preventive effects on NAFLD in leptin-insensitive individuals. We used liver tissue, serum exosomes and isolated hepatocytes from high-fat diet (HFD)-induced Zucker diabetic fatty (ZDF) rats and leptin receptor (Lepr) knockout rats to investigate the correlation between hepatic Lepr defective and liver damage caused by metformin. Through immunostaining, RT-PCR and glucose uptake monitoring, we showed that metformin treatment activates adenosine monophosphate (AMP)-activated protein kinase (AMPK) and its downstream cytochrome C oxidase (CCO). This leads to overactivation of glucose catabolism-related genes, excessive energy repertoire consumption, and subsequent hepatocyte pyroptosis. Single-cell RNA sequencing further confirmed the hyper-activation of glucose catabolism after metformin treatment. Altogether, we showed that functional Lepr is necessary for metformin treatment to be effective, and that long-term metformin treatment might promote NAFLD progression in leptin-insensitive individuals. This provides important insight into the clinical application of metformin.
Collapse
Affiliation(s)
- Bingli Liu
- Department of Orthopedics, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China
| | - Jingyuan Xu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Linyao Lu
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Lili Gao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Shengjuan Zhu
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Yi Sui
- Department of Nutrition, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ting Cao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Tao Yang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
19
|
Li W, Twaddle NC, Spray B, Nounamo B, Monzavi-Karbassi B, Hakkak R. Feeding Soy Protein Concentrates with Low and High Isoflavones Alters 9 and 18 Weeks Serum Isoflavones and Inflammatory Protein Levels in Lean and Obese Zucker Rats. J Med Food 2023; 26:120-127. [PMID: 36720082 DOI: 10.1089/jmf.2022.0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Soy's anti-inflammatory properties contribute to the health benefits of soy foods. This study was designed to investigate the bioavailability of soy isoflavones and whether the isoflavone content of soy protein concentrate diet would affect serum inflammatory proteins in an obese (fa/fa) Zucker rat model. Six-week-old male lean (L) and obese (O) Zucker rats were fed a casein control diet (C), soy protein concentrate with low isoflavones (SPC-LIF), or soy protein concentrate with high isoflavones (SPC-HIF) (7 rats/dietary group) before being killed at 9 and 18 weeks. Serum samples were analyzed for isoflavones and inflammatory proteins. At both time points, serum total (aglycone + conjugates) genistein, daidzein, and equol concentrations were significantly higher in L-SPC-HIF and O-SPC-HIF groups compared with L-SPC-LIF and O-SPC-LIF groups, respectively, and were not detectable in either L-C or O-C groups. At week 9, serum C-reactive protein (CRP) concentration was significantly lower in O-SPC-HIF group compared with O-C and O-SPC-LIF group, whereas proteins tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels did not differ between any groups. At week 18, serum CRP levels in both O-SPC-HIF and O-SPC-LIF groups were significantly lower compared with the O-C group. TNF-α level was higher in the O-SPC-LIF group compared with both O-C and O-SPC-HIF groups, whereas IL-6 levels were not different between any groups. Taken together, feeding Zucker rats SPC-LIF and SPC-HIF diets led to different serum isoflavone concentrations in both L and O Zucker rats and altered CRP and TNF-α levels in obese Zucker rats compared with controls.
Collapse
Affiliation(s)
- Wei Li
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Nathan C Twaddle
- Division of Biochemical Toxicology of National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Beverly Spray
- Division of Biostatistics Core, Arkansas Children's Research Institute, Little Rock, Arkansas, USA
| | - Bernice Nounamo
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Reza Hakkak
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Division of Biostatistics Core, Arkansas Children's Research Institute, Little Rock, Arkansas, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
20
|
Gomes MFP, de Moura EDOC, Cardoso NM, da Silva GA, Dos Santos ACC, de Souza FS, Estadella D, Lambertucci RH, Lago JHG, Medeiros A. Supplementation with okra combined or not with exercise training is able to protect the heart of animals with metabolic syndrome. Sci Rep 2023; 13:1468. [PMID: 36702820 PMCID: PMC9879946 DOI: 10.1038/s41598-023-28072-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
The metabolic syndrome (MetS) is a clinical manifestation strongly associated with cardiovascular disease, the main cause of death worldwide. In view of this scenario, many therapeutic proposals have appeared in order to optimize the treatment of individuals with MetS, including the practice of exercise training (ET) and the consumption of okra (O). The aim of the present study was to evaluate the effect of O consumption and/or ET in animals with MetS. In all, 32 male Zucker rats (fa/fa) at 10 weeks old were randomly distributed into four groups of 8 animals each: MetS, MetS+O, MetS+ET and MetS+ET+O, and 8 lean Zucker rats (fa/ +) comprised the control group. Okra was administered by orogastric gavage 2x/day (morning and night, 100 mg/kg), 5 days/week, for 6 weeks. The ET was performed on a treadmill 1x/day (afternoon), 5 days/week, 60 min/day, in an intensity of 70% of maximal capacity, for the same days of O treatment. It was found that, O consumption alone was able to promote improved insulin sensitivity (MetS 93.93 ± 8.54 mg/dL vs. MetS+O 69.95 ± 18.7 mg/dL, p ≤ 0.05, d = 1.65, CI = 50.32 -89.58, triglyceride reduction (MetS 492.9 ± 97.8 mg/dL vs. MetS+O 334.9 ± 98.0 mg/dL, p ≤ 0.05, d = 1.61, CI = 193.2-398.7). In addition, it promoted a reduction in systolic blood pressure (MetS 149.0 ± 9.3 mmHg vs. MetS+O 132.0 ± 11.4 mmHg, p ≤ 0.05, d = 1.63, CI = 120-140), prevented an increase in cardiac collagen (MetS 12.60 ± 2.08% vs. MetS+O 7.52 ± 0.77%, p ≤ 0.05, d = 3.24, CI = 6.56-8.49). When associated with ET, the results were similar. Thus, we conclude that O consumption combined or not with aerobic ET can have a protective effect on the cardiac tissue of rats with MetS.
Collapse
Affiliation(s)
- Moisés Felipe Pereira Gomes
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil.
- Center for Applied Social Sciences and Health, Universidade Católica de Santos (Unisantos), Av. Conselheiro Nébias, 300, Vila Matias, Santos, SP, 11015-002, Brazil.
| | | | - Naiara Magalhães Cardoso
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Graziele Aparecida da Silva
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Ana Carolina Cardoso Dos Santos
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Fernanda Samantha de Souza
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), Rua Prof. Artur Riedel, n° 275, Eldorado, Diadema, SP, 09972-270, Brazil
| | - Débora Estadella
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - Rafael Herling Lambertucci
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| | - João Henrique Ghilardi Lago
- Center of Natural and Human Sciences, Universidade Federal Do ABC, Av. Dos Estados, 500, Bangú, Santo André, SP, 09210-580, Brazil
| | - Alessandra Medeiros
- Department of Bioscience, Universidade Federal de São Paulo (UNIFESP), R. Silva Jardim, 136 - Vila Matias, Santos, SP, 11015-020, Brazil
| |
Collapse
|
21
|
Dupak R, Hrnkova J, Simonova N, Kovac J, Ivanisova E, Kalafova A, Schneidgenova M, Prnova MS, Brindza J, Tokarova K, Capcarova M. The consumption of sea buckthorn (Hippophae rhamnoides L.) effectively alleviates type 2 diabetes symptoms in spontaneous diabetic rats. Res Vet Sci 2022; 152:261-269. [PMID: 36063603 DOI: 10.1016/j.rvsc.2022.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022]
Abstract
Sea buckthorn (Hippophae rhamnoides L.) is described by various beneficial effects as it contains several bioactive substances characterized by antioxidant effects. These effects are closely related to the reduction of oxidative stress that is involved in the development of the disease. One such diseases is Diabetes mellitus, the prevalence of which is growing and is associated primarily with diet, lack of exercise and/or genetics. This study intends to examine the effects of sea buckthorn and metformin on body weight, water and feed intake, glycaemia, insulinemia, sorbitol accumulation and cataract development in Zucker diabetic fatty rats, which represent an animal model of type 2 Diabetes mellitus, as well as to characterize the individual content of bioactive substances and the antioxidant activity of sea buckthorn. Particular concentrations were applied (500 and 1000 mg.kg-1 body weight of sea buckthorn, and combinations with 150 mg.kg-1 body weight of metformin) by gastric gavage. The total antioxidant capacity and bioactive compounds were determined by spectrophotometric analysis. The best results of the study showed suppression of hyperglycaemia, water intake, decreased sorbitol levels in the lens of the eyes after sea buckthorn treatment. Determination of bioactive compounds showed significantly higher values in dry berries when compared to fresh berries of sea buckthorn and high total antioxidant capacity. Our results represent an interest in sea buckthorn and its potential use in the treatment of Diabetes mellitus as well as other experimental studies.
Collapse
Affiliation(s)
- Rudolf Dupak
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Jana Hrnkova
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Nikoleta Simonova
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Jan Kovac
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Eva Ivanisova
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Anna Kalafova
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Monika Schneidgenova
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Marta Soltesova Prnova
- Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Science, Dubravska cesta 9, 841 04 Bratislava, Slovak Republic.
| | - Jan Brindza
- Institute of Plant and Environmental Sciences, Faculty of Agrobiology and Food Resources, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Katarina Tokarova
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| | - Marcela Capcarova
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda Andreja Hlinku 2, 949 76 Nitra, Slovak Republic.
| |
Collapse
|
22
|
Reduced Tyrosine and Serine-632 Phosphorylation of Insulin Receptor Substrate-1 in the Gastrocnemius Muscle of Obese Zucker Rat. Curr Issues Mol Biol 2022; 44:6015-6027. [PMID: 36547071 PMCID: PMC9777198 DOI: 10.3390/cimb44120410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Obesity has become a serious health problem in the world, with increased morbidity, mortality, and financial burden on patients and health-care providers. The skeletal muscle is the most extensive tissue, severely affected by a sedentary lifestyle, which leads to obesity and type 2 diabetes. Obesity disrupts insulin signaling in the skeletal muscle, resulting in decreased glucose disposal, a condition known as insulin resistance. Although there is a large body of evidence on obesity-induced insulin resistance in various skeletal muscles, the molecular mechanism of insulin resistance due to a disruption in insulin receptor signaling, specifically in the gastrocnemius skeletal muscle of obese Zucker rats (OZRs), is not fully understood. This study subjected OZRs to a glucose tolerance test (GTT) to analyze insulin sensitivity. In addition, immunoprecipitation and immunoblotting techniques were used to determine the expression and tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1) and insulin receptor-β (IRβ), and the activation of serine-632-IRS-1 phosphorylation in the gastrocnemius muscle of Zucker rats. The results show that the GTT in the OZRs was impaired. There was a significant decrease in IRS-1 levels, but no change was observed in IRβ in the gastrocnemius muscle of OZRs, compared to Zucker leans. Obese rats had a higher ratio of tyrosine phosphorylation of IRS-1 and IRβ than lean rats. In obese rats, however, insulin was unable to induce tyrosine phosphorylation. Moreover, insulin increased the phosphorylation of serine 632-IRS-1 in the gastrocnemius muscle of lean rats. However, obese rats had a low basal level of serine-632-IRS-1 and insulin only mildly increased serine phosphorylation in obese rats, compared to those without insulin. Thus, we addressed the altered steps of the insulin receptor signal transduction in the gastrocnemius muscle of OZRs. These findings may contribute to a better understanding of human obesity and type 2 diabetes.
Collapse
|
23
|
Manuel Sánchez DM, Limón D, Silva Gómez AB. Obese male Zucker rats exhibit dendritic remodeling in neurons of the hippocampal trisynaptic circuit as well as spatial memory deficits. Hippocampus 2022; 32:828-838. [PMID: 36177907 DOI: 10.1002/hipo.23473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/26/2022] [Accepted: 09/17/2022] [Indexed: 01/07/2023]
Abstract
Obesity is characterized by excessive fat accumulation. The Zucker rat displays genetic obesity due to a mutation in the leptin receptor gene; this model is of great interest because of its similarity to human obesity. Brain regions may be affected by obesity, but detailed information is lacking. In the present study, we analyzed the morphology of neurons in the hippocampal trisynaptic circuit as well as the spatial memory of obese Zucker rats. We performed two experiments. Each experiment contained two experimental groups: the control group (male Long Evans rats) and the study group (obese male Zucker rats). We monitored the body weights of all rats over 4 weeks. In the first experiment, we analyzed the morphology of hippocampal neurons. Under anesthesia, we measured the abdominal and hip circumferences and collected at least 1 ml of blood to assess serum glucose (GLU), triglyceride (TGC), and cholesterol (COL) concentrations. We perfused the brains of these rats with 0.9% saline solution, incubated the brains in Golgi-Cox solution, and subsequently evaluated the morphology of pyramidal neurons in the hippocampus (the CA1-CA3 regions) and the entorhinal cortex as well as the morphology of granule neurons in the dentate gyrus. In the second experiment, we assessed the spatial memory of animals with the Morris water maze. The Zucker rats had an obese phenotype, as indicated by their elevated body weight and increased abdominal and hip circumferences as well as elevated GLU, COL, and TGC concentrations. Analysis of neurons from the specified regions in obese male Zucker rats indicated reduced dendritic arborization and reduced dendritic spine density. In terms of spatial learning and memory, the obese Zucker rats exhibited intact spatial learning (i.e., of platform location) but deficits in spatial memory. These data provide evidence that obesity alters the morphology and function of hippocampal neurons.
Collapse
Affiliation(s)
- Dulce María Manuel Sánchez
- Laboratorio de Neurofisiología Experimental, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Adriana Berenice Silva Gómez
- Laboratorio de Neurofisiología Experimental, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
24
|
Kroon T, Hagstedt T, Alexandersson I, Ferm A, Petersson M, Maurer S, Zarrouki B, Wallenius K, Oakes ND, Boucher J. Chronotherapy with a glucokinase activator profoundly improves metabolism in obese Zucker rats. Sci Transl Med 2022; 14:eabh1316. [DOI: 10.1126/scitranslmed.abh1316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Circadian rhythms play a critical role in regulating metabolism, including daily cycles of feeding/fasting. Glucokinase (GCK) is central for whole-body glucose homeostasis and oscillates according to a circadian clock. GCK activators (GKAs) effectively reduce hyperglycemia, but their use is also associated with hypoglycemia, hyperlipidemia, and hepatic steatosis. Given the circadian rhythmicity and natural postprandial activation of GCK, we hypothesized that GKA treatment would benefit from being timed specifically during feeding periods. Acute treatment of obese Zucker rats with the GKA AZD1656 robustly increased flux into all major metabolic pathways of glucose disposal, enhancing glucose elimination. Four weeks of continuous AZD1656 treatment of obese Zucker rats improved glycemic control; however, hepatic steatosis and inflammation manifested. In contrast, timing AZD1656 to feeding periods robustly reduced hepatic steatosis and inflammation in addition to improving glycemia, whereas treatment timed to fasting periods caused overall detrimental metabolic effects. Mechanistically, timing AZD1656 to feeding periods diverted newly synthesized lipid toward direct VLDL secretion rather than intrahepatic storage. In line with increased hepatic insulin signaling, timing AZD1656 to feeding resulted in robust activation of AKT, mTOR, and SREBP-1C after glucose loading, pathways known to regulate VLDL secretion and hepatic de novo lipogenesis. In conclusion, intermittent AZD1656 treatment timed to feeding periods promotes glucose disposal when needed the most, restores metabolic flexibility and hepatic insulin sensitivity, and thereby avoids hepatic steatosis. Thus, chronotherapeutic approaches may benefit the development of GKAs and other drugs acting on metabolic targets.
Collapse
Affiliation(s)
- Tobias Kroon
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
- Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothernburg 41345, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothernburg 40530 Sweden
| | - Therese Hagstedt
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Ida Alexandersson
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Annett Ferm
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Marie Petersson
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Stefanie Maurer
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Bader Zarrouki
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Kristina Wallenius
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Nicholas D. Oakes
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg 43183, Sweden
| | - Jeremie Boucher
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43183, Sweden
- Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothernburg 41345, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothernburg 40530 Sweden
| |
Collapse
|
25
|
Mao Y, Zhao K, Li P, Sheng Y. The emerging role of leptin in obesity-associated cardiac fibrosis: evidence and mechanism. Mol Cell Biochem 2022; 478:991-1011. [PMID: 36214893 DOI: 10.1007/s11010-022-04562-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022]
Abstract
Cardiac fibrosis is a hallmark of various cardiovascular diseases, which is quite commonly found in obesity, and may contribute to the increased incidence of heart failure arrhythmias, and sudden cardiac death in obese populations. As an endogenous regulator of adiposity metabolism, body mass, and energy balance, obesity, characterized by increased circulating levels of the adipocyte-derived hormone leptin, is a critical contributor to the pathogenesis of cardiac fibrosis. Although there are some gaps in our knowledge linking leptin and cardiac fibrosis, this review will focus on the interplay between leptin and major effectors involved in the pathogenesis underlying cardiac fibrosis at both cellular and molecular levels based on the current reports. The profibrotic effect of leptin is predominantly mediated by activated cardiac fibroblasts but may also involve cardiomyocytes, endothelial cells, and immune cells. Moreover, a series of molecular signals with a known profibrotic property is closely involved in leptin-induced fibrotic events. A more comprehensive understanding of the underlying mechanisms through which leptin contributes to the pathogenesis of cardiac fibrosis may open up a new avenue for the rapid emergence of a novel therapy for preventing or even reversing obesity-associated cardiac fibrosis.
Collapse
Affiliation(s)
- Yukang Mao
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China. .,Department of Cardiology, Jiangsu Province Hospital, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
26
|
Couto AR, Parreira B, Power DM, Pinheiro L, Madruga Dias J, Novofastovski I, Eshed I, Sarzi-Puttini P, Pappone N, Atzeni F, Verlaan JJ, Kuperus J, Bieber A, Ambrosino P, Kiefer D, Khan MA, Mader R, Baraliakos X, Bruges-Armas J. Evidence for a genetic contribution to the ossification of spinal ligaments in Ossification of Posterior Longitudinal Ligament and Diffuse idiopathic skeletal hyperostosis: A narrative review. Front Genet 2022; 13:987867. [PMID: 36276944 PMCID: PMC9586552 DOI: 10.3389/fgene.2022.987867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022] Open
Abstract
Diffuse Idiopathic Skeletal Hyperostosis (DISH) and Ossification of the Posterior Longitudinal Ligament (OPLL) are common disorders characterized by the ossification of spinal ligaments. The cause for this ossification is currently unknown but a genetic contribution has been hypothesized. Over the last decade, many studies on the genetics of ectopic calcification disorders have been performed, mainly on OPLL. Most of these studies were based on linkage analysis and case control association studies. Animal models have provided some clues but so far, the involvement of the identified genes has not been confirmed in human cases. In the last few years, many common variants in several genes have been associated with OPLL. However, these associations have not been at definitive levels of significance and evidence of functional significance is generally modest. The current evidence suggests a multifactorial aetiopathogenesis for DISH and OPLL with a subset of cases showing a stronger genetic component.
Collapse
Affiliation(s)
- Ana Rita Couto
- Hospital de Santo Espirito da Ilha Terceira EPER, SEEBMO, Angra do Heroísmo, Portugal
- Comprehensive Health Research Centre, Hospital de Santo Espírito da Ilha Terceira, Lisbon, Portugal
| | - Bruna Parreira
- Hospital de Santo Espirito da Ilha Terceira EPER, SEEBMO, Angra do Heroísmo, Portugal
- Comprehensive Health Research Centre, Hospital de Santo Espírito da Ilha Terceira, Lisbon, Portugal
| | - Deborah M. Power
- University of Algarve, Center of Marine Science (CCMAR), Faro, Portugal
| | - Luís Pinheiro
- Hospital de Santo Espirito da Ilha Terceira EPER, Orthopedics Service, Angra do Heroísmo, Portugal
| | - João Madruga Dias
- Centro Hospitalar Do Medio Tejo EPE Unidade de Torres Novas, Rheumatology Department, Santarém, Portugal
- CHRC Campus Nova Medical School, EpiDoc Research Unit, CEDOC, Lisboa, Portugal
| | | | | | | | - Nicola Pappone
- Istituti Clinici Scientifici Maugeri IRCCS, Neuromotor Rehabilitation Unit of Telese Terme Institute, Pavia, Italy
| | - Fabiola Atzeni
- Universita Degli Studi di Messina, Rheumatology Unit, Clinical and Experimental Medicine, Messina, Italy
| | - Jorrit-Jan Verlaan
- University Medical Centre, Department of Orthopedics, Utrecht, Netherlands
| | | | - Amir Bieber
- Emek Medical Center, Rheumatology Unit, Afula, Israel
| | - Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, Pavia, Italy
| | - David Kiefer
- Ruhr-Universitat Bochum, Rheumazentrum Ruhrgebiet, Bochum, Germany
| | | | - Reuven Mader
- Emek Medical Center, Rheumatology Unit, Afula, Israel
- Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Ruhr University Bochum, Rheumazentrum Ruhrgebiet, Herne, Germany
| | | | - Jácome Bruges-Armas
- Hospital de Santo Espirito da Ilha Terceira EPER, SEEBMO, Angra do Heroísmo, Portugal
- Comprehensive Health Research Centre, Hospital de Santo Espírito da Ilha Terceira, Lisbon, Portugal
| |
Collapse
|
27
|
Amorim MR, Aung O, Mokhlesi B, Polotsky VY. Leptin-mediated neural targets in obesity hypoventilation syndrome. Sleep 2022; 45:zsac153. [PMID: 35778900 PMCID: PMC9453616 DOI: 10.1093/sleep/zsac153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/20/2022] [Indexed: 07/30/2023] Open
Abstract
Obesity hypoventilation syndrome (OHS) is defined as daytime hypercapnia in obese individuals in the absence of other underlying causes. In the United States, OHS is present in 10%-20% of obese patients with obstructive sleep apnea and is linked to hypoventilation during sleep. OHS leads to high cardiorespiratory morbidity and mortality, and there is no effective pharmacotherapy. The depressed hypercapnic ventilatory response plays a key role in OHS. The pathogenesis of OHS has been linked to resistance to an adipocyte-produced hormone, leptin, a major regulator of metabolism and control of breathing. Mechanisms by which leptin modulates the control of breathing are potential targets for novel therapeutic strategies in OHS. Recent advances shed light on the molecular pathways related to the central chemoreceptor function in health and disease. Leptin signaling in the nucleus of the solitary tract, retrotrapezoid nucleus, hypoglossal nucleus, and dorsomedial hypothalamus, and anatomical projections from these nuclei to the respiratory control centers, may contribute to OHS. In this review, we describe current views on leptin-mediated mechanisms that regulate breathing and CO2 homeostasis with a focus on potential therapeutics for the treatment of OHS.
Collapse
Affiliation(s)
- Mateus R Amorim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - O Aung
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Babak Mokhlesi
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Vsevolod Y Polotsky
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Hughey CC, Puchalska P, Crawford PA. Integrating the contributions of mitochondrial oxidative metabolism to lipotoxicity and inflammation in NAFLD pathogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159209. [DOI: 10.1016/j.bbalip.2022.159209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/25/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022]
|
29
|
Wen Y, Liu Y, Huang Q, Farag MA, Li X, Wan X, Zhao C. Nutritional assessment models for diabetes and aging. FOOD FRONTIERS 2022. [DOI: 10.1002/fft2.168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Yuxi Wen
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Yuanyuan Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Qihui Huang
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Department of Analytical and Food Chemistry Universidade de Vigo, Nutrition and Bromatology Group, Faculty of Sciences Ourense Spain
| | - Mohamed A. Farag
- Pharmacognosy Department, College of Pharmacy Cairo University Cairo Egypt
| | - Xiaoqing Li
- School of Food Science and Engineering South China University of Technology Guangzhou China
| | - Xuzhi Wan
- College of Biosystem Engineering and Food Science Zhejiang University Hangzhou China
| | - Chao Zhao
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
30
|
Gvazava IG, Karimova MV, Vasiliev AV, Vorotelyak EA. Type 2 Diabetes Mellitus: Pathogenic Features and Experimental Models in Rodents. Acta Naturae 2022; 14:57-68. [PMID: 36348712 PMCID: PMC9611859 DOI: 10.32607/actanaturae.11751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is the most common endocrine disorder (90%) in the world; it has numerous clinical, immunological, and genetic differences from type 1 diabetes mellitus. The pathogenesis of T2DM is complex and not fully clear. To date, animal models remain the main tool by which to study the pathophysiology and therapy of T2DM. Rodents are considered the best choice among animal models, because they are characterized by a small size, short induction period, easy diabetes induction, and economic efficiency. This review summarizes data on experimental models of T2DM that are currently used, evaluates their advantages and disadvantages vis-a-vis research, and describes in detail the factors that should be taken into account when using these models. Selection of a suitable model for tackling a particular issue is not always trivial; it affects study results and their interpretation.
Collapse
Affiliation(s)
- I. G. Gvazava
- Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - M. V. Karimova
- Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - A. V. Vasiliev
- Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234 Russia
| | - E. A. Vorotelyak
- Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234 Russia
| |
Collapse
|
31
|
Greenman AC, Diffee GM, Power AS, Wilkins GT, Gold OMS, Erickson JR, Baldi JC. Treadmill running increases the calcium sensitivity of myofilaments in diabetic rats. J Appl Physiol (1985) 2022; 132:1350-1360. [PMID: 35482324 DOI: 10.1152/japplphysiol.00785.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cardiovascular benefits of regular exercise are unequivocal, yet patients with type 2 diabetes respond poorly to exercise due to a reduced cardiac reserve. The contractile response of diabetic cardiomyocytes to beta-adrenergic stimulation is attenuated, which may result in altered myofilament calcium sensitivity and post-translational modifications of cardiac troponin I (cTnI). Treadmill running increases myofilament calcium sensitivity in non‑diabetic rats, and thus we hypothesized that endurance training would increase calcium sensitivity of diabetic cardiomyocytes and alter site-specific phosphorylation of cTnI. Calcium sensitivity, or pCa50, was measured in Zucker Diabetic Fatty (ZDF) non-diabetic (nDM) and diabetic (DM) rat hearts after 8 weeks of either a sedentary (SED) or progressive treadmill running (TR) intervention. Skinned cardiomyocytes were connected to a capacitance-gauge transducer and a torque motor to measure force as a function of pCa (‑log[Ca2+]). Specific phospho-sites on cTnI and O‑GlcNAcylation were quantified by immunoblot and total protein phosphorylation by fluorescent gel staining (ProQ Diamond). The novel finding in this study was that training increased pCa50 in both DM and nDM cardiomyocytes (p = 0.009). Phosphorylation of cTnI amino acid residues Ser23/24, a crucial protein kinase A site, and Threonine (Thr)144, was lower in DM hearts, but there was no effect of training on site-specific phosphorylation. Additionally, total phosphorylation and O-GlcNAcylation levels were not different between SED and TR groups. These findings suggest that regular exercise may benefit the diabetic heart by specifically targeting myofilament contractile function.
Collapse
Affiliation(s)
- Angela Claire Greenman
- Department of Medicine, Otago Medical School, University of Otago, Dunedin, New Zealand.,Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Gary M Diffee
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI
| | - Amelia S Power
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Gerard T Wilkins
- Department of Medicine, Otago Medical School, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Olivia M S Gold
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Jeffrey R Erickson
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - James C Baldi
- Department of Medicine, Otago Medical School, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
32
|
Wang T, Tang X, Hu X, Wang J, Chen G. Reduction in the Dietary VA Status Prevents Type 2 Diabetes and Obesity in Zucker Diabetic Fatty Rats. Biomolecules 2022; 12:biom12040528. [PMID: 35454117 PMCID: PMC9032907 DOI: 10.3390/biom12040528] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023] Open
Abstract
We hypothesized that the vitamin A (VA) status regulates type 2 diabetes (T2D) development in Zucker diabetic fatty (ZDF) rats. Zucker Lean and ZDF rats at weaning were fed a VA deficient with basal fat (VAD-BF, no VA and 22.1% fat energy), VA marginal with BF (VAM-BF, 0.35 mg retinyl palmitate (RP)/kg), VA sufficient with BF (VAS-BF, 4.0 mg RP/kg), VAD with high fat (VAD-HF, 60% fat energy), VAM-HF or VAS-HF diet for 8 weeks, including an oral glucose tolerance test (OGTT) at week 7.5. The hepatic mRNA and proteins levels were determined using real-time PCR and Western blot, respectively. The VAD-BF/HF and VAM-BF/HF diets prevented peripheral hyperglycemia and attenuated obesity in ZDF rats, which occurred in the presence of the VAS-BF/HF diets. This lowered VA status reduced venous blood hyperglycemia, hyperinsulinemia and hyperlipidemia, and improved OGTT and homeostasis model assessment for insulin resistance results in ZDF rats. The expression levels of key hepatic genes for glucose and fat metabolism were regulated by VA status and dietary fat contents. An interaction between VA and HF condition was also observed. We conclude that the reduction in the dietary VA status in both BF and HF conditions prevents T2D and obesity in ZDF rats.
Collapse
Affiliation(s)
- Tiannan Wang
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, TN 37996, USA; (T.W.); (X.H.)
| | - Xia Tang
- College of Food Science and Technology, Hebei Agriculture University, Baoding 071001, China;
| | - Xinge Hu
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, TN 37996, USA; (T.W.); (X.H.)
| | - Jing Wang
- College of Pharmacy, South-Central University for Nationalities, Wuhan 430074, China;
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, TN 37996, USA; (T.W.); (X.H.)
- Correspondence: ; Tel.:+1-865-974-6257
| |
Collapse
|
33
|
Hossain MJ, Kendig MD, Letton ME, Morris MJ, Arnold R. Peripheral Neuropathy Phenotyping in Rat Models of Type 2 Diabetes Mellitus: Evaluating Uptake of the Neurodiab Guidelines and Identifying Future Directions. Diabetes Metab J 2022; 46:198-221. [PMID: 35385634 PMCID: PMC8987683 DOI: 10.4093/dmj.2021.0347] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/25/2022] [Indexed: 11/08/2022] Open
Abstract
Diabetic peripheral neuropathy (DPN) affects over half of type 2 diabetes mellitus (T2DM) patients, with an urgent need for effective pharmacotherapies. While many rat and mouse models of T2DM exist, the phenotyping of DPN has been challenging with inconsistencies across laboratories. To better characterize DPN in rodents, a consensus guideline was published in 2014 to accelerate the translation of preclinical findings. Here we review DPN phenotyping in rat models of T2DM against the 'Neurodiab' criteria to identify uptake of the guidelines and discuss how DPN phenotypes differ between models and according to diabetes duration and sex. A search of PubMed, Scopus and Web of Science databases identified 125 studies, categorised as either diet and/or chemically induced models or transgenic/spontaneous models of T2DM. The use of diet and chemically induced T2DM models has exceeded that of transgenic models in recent years, and the introduction of the Neurodiab guidelines has not appreciably increased the number of studies assessing all key DPN endpoints. Combined high-fat diet and low dose streptozotocin rat models are the most frequently used and well characterised. Overall, we recommend adherence to Neurodiab guidelines for creating better animal models of DPN to accelerate translation and drug development.
Collapse
Affiliation(s)
- Md Jakir Hossain
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Michael D. Kendig
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Meg E. Letton
- Department of Exercise Physiology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Ria Arnold
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
- Department of Exercise Physiology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
- Department of Exercise and Rehabilitation, School of Medical, Indigenous and Health Science, University of Wollongong, Wollongong, Australia
- Corresponding author: Ria Arnold https://orcid.org/0000-0002-7469-6587 Department of Exercise Physiology, School of Health Sciences, UNSW Sydney, Sydney, NSW 2052, Australia E-mail:
| |
Collapse
|
34
|
Wang AN, Carlos J, Fraser GM, McGuire JJ. Zucker Diabetic Sprague Dawley rat (ZDSD): type 2 diabetes translational research model. Exp Physiol 2022; 107:265-282. [PMID: 35178802 PMCID: PMC9314054 DOI: 10.1113/ep089947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 02/02/2022] [Indexed: 11/30/2022]
Abstract
New Findings What is the topic of this review? The Zucker Diabetic‐Sprague Dawley (ZDSD) rat is in the early adoption phase of use by researchers in the fields of diabetes, including prediabetes, obesity and metabolic syndrome. It is essential that physiology researchers choose preclinical models that model human type 2 diabetes appropriately and are aware of the limitations on experimental design. What advances does it highlight? Our review of the scientific literature finds that although sex, age and diets contribute to variability, the ZDSD phenotype and disease progression model the characteristics of humans who have prediabetes and diabetes, including co‐morbidities.
Abstract Type 2 diabetes (T2D) is a prevalent disease and a significant concern for global population health. For persons with T2D, clinical treatments target not only the characteristics of hyperglycaemia and insulin resistance, but also co‐morbidities, such as obesity, cardiovascular and renal disease, neuropathies and skeletal bone conditions. The Zucker Diabetic‐Sprague Dawley (ZDSD) rat is a rodent model developed for experimental studies of T2D. We reviewed the scientific literature to highlight the characteristics of T2D development and the associated phenotypes, such as metabolic syndrome, cardiovascular complications and bone and skeletal pathologies in ZDSD rats. We found that ZDSD phenotype characteristics are independent of leptin receptor signalling. The ZDSD rat develops prediabetes, then progresses to overt diabetes that is accelerated by introduction of a timed high‐fat diet. In male ZDSD rats, glycated haemoglobin (HbA1c) increases at a constant rate from 7 to >30 weeks of age. Diabetic ZDSD rats are moderately hypertensive compared with other rat strains. Diabetes in ZDSD rats leads to endothelial dysfunction in specific vasculatures, impaired wound healing, decreased systolic and diastolic cardiac function, neuropathy and nephropathy. Changes to bone composition and the skeleton increase the risk of bone fractures. Zucker Diabetic‐Sprague Dawley rats have not yet achieved widespread use by researchers. We highlight sex‐related differences in the ZDSD phenotype and gaps in knowledge for future studies. Overall, scientific data support the premise that the phenotype and disease progression in ZDSD rats models the characteristics in humans. We conclude that ZDSD rats are an advantageous model to advance understanding and discovery of treatments for T2D through preclinical research.
Collapse
Affiliation(s)
- Andrea N Wang
- Departments of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Joselia Carlos
- Departments of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Graham M Fraser
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland, Canada
| | - John J McGuire
- Departments of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.,Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
35
|
Carpéné C, Marti L, Morin N. Increased monoamine oxidase activity and imidazoline binding sites in insulin-resistant adipocytes from obese Zucker rats. World J Biol Chem 2022; 13:15-34. [PMID: 35126867 PMCID: PMC8790288 DOI: 10.4331/wjbc.v13.i1.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/09/2021] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Despite overt insulin resistance, adipocytes of genetically obese Zucker rats accumulate the excess of calorie intake in the form of lipids.
AIM To investigate whether factors can replace or reinforce insulin lipogenic action by exploring glucose uptake activation by hydrogen peroxide, since it is produced by monoamine oxidase (MAO) and semicarbazide-sensitive amine oxidase (SSAO) in adipocytes.
METHODS 3H-2-deoxyglucose uptake (2-DG) was determined in adipocytes from obese and lean rats in response to insulin or MAO and SSAO substrates such as tyramine and benzylamine. 14C-tyramine oxidation and binding of imidazolinic radioligands [3H-Idazoxan, 3H-(2-benzofuranyl)-2-imidazoline] were studied in adipocytes, the liver, and muscle. The influence of in vivo administration of tyramine + vanadium on glucose handling was assessed in lean and obese rats.
RESULTS 2-DG uptake and lipogenesis stimulation by insulin were dampened in adipocytes from obese rats, when compared to their lean littermates. Tyramine and benzylamine activation of hexose uptake was vanadate-dependent and was also limited, while MAO was increased and SSAO decreased. These changes were adipocyte-specific and accompanied by a greater number of imidazoline I2 binding sites in the obese rat, when compared to the lean. In vitro, tyramine precluded the binding to I2 sites, while in vivo, its administration together with vanadium lowered fasting plasma levels of glucose and triacylglycerols in obese rats.
CONCLUSION The adipocytes from obese Zucker rats exhibit increased MAO activity and imidazoline binding site number. However, probably as a consequence of SSAO down-regulation, the glucose transport stimulation by tyramine is decreased as much as that of insulin in these insulin-resistant adipocytes. The adipocyte amine oxidases deserve more studies with respect to their putative contribution to the management of glucose and lipid handling.
Collapse
Affiliation(s)
- Christian Carpéné
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, Toulouse 31342, France
| | - Luc Marti
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, Toulouse 31342, France
| | - Nathalie Morin
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, Toulouse 31342, France
- Faculté de Pharmacie de Paris, Paris University, Paris 75270, France
| |
Collapse
|
36
|
Jankauskas SS, Kansakar U, Varzideh F, Wilson S, Mone P, Lombardi A, Gambardella J, Santulli G. Heart failure in diabetes. Metabolism 2021; 125:154910. [PMID: 34627874 PMCID: PMC8941799 DOI: 10.1016/j.metabol.2021.154910] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Heart failure and cardiovascular disorders represent the leading cause of death in diabetic patients. Here we present a systematic review of the main mechanisms underlying the development of diabetic cardiomyopathy. We also provide an excursus on the relative contribution of cardiomyocytes, fibroblasts, endothelial and smooth muscle cells to the pathophysiology of heart failure in diabetes. After having described the preclinical tools currently available to dissect the mechanisms of this complex disease, we conclude with a section on the most recent updates of the literature on clinical management.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Urna Kansakar
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Fahimeh Varzideh
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Scott Wilson
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Angela Lombardi
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Jessica Gambardella
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; International Translational Research and Medical Education (ITME), Department of Advanced Biomedical Science, "Federico II" University, 80131 Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; International Translational Research and Medical Education (ITME), Department of Advanced Biomedical Science, "Federico II" University, 80131 Naples, Italy.
| |
Collapse
|
37
|
Glenny EM, Coleman MF, Giles ED, Wellberg EA, Hursting SD. Designing Relevant Preclinical Rodent Models for Studying Links Between Nutrition, Obesity, Metabolism, and Cancer. Annu Rev Nutr 2021; 41:253-282. [PMID: 34357792 PMCID: PMC8900211 DOI: 10.1146/annurev-nutr-120420-032437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Diet and nutrition are intricately related to cancer prevention, growth, and treatment response. Preclinical rodent models are a cornerstone to biomedical research and remain instrumental in our understanding of the relationship between cancer and diet and in the development of effective therapeutics. However, the success rate of translating promising findings from the bench to the bedside is suboptimal. Well-designed rodent models will be crucial to improving the impact basic science has on clinical treatment options. This review discusses essential experimental factors to consider when designing a preclinical cancer model with an emphasis on incorporatingthese models into studies interrogating diet, nutrition, and metabolism. The aims of this review are to (a) provide insight into relevant considerations when designing cancer models for obesity, nutrition, and metabolism research; (b) identify common pitfalls when selecting a rodent model; and (c) discuss strengths and limitations of available preclinical models.
Collapse
Affiliation(s)
- Elaine M Glenny
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Michael F Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Erin D Giles
- Department of Nutrition, Texas A&M University, College Station, Texas 77843, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, USA
| |
Collapse
|
38
|
Greenman AC, Diffee GM, Power AS, Wilkins GT, Gold OMS, Erickson JR, Baldi JC. Increased myofilament calcium sensitivity is associated with decreased cardiac troponin I phosphorylation in the diabetic rat heart. Exp Physiol 2021; 106:2235-2247. [PMID: 34605091 DOI: 10.1113/ep089730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/23/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? In Zucker Diabetic Fatty rats, does cardiomyocyte myofilament function change through the time course of diabetes and what are the mechanisms behind alterations in calcium sensitivity? What is the main finding and its importance? Zucker Diabetic Fatty rats had increased myofilament calcium sensitivity and reduced phosphorylation at cardiac troponin I without differential O-GlcNAcylation. ABSTRACT The diabetic heart has impaired systolic and diastolic function independent of other comorbidities. The availability of calcium is altered, but does not fully explain the cardiac dysfunction seen in the diabetic heart. Thus, we explored if myofilament calcium regulation of contraction is altered while also categorizing the levels of phosphorylation and O-GlcNAcylation in the myofilaments. Calcium sensitivity (pCa50 ) was measured in Zucker Diabetic Fatty (ZDF) rat hearts at the initial stage of diabetes (12 weeks old) and after 8 weeks of uncontrolled hyperglycaemia (20 weeks old) and in non-diabetic (nDM) littermates. Skinned cardiomyocytes were connected to a capacitance-gauge transducer and a torque motor to measure force as a function of pCa (-log[Ca2+ ]). Fluorescent gel stain (ProQ Diamond) was used to measure total protein phosphorylation. Specific phospho-sites on cardiac troponin I (cTnI) and total cTnI O-GlcNAcylation were quantified using immunoblot. pCa50 was greater in both 12- and 20-week-old diabetic (DM) rats compared to nDM littermates (P = 0.0001). Total cTnI and cTnI serine 23/24 phosphorylation were lower in DM rats (P = 0.003 and P = 0.01, respectively), but cTnI O-GlcNAc protein expression was not different. pCa50 is greater in DM rats and corresponds with an overall reduction in cTnI phosphorylation. These findings indicate that myofilament calcium sensitivity is increased and cTnI phosphorylation is reduced in ZDF DM rats and suggests an important role for cTnI phosphorylation in the DM heart.
Collapse
Affiliation(s)
- Angela C Greenman
- Department of Medicine, Otago Medical School, University of Otago, Dunedin, New Zealand.,Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Gary M Diffee
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, USA
| | - Amelia S Power
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Gerard T Wilkins
- Department of Medicine, Otago Medical School, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Olivia M S Gold
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - Jeffrey R Erickson
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| | - James C Baldi
- Department of Medicine, Otago Medical School, University of Otago, Dunedin, New Zealand.,HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
39
|
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: 113] [Impact Index Per Article: 28.3] [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
|
40
|
Marques Miranda C, de Lima Campos M, Leite-Almeida H. Diet, body weight and pain susceptibility - A systematic review of preclinical studies. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100066. [PMID: 34195483 PMCID: PMC8237587 DOI: 10.1016/j.ynpai.2021.100066] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/26/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
Obesity has been associated with increased chronic pain susceptibility but causes are unclear. In this review, we systematize and analyze pain outcomes in rodent models of obesity as these can be important tools for mechanistic studies. Studies were identified using MEDLINE/PubMed and Scopus databases using the following search query: (((pain) OR (nociception)) AND (obesity)) AND (rat OR (mouse) OR (rodent))). From each eligible record we extracted the following data: species, strain, sex, pain/obesity model and main behavioral readouts. Out of 695 records 33 were selected for inclusion. 27 studies assessed nociception/acute pain and 17 studies assessed subacute or chronic pain. Overall genetic and dietary models overlapped in pain-related outcomes. Most acute pain studies reported either decreased or unaltered responses to noxious painful stimuli. However, decreased thresholds to mechanical innocuous stimuli, i.e. allodynia, were frequently reported. In most studies using subacute and chronic pain models, namely of subcutaneous inflammation, arthritis and perineural inflammation, decreased thresholds and/or prolonged pain manifestations were reported in obesity models. Strain comparisons and longitudinal observations indicate that genetic factors and the time course of the pathology might account for some of the discrepancies observed across studies. Two studies reported increased pain in animals subjected to high fat diet in the absence of weight gain. Pain-related outcomes in experimental models and clinical obesity are aligned indicating that the rodent can be an useful tool to study the interplay between diet, obesity and pain. In both cases weight gain might represent only a minor contribution to abnormal pain manifestation.
Collapse
Affiliation(s)
- Carolina Marques Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mariana de Lima Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Hugo Leite-Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
41
|
Hsieh PS, Ho HH, Tsao SP, Hsieh SH, Lin WY, Chen JF, Kuo YW, Tsai SY, Huang HY. Multi-strain probiotic supplement attenuates streptozotocin-induced type-2 diabetes by reducing inflammation and β-cell death in rats. PLoS One 2021; 16:e0251646. [PMID: 34166387 PMCID: PMC8224959 DOI: 10.1371/journal.pone.0251646] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
Probiotics are health beneficial bacterial populations colonizing the human gut and skin. Probiotics are believed to be involved in immune system regulation, gut microbiota stabilization, prevention of infectious diseases, and adjustments of host metabolic activities. Probiotics such as Lactobacillus and Bifidobacterium affect glycemic levels, blood lipids, and protein metabolism. However, the interactions between probiotics and metabolic diseases as well as the underlying mechanisms remain unclear. We used streptozotocin (STZ)-induced diabetic animal models to study the effect of ProbiogluTM, a multi-strain probiotic supplement including Lactobaccilus salivarius subsp. salicinius AP-32, L. johnsonii MH-68, L. reuteri GL-104, and Bifidobacterium animalis subsp. lactis CP-9, on the regulation of physiochemical parameters related to type-2 diabetes. Experimental rats were randomly assigned into five groups, control group, streptozotocin (STZ)-treated rats (STZ group), STZ + 1× ProbiogluTM group, STZ + 5× ProbiogluTM group, and STZ + 10× ProbiogluTM group, and physiological data were measured at weeks 0, 2, 4, 6, and 8. Our results indicate that supplementation with ProbiogluTM significantly improved glucose tolerance, glycemic levels, insulin levels, and insulin resistance (HOMA-IR). Furthermore, we observed reduction in urea and blood lipid levels, including low-density lipoprotein (LDL), triglycerides (TG), and total cholesterol (TC). ProbiogluTM administration increased the β-cell mass in STZ-induced diabetic animal models, whereas it reduced the levels of proinflammatory cytokines TNF-α, IL-6, and IL-1β. In addition, the enhancement of oxidative stress biomarkers and superoxide dismutase (SOD) activities was associated with a decrease in malondialdehyde (MDA) levels. We conclude that ProbiogluTM attenuates STZ-induced type-2 diabetes by protecting β-cells, stabilizing glycemic levels, and reducing inflammation. Among all probiotic treating groups, the 10×ProbiogluTM treatment revealed the best results. However, these experimental results still need to be validated by different animal models of type-2 diabetes and human clinical trials in the future.
Collapse
Affiliation(s)
- Pei-Shan Hsieh
- Functional Research Division, Department of Research and Design, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Hsieh-Hsun Ho
- Functional Research Division, Department of Research and Design, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Shu Ping Tsao
- Department of Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Shih-Hung Hsieh
- Functional Research Division, Department of Research and Design, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Wen-Yang Lin
- Functional Research Division, Department of Research and Design, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Jui-Fen Chen
- Functional Research Division, Department of Research and Design, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Yi-Wei Kuo
- Functional Research Division, Department of Research and Design, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Shin-Yu Tsai
- Product Division, Department of Research and Design Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Hui-Yu Huang
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
42
|
Leptin Receptors Are Not Required for Roux-en-Y Gastric Bypass Surgery to Normalize Energy and Glucose Homeostasis in Rats. Nutrients 2021; 13:nu13051544. [PMID: 34064308 PMCID: PMC8147759 DOI: 10.3390/nu13051544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022] Open
Abstract
Sensitization to the adipokine leptin is a promising therapeutic strategy against obesity and its comorbidities and has been proposed to contribute to the lasting metabolic benefits of Roux-en-Y gastric bypass (RYGB) surgery. We formally tested this idea using Zucker fatty fa/fa rats as an established genetic model of obesity, glucose intolerance, and fatty liver due to leptin receptor deficiency. We show that the changes in body weight in these rats following RYGB largely overlaps with that of diet-induced obese Wistar rats with intact leptin receptors. Further, food intake and oral glucose tolerance were normalized in RYGB-treated Zucker fatty fa/fa rats to the levels of lean Zucker fatty fa/+ controls, in association with increased glucagon-like peptide 1 (GLP-1) and insulin release. In contrast, while fatty liver was also normalized in RYGB-treated Zucker fatty fa/fa rats, their circulating levels of the liver enzyme alanine aminotransferase (ALT) remained elevated at the level of obese Zucker fatty fa/fa controls. These findings suggest that the leptin system is not required for the normalization of energy and glucose homeostasis associated with RYGB, but that its potential contribution to the improvements in liver health postoperatively merits further investigation.
Collapse
|
43
|
Grattan DR, Andrews ZB. Insulin as a neuroendocrine hormone. J Neuroendocrinol 2021; 33:e12966. [PMID: 33786903 DOI: 10.1111/jne.12966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
The year 2021 marks 100 years since the discovery of insulin and this Special Issue of the Journal of Neuroendocrinology was conceived as a way to mark that historic breakthrough. The discovery of insulin and its subsequent use in the treatment of diabetes is one of the most striking success stories in biomedical research. From a neuroendocrinology perspective, the recognition that insulin also exerts widespread and varied actions in the brain is more recent, but potentially also of equal importance with relevance for conditions ranging from obesity to dementia. The reviews contained in this Special Issue were selected to cover the range of known actions of insulin in neuroendocrine function, and also to highlight areas where further understanding of insulin actions in the brain hold great promise for further improvements in human health.
Collapse
Affiliation(s)
- David R Grattan
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Zane B Andrews
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
44
|
Abstract
Diabetic heart disease is a growing and important public health risk. Apart from the risk of coronary artery disease or hypertension, diabetes mellitus (DM) is a well-known risk factor for heart failure in the form of diabetic cardiomyopathy (DiaCM). Currently, DiaCM is defined as myocardial dysfunction in patients with DM in the absence of coronary artery disease and hypertension. The underlying pathomechanism of DiaCM is partially understood, but accumulating evidence suggests that metabolic derangements, oxidative stress, increased myocardial fibrosis and hypertrophy, inflammation, enhanced apoptosis, impaired intracellular calcium handling, activation of the renin-angiotensin-aldosterone system, mitochondrial dysfunction, and dysregulation of microRNAs, among other factors, are involved. Numerous animal models have been used to investigate the pathomechanisms of DiaCM. Despite some limitations, animal models for DiaCM have greatly advanced our understanding of pathomechanisms and have helped in the development of successful disease management strategies. In this review, we summarize the current pathomechanisms of DiaCM and provide animal models for DiaCM according to its pathomechanisms, which may contribute to broadening our understanding of the underlying mechanisms and facilitating the identification of possible new therapeutic targets.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Corresponding authors: Wang-Soo Lee https://orcid.org/0000-0002-8264-0866 Division of Cardiology, Department of Internal Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul 06973, Korea E-mail:
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Corresponding authors: Wang-Soo Lee https://orcid.org/0000-0002-8264-0866 Division of Cardiology, Department of Internal Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul 06973, Korea E-mail:
| |
Collapse
|
45
|
Cellular and Molecular Players in the Interplay between Adipose Tissue and Breast Cancer. Int J Mol Sci 2021; 22:ijms22031359. [PMID: 33572982 PMCID: PMC7866411 DOI: 10.3390/ijms22031359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence and severity of obesity are rising in most of the world. In addition to metabolic disorders, obesity is associated with an increase in the incidence and severity of a variety of types of cancer, including breast cancer (BC). The bidirectional interaction between BC and adipose cells has been deeply investigated, although the molecular and cellular players involved in these mechanisms are far from being fully elucidated. Here, we review the current knowledge on these interactions and describe how preclinical research might be used to clarify the effects of obesity over BC progression and morbidity, with particular attention paid to promising therapeutic interventions.
Collapse
|
46
|
Kato Y, Sakoh M, Nagai T, Yoshida A, Ishida H, Inoue N, Yanagita T, Nagao K. Ozonated Olive Oil Alleviates Hepatic Steatosis in Obese Zucker (<i>fa/fa</i>) Rats. J Oleo Sci 2021; 71:599-607. [DOI: 10.5650/jos.ess21377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Masami Sakoh
- Department of Biological Resource Science, Saga University
| | | | | | | | - Nao Inoue
- Department of Biological Resource Science, Saga University
| | | | - Koji Nagao
- Department of Biological Resource Science, Saga University
| |
Collapse
|
47
|
Krskova K, Balazova L, Dobrocsyova V, Olszanecki R, Suski M, Chai SY, Zorad Š. Insulin-Regulated Aminopeptidase Inhibition Ameliorates Metabolism in Obese Zucker Rats. Front Mol Biosci 2020; 7:586225. [PMID: 33344504 PMCID: PMC7746680 DOI: 10.3389/fmolb.2020.586225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
The aim of our study was to determine the influence of inhibition of insulin-regulated aminopeptidase/oxytocinase (IRAP) on glucose tolerance and metabolism of skeletal muscle and visceral adipose tissue in obese Zucker rats. Obese Zucker rats administered with IRAP inhibitor-HFI-419 at a dose of 29 μg/100 g BW/day by osmotic minipumps implanted subcutaneously for 2 weeks. Two-hour intraperitoneal glucose tolerance test (ipGTT) was performed in fasting rats. Plasma oxytocin levels were measured by enzyme immunoassay after plasma extraction. In the musculus quadriceps and epididymal adipose tissue, the expression of factors affecting tissue oxidative status and metabolism was determined by real-time qPCR and/or Western blot analysys. The plasma and tissue enzymatic activities were determined by colorimetric or fluorometric method. Circulated oxytocin levels in obese animals strongly tended to increase after HFI-419 administration. This was accompanied by significantly improved glucose utilization during ipGTT and decreased area under the curve (AUC) for glucose. In skeletal muscle IRAP inhibitor treatment up-regulated enzymes of antioxidant defense system - superoxide dismutase 1 and 2 and improved insulin signal transduction pathway. HFI-419 increased skeletal muscle aminopeptidase A expression and activity and normalized its plasma levels in obese animals. In epididymal adipose tissue, gene expression of markers of inflammation and adipocyte hypertrophy was down-regulated in obese rats after HFI-419 treatment. Our results demonstrate that IRAP inhibition improves whole-body glucose tolerance in insulin-resistant Zucker fatty rats and that this metabolic effect of HFI-419 involves ameliorated redox balance in skeletal muscle.
Collapse
Affiliation(s)
- Katarina Krskova
- Institute of Experimental Endocrinology, Biomedical Research Center, Department of Endocrine Regulations and Psychofarmacology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Balazova
- Institute of Experimental Endocrinology, Biomedical Research Center, Department of Endocrine Regulations and Psychofarmacology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Viktoria Dobrocsyova
- Institute of Experimental Endocrinology, Biomedical Research Center, Department of Endocrine Regulations and Psychofarmacology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Rafal Olszanecki
- Department of Pharmacology, Jagiellonian University Medical College, Cracow, Poland
| | - Maciej Suski
- Department of Pharmacology, Jagiellonian University Medical College, Cracow, Poland
| | - Siew Yeen Chai
- Monash Biomedicine Discovery Institute, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Štefan Zorad
- Institute of Experimental Endocrinology, Biomedical Research Center, Department of Endocrine Regulations and Psychofarmacology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
48
|
Martin WP, le Roux CW, Docherty NG. Impact of Metabolic Surgery on Renal Injury in Pre-Clinical Models of Diabetic Kidney Disease. Nephron Clin Pract 2020; 145:585-594. [PMID: 33264793 DOI: 10.1159/000511790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 09/17/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Surgical approaches to the treatment of obesity and type 2 diabetes, most notably the Roux-en-Y gastric bypass (RYGB) procedure, have been shown to be renoprotective, reducing the incidence of albuminuria and end-stage kidney disease over 15- to 20-year follow-up in patients with obesity. The tissue level effects of metabolic surgery on the diabetic kidney are not easily interrogated in clinical samples. However, elucidation of the cellular and molecular basis for the renoprotective effects of metabolic surgery is now emerging from a body of pre-clinical work in rodent models of diabetic kidney disease (DKD). SUMMARY Experimental metabolic surgery (RYGB, sleeve gastrectomy [SG], Roux-en-Y oesophagojejunostomy, and duodenojejunal bypass) exerts a pronounced albuminuria-lowering effect in rat models of DKD. Following RYGB in the Zucker diabetic fatty rat, glomerular histology is improved as demonstrated by reductions in podocyte stress, glomerulomegaly, and glomerulosclerosis. Glomerular ultrastructure improves after RYGB and after SG, manifested by quantifiable reductions in podocyte foot process effacement. The transcriptional programme underpinning these structural improvements has been characterized at the pathway level using RNA sequencing and is associated with a significant reduction in the activation of inflammatory and fibrotic responses. Key Messages: Experimental metabolic surgery reduces biochemical, histological, and molecular indices of DKD. These pre-clinical data support a growing interest in the potential utility of metabolic surgery as a therapeutic approach to slow renal functional decline in patients with obesity and DKD.
Collapse
Affiliation(s)
- William P Martin
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland,
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland.,Division of Investigative Science, Imperial College London, London, United Kingdom
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
49
|
Bioactive Agent Discovery from the Natural Compounds for the Treatment of Type 2 Diabetes Rat Model. Molecules 2020; 25:molecules25235713. [PMID: 33287318 PMCID: PMC7731446 DOI: 10.3390/molecules25235713] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/27/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022] Open
Abstract
Diabetes mellitus is a well-known chronic metabolic disease that poses a long-term threat to human health and is characterized by a relative or absolute lack of insulin, resulting in hyperglycemia. Type 2 diabetes mellitus (T2DM) typically affects many metabolic pathways, resulting in β-cell dysfunction, insulin resistance, abnormal blood glucose levels, inflammatory processes, excessive oxidative reactions, and impaired lipid metabolism. It also leads to diabetes-related complications in many organ systems. Antidiabetic drugs have been approved for the treatment of hyperglycemia in T2DM; these are beneficial for glucose metabolism and promote weight loss, but have the risk of side effects, such as nausea or an upset stomach. A wide range of active components, derived from medicinal plants, such as alkaloids, flavonoids, polyphenol, quinones, and terpenoids may act as alternative sources of antidiabetic agents. They are usually attributed to improvements in pancreatic function by increasing insulin secretions or by reducing the intestinal absorption of glucose. Ease of availability, low cost, least undesirable side effects, and powerful pharmacological actions make plant-based preparations the key player of all available treatments. Based on the study of therapeutic reagents in the pathogenesis of humans, we use the appropriate animal models of T2DM to evaluate medicinal plant treatments. Many of the rat models have characteristics similar to those in humans and have the advantages of ease of genetic manipulation, a short breeding span, and access to physiological and invasive testing. In this review, we summarize the pathophysiological status of T2DM rat models and focus on several bioactive compounds from herbal medicine with different functional groups that exhibit therapeutic potential in the T2DM rat models, in turn, may guide future approach in treating diabetes with natural drugs.
Collapse
|
50
|
Pandey S, Dvorakova MC. Future Perspective of Diabetic Animal Models. Endocr Metab Immune Disord Drug Targets 2020; 20:25-38. [PMID: 31241444 PMCID: PMC7360914 DOI: 10.2174/1871530319666190626143832] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/06/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
Abstract
Objective The need of today’s research is to develop successful and reliable diabetic animal models for understanding the disease susceptibility and pathogenesis. Enormous success of animal models had already been acclaimed for identifying key genetic and environmental factors like Idd loci and effects of microorganisms including the gut microbiota. Furthermore, animal models had also helped in identifying many therapeutic targets and strategies for immune-intervention. In spite of a quite success, we have acknowledged that many of the discovered immunotherapies are working on animals and did not have a significant impact on human. Number of animal models were developed in the past to accelerate drug discovery pipeline. However, due to poor initial screening and assessment on inequivalent animal models, the percentage of drug candidates who succeeded during clinical trials was very low. Therefore, it is essential to bridge this gap between pre-clinical research and clinical trial by validating the existing animal models for consistency. Results and Conclusion In this review, we have discussed and evaluated the significance of animal models on behalf of published data on PUBMED. Amongst the most popular diabetic animal models, we have selected six animal models (e.g. BioBreeding rat, “LEW IDDM rat”, “Nonobese Diabetic (NOD) mouse”, “STZ RAT”, “LEPR Mouse” and “Zucker Diabetic Fatty (ZDF) rat” and ranked them as per their published literature on PUBMED. Moreover, the vision and brief imagination for developing an advanced and robust diabetic model of 21st century was discussed with the theme of one mice-one human concept including organs-on-chips.
Collapse
Affiliation(s)
- Shashank Pandey
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Magdalena C Dvorakova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.,Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|