1
|
Yao TF, Wang ZY, Sun L, Yu SX, Yu HD, Yang ZZ, Li WZ, Niu L, Sun D, Shi YH, Li JQ, Liu WQ, Liu XZ, Zuo ZF. DNMT3b-mediated CpA methylation facilitates REST binding and gene silencing and exacerbates hippocampal demyelination in diabetic mice. J Biol Chem 2024:108137. [PMID: 39730060 DOI: 10.1016/j.jbc.2024.108137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024] Open
Abstract
The remyelination process within the diabetes mellitus (DM) brain is inhibited, and dynamic interactions between DNA methylation and transcription factors are critical for this process. Repressor element-1 silencing transcription factor (REST) is a major regulator of oligodendrocyte differentiation, and the role of REST on DM remyelination remains to be investigated. Here, we investigated the effects of REST and DNA methylation on DM remyelination and explored the underlying mechanisms. In this study, using a diabetic mouse model, we found that myelin damage preceded neuronal damage and caused cognitive impairment in DM mice. Inhibition of REST by X5050 and DNMT3b by Naomycin A promoted myelin regeneration in the hippocampus and ameliorated cognitive deficits in DM mice. In addition, CpA methylation of the RE-1 locus of the CNTN1 gene was able to increase the binding capacity of REST. We also observed that CNTN1 promotes oligodendrocyte maturation, facilitates the ratio of microglia to pro-regenerative phenotype as well as enhances the ability of microglia to remove myelin debris. Our findings suggest that that REST and DNMT3b expression inhibit CNTN1 expression and exacerbate myelin damage. This mechanism of gene silencing may be associated with DNMT3b-mediated CpA methylation of the REST binding site in the promoter region of the CNTN1 gene. We also identified a role for CNTN1 in promoting oligodendrocyte precursor cell maturation and myelin debris removal during remyelination.
Collapse
Affiliation(s)
- Tie-Feng Yao
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Zhi-Yun Wang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China
| | - Lu Sun
- Department of Pathology, Jinzhou Medical University, Jinzhou, China
| | - Sheng-Xue Yu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Hong Dan Yu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Zheng-Zhong Yang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Wan-Ze Li
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Lin Niu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Die Sun
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Ya-Hui Shi
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Jun-Qi Li
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Wen-Qiang Liu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China
| | - Xue-Zheng Liu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China.
| | - Zhong-Fu Zuo
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, China; Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
2
|
Merlin E, Salio C, Ferrini F. Painful Diabetic Neuropathy: Sex-Specific Mechanisms and Differences from Animal Models to Clinical Outcomes. Cells 2024; 13:2024. [PMID: 39682771 DOI: 10.3390/cells13232024] [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: 11/11/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Diabetes is a chronic and progressive disease associated with high blood glucose levels. Several co-morbidities arise from diabetes, the most common and severe one is diabetic neuropathy whose symptoms also include pain hypersensitivity. Currently, there are no effective therapies to counteract painful diabetic neuropathy or slow down the progression of the disease, and the underlying mechanisms are yet to be fully understood. Emerging data in recent decades have provided compelling evidence that the molecular and cellular mechanisms underlying chronic pain are different across the sexes. Interestingly, relevant differences have also been observed in the course and clinical presentation of painful diabetic neuropathy in humans. Here, we reviewed the current state of the art on sex differences in diabetic neuropathy, from animal models to clinical data. Comparing the output of both preclinical and clinical studies is necessary for properly orienting future choices in pain research, refining animal models, and interpreting clinical data. The identification of sex-specific mechanisms may help to develop more targeted therapies to counteract pain symptoms in diabetes.
Collapse
Affiliation(s)
- Emma Merlin
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| | - Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, TO, Italy
- Department of Psychiatry and Neurosciences, Université Laval, Québec, QC G1K 7P4, Canada
| |
Collapse
|
3
|
Mani AM, Lamin V, Peach RC, Friesen EH, Wong T, Singh MV, Dokun AO. miRNA-6236 Regulation of Postischemic Skeletal Muscle Angiogenesis. J Am Heart Assoc 2024:e035923. [PMID: 39604034 DOI: 10.1161/jaha.124.035923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Peripheral arterial disease affects >200 million people worldwide and is characterized by impaired blood flow to the lower extremities. There are no effective medical treatments available. Using the mouse hind-limb ischemia model and miRNA sequencing, we identified a novel miRNA, miR-6236, whose expression significantly elevated in ischemic mouse limbs compared with nonischemic limbs. The role of miR-6236 in general or in postischemic angiogenesis is not known. Here we describe its role using in vivo and in vitro models of peripheral arterial disease. METHODS AND RESULTS In primary mouse and human endothelial cells, we studied the effect of simulated ischemia on miR-6236 expression and assessed its role in cell viability, apoptosis, migration, and tube formation during ischemia. Furthermore, we developed miR-6236 null mice and tested its role in postischemic perfusion recovery using the hind-limb ischemia model. Lastly, using bioinformatics and gene expression analysis, we identified putative angiogenic miR-6236 targets. In vitro simulated ischemia-enhanced miR-6236 expression in mouse and human endothelial cells, whereas its inhibition improved viability, migration, tube formation, and reduced apoptosis. In vivo ischemic mouse skeletal muscle tissue showed higher miR-6236 expression compared with nonischemic muscles. Loss of miR-6236 improved impaired postischemic perfusion recovery and poor angiogenesis associated with streptozotocin-induced diabetes in mice. Six of the 8 miR-6236 predicted angiogenic target mRNAs showed expression consistent with regulation by miR-6236 in ischemic skeletal muscle. CONCLUSIONS Our results show for the first time that miR-6236 plays a key role in regulating postischemic perfusion recovery and angiogenesis.
Collapse
Affiliation(s)
- Arul M Mani
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Victor Lamin
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ronan C Peach
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Eli H Friesen
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Thomas Wong
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Madhu V Singh
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
| | - Ayotunde O Dokun
- Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa Iowa City IA USA
- Fraternal Order of Eagles Diabetes Research Centre, Carver College of Medicine University of Iowa Iowa City IA USA
| |
Collapse
|
4
|
Kusumo LE, Gilley-Connor KR, Johnson MG, Hall GM, Gillett AE, McCready RG, Vichaya EG. Hyperglycemia sensitizes female mice to stress-induced depressive-like behavior in an inflammation-independent manner. Psychoneuroendocrinology 2024; 169:107151. [PMID: 39098101 DOI: 10.1016/j.psyneuen.2024.107151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Depression is a multifaceted disorder that represents one of the most common causes of disability. The risk for developing depression is increased in women and among individuals with chronic diseases. For example, individuals in the United States with diabetes mellitus (DM) are at a twofold increased risk of developing depression compared to the general population and approximately one-quarter of women with diabetes have comorbid depression. The neurobiological mechanisms underlying this association between diabetes and depression is not fully understood and is particularly under-investigated in female models. We sought to explore the role of neuroinflammation in diabetes-induced depression in a female mouse model of hyperglycemia. METHODS To this end, we utilized female C57BL/6 J mice to (1) characterize the depressive-like symptoms in response to 75 mg/kg/day dose of streptozotocin (STZ) over 5 days, a dose reported to induce hyperglycemia in female mice (n=20), (2) determine if female hyperglycemic mice are sensitized to unpredictable chronic mild stress (UCMS)-induced depressive-like behavior and neuroinflammation (n=28), and (3) investigate if female hyperglycemic mice are primed to respond to a subthreshold dose of lipopolysaccharide (LPS), an acute inflammatory challenge (n=21). RESULTS Our results demonstrate that female mice exhibit robust hyperglycemia but limited evidence of depressive-like behavior in response to 75 mg/kg STZ. Additionally, we observe that healthy female mice have limited response to our stress protocol; however, hyperglycemic mice display increased stress-sensitivity as indicated by increased immobility in the forced swim test. While STZ mice show evidence of mild neuroinflammation, this effect was blunted by stress. Further, STZ mice failed to display a sensitization to inflammation-induced depressive-like behavior. CONCLUSION We interpret this data to indicate that while STZ-induced hyperglycemia does increase vulnerability to stress-induced depressive-like behavior, this effect is not a consequence of neuroinflammatory priming. Future studies will seek to better understand the mechanisms underlying this sensitization.
Collapse
Affiliation(s)
- Laura E Kusumo
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Kayla R Gilley-Connor
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Madilyn G Johnson
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Grace M Hall
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Avery E Gillett
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Riley G McCready
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Elisabeth G Vichaya
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States.
| |
Collapse
|
5
|
Chen Y, Schlotterer A, Lin J, Dietrich N, Fleming T, Lanzinger S, Holl RW, Hammes HP. Sex differences in the development of experimental diabetic retinopathy. Sci Rep 2024; 14:22812. [PMID: 39354039 PMCID: PMC11445250 DOI: 10.1038/s41598-024-73279-x] [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: 03/02/2024] [Accepted: 09/15/2024] [Indexed: 10/03/2024] Open
Abstract
This study aimed to characterize the role of female sex in the pathogenesis of diabetic retinopathy. In the retinae of female Ins2Akita-diabetic mice (F-IA), ovariectomized female Ins2Akita-diabetic mice (F-IA/OVX), male Ins2Akita-diabetic mice (M-IA), and female STZ-diabetic mice (F-STZ), the formation of reactive metabolites and post-translational modifications, damage to the neurovascular unit, and expression of cellular stress response genes were analyzed. Compared to the male diabetic retina, the concentrations of the glycation adduct fructosyl-lysine, the Maillard product 3-deoxyglucosone, and the reactive metabolite methylglyoxal were significantly reduced in females. In females, there was also less evidence of diabetic damage to the neurovascular unit, as shown by decreased pericyte loss and reduced microglial activation. In the male diabetic retina, the expression of several members of the crystallin gene family (Cryab, Cryaa, Crybb2, Crybb1, and Cryba4) was increased. Clinical data from type 1 diabetic females showed that premenopausal women had a significantly lower prevalence of diabetic retinopathy compared to postmenopausal women stratified for disease duration and glycemic control. These data emphasize the importance of estradiol in protecting the diabetic retina and highlight the pathogenic relevance of sex in diabetic retinopathy.
Collapse
Affiliation(s)
- Ying Chen
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Andrea Schlotterer
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jihong Lin
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nadine Dietrich
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Stefanie Lanzinger
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Reinhard W Holl
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Hans-Peter Hammes
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
6
|
Talbot SR, Heider M, Wirth M, Jörns A, Naujok O. Exploring dose-response variability and relative severity assessment in STZ-induced diabetes male NSG mice. Sci Rep 2024; 14:16559. [PMID: 39020093 PMCID: PMC11255292 DOI: 10.1038/s41598-024-67490-z] [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: 03/12/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024] Open
Abstract
NSG mice are among the most immunodeficient mouse model being used in various scientific branches. In diabetelogical research diabetic NSG mice are an important asset as a xenotransplantation model for human pancreatic islets or pluripotent stem cell-derived islets. The treatment with the beta cell toxin streptozotocin is the standard procedure for triggering a chemically induced diabetes. Surprisingly, little data has been published about the reproducibility, stress and animal suffering in these NSG mice during diabetes induction. The 3R rules, however, are a constant reminder that existing methods can be further refined to minimize suffering. In this pilot study the dose-response relationship of STZ in male NSG mice was investigated and additionally animal suffering was charted by applying the novel 'Relative Severity Assessment' algorithm. By this we successfully explored an STZ dose that reliably induced diabetes while reduced stress and pain to the animals to a minimum using evidence-based and objective parameters rather than criteria that might be influenced by human bias.
Collapse
Affiliation(s)
| | - Miriam Heider
- Institute for Laboratory Animal Science, Hannover, Germany
| | - Martin Wirth
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Ortwin Naujok
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
7
|
Umek N, Pušnik L, Ugwoke CK, Šink Ž, Horvat S, Janáček J, Cvetko E. Skeletal muscle myosin heavy chain expression and 3D capillary network changes in streptozotocin-induced diabetic female mice. BIOMOLECULES & BIOMEDICINE 2024; 24:582-592. [PMID: 37902457 PMCID: PMC11088899 DOI: 10.17305/bb.2023.9843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 10/31/2023]
Abstract
It is not well-understood how type 1 diabetes (T1DM) affects skeletal muscle histological phenotype, particularly capillarisation. This study aimed to analyze skeletal muscle myosin heavy chain (MyHC) fibre type changes and 3D capillary network characteristics in experimental T1DM mice. Female C57BL/6J-OlaHsd mice were categorized into streptozotocin (STZ)-induced diabetic (n = 12) and age-matched non-diabetic controls (n =12). The muscle fibre phenotype of the soleus, gluteus maximus, and gastrocnemius muscles were characterized based on the expression of MyHC isoforms, while capillaries of the gluteus maximus were assessed with immunofluorescence staining, confocal laser microscopy and 3D image analysis. STZ-induced diabetic mice exhibited elevated glucose levels, reduced body weight, and prolonged thermal latency, verifying the T1DM phenotype. In both T1DM and non-diabetic mice, the gluteus maximus and gastrocnemius muscles predominantly expressed fast-twitch type 2b fibers, with no significant differences noted. However, the soleus muscle in non-diabetic mice had a greater proportion of type 2a fibers and comparable type 1 fiber densities (26.2 ± 14.6% vs 21.9 ± 13.5%) relative to diabetic mice. T1DM mice showed reduced fiber diameters (P = 0.026), and the 3D capillary network analysis indicated a higher capillary length per muscle volume in the gluteus maximus of diabetic mice compared to controls (P < 0.05). Overall, T1DM induced significant changes in the skeletal muscle, including shifts in MyHC fibre types, decreased fibre diameters, and increased relative capillarisation, possibly due to muscle fibre atrophy. Our findings emphasize the superior detail provided by the 3D analytical method for characterizing skeletal muscle capillary architecture, highlighting caution in interpreting 2D data for capillary changes in T1DM.
Collapse
Affiliation(s)
- Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Luka Pušnik
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Žiga Šink
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Simon Horvat
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Jiří Janáček
- Laboratory of Biomathematics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Erika Cvetko
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
8
|
Takahashi N, Ichii O, Hiraishi M, Namba T, Otani Y, Nakamura T, Kon Y. Phenotypes of streptozotocin-induced gestational diabetes mellitus in mice. PLoS One 2024; 19:e0302041. [PMID: 38626157 PMCID: PMC11020761 DOI: 10.1371/journal.pone.0302041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/27/2024] [Indexed: 04/18/2024] Open
Abstract
Gestational diabetes mellitus (GDM) in human patients disrupts glucose metabolism post-pregnancy, affecting fetal development. Although obesity and genetic factors increase GDM risk, a lack of suitable models impedes a comprehensive understanding of its pathology. To address this, we administered streptozotocin (STZ, 75 mg/kg) to C57BL/6N mice for two days before pregnancy, establishing a convenient GDM model. Pregnant mice exposed to STZ (STZ-pregnant) were compared with STZ-injected virgin mice (STZ-virgin), citrate buffer-injected virgin mice (CB-virgin), and pregnant mice injected with citrate buffer (CB-pregnant). STZ-pregnant non-obese mice exhibited elevated blood glucose levels on gestational day 15.5 and impaired glucose tolerance. They also showed fewer normal fetuses compared to CB-pregnant mice. Additionally, STZ-pregnant mice had the highest plasma C-peptide levels, with decreased pancreatic islets or increased alpha cells compared to CB-pregnant mice. Kidneys isolated from STZ-pregnant mice did not display histological alterations or changes in gene expression for the principal glucose transporters (GLUT2 and SGLT2) and renal injury-associated markers. Notably, STZ-pregnant mice displayed decreased gene expression of insulin-receiving molecules (ISNR and IGFR1), indicating heightened insulin resistance. Liver histology in STZ-pregnant mice remained unchanged except for a pregnancy-related increase in lipid droplets within hepatocytes. Furthermore, the duodenum of STZ-pregnant mice exhibited increased gene expression of ligand-degradable IGFR2 and decreased expression of GLUT5 and GLUT12 (fructose and glucose transporters, respectively) compared to STZ-virgin mice. Thus, STZ-pregnant mice displayed GDM-like symptoms, including fetal abnormalities, while organs adapted to impaired glucose metabolism by altering glucose transport and insulin reception without histopathological changes. STZ-pregnant mice offer a novel model for studying mild onset non-obese GDM and species-specific differences in GDM features between humans and animals.
Collapse
Affiliation(s)
- Narumi Takahashi
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Masaya Hiraishi
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Namba
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Otani
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Teppei Nakamura
- Laboratory of Agrobiomedical Science, Faculty of Agriculture, Hokkaido University, Sapporo, Japan
- Laboratory of Laboratory Animal Science and Medicine, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
9
|
Zhao A, Jiang H, Palomares AR, Larsson A, He W, Grünler J, Zheng X, Rodriguez Wallberg KA, Catrina SB, Deng Q. Appropriate glycemic management protects the germline but not the uterine environment in hyperglycemia. EMBO Rep 2024; 25:1752-1772. [PMID: 38491313 PMCID: PMC11014859 DOI: 10.1038/s44319-024-00097-7] [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: 11/21/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 03/18/2024] Open
Abstract
Emerging evidence indicates that parental diseases can impact the health of subsequent generations through epigenetic inheritance. Recently, it was shown that maternal diabetes alters the metaphase II oocyte transcriptome, causing metabolic dysfunction in offspring. However, type 1 diabetes (T1D) mouse models frequently utilized in previous studies may be subject to several confounding factors due to severe hyperglycemia. This limits clinical translatability given improvements in glycemic control for T1D subjects. Here, we optimize a T1D mouse model to investigate the effects of appropriately managed maternal glycemic levels on oocytes and intrauterine development. We show that diabetic mice with appropriate glycemic control exhibit better long-term health, including maintenance of the oocyte transcriptome and chromatin accessibility. We further show that human oocytes undergoing in vitro maturation challenged with mildly increased levels of glucose, reflecting appropriate glycemic management, also retain their transcriptome. However, fetal growth and placental function are affected in mice despite appropriate glycemic control, suggesting the uterine environment rather than the germline as a pathological factor in developmental programming in appropriately managed diabetes.
Collapse
Affiliation(s)
- Allan Zhao
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hong Jiang
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Alice Larsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Wenteng He
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jacob Grünler
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Xiaowei Zheng
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Kenny A Rodriguez Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Division of Gynecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Diabetes, Academic Specialist Centrum, Stockholm, Sweden
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
10
|
Surico PL, Narimatsu A, Forouzanfar K, Singh RB, Shoushtari S, Dana R, Blanco T. Effects of Diabetes Mellitus on Corneal Immune Cell Activation and the Development of Keratopathy. Cells 2024; 13:532. [PMID: 38534376 DOI: 10.3390/cells13060532] [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: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
Diabetes mellitus (DM) is one of the most prevalent diseases globally, and its prevalence is rapidly increasing. Most patients with a long-term history of DM present with some degree of keratopathy (DK). Despite its high incidence, the underlying inflammatory mechanism of DK has not been elucidated yet. For further insights into the underlying immunopathologic processes, we utilized streptozotocin-induced mice to model type 1 DM (T1D) and B6.Cg-Lepob/J mice to model type 2 DM (T2D). We evaluated the animals for the development of clinical manifestations of DK. Four weeks post-induction, the total frequencies of corneal CD45+CD11b+Ly-6G- myeloid cells, with enhanced gene and protein expression levels for the proinflammatory cytokines TNF-α and IL-1β, were higher in both T1D and T2D animals. Additionally, the frequencies of myeloid cells/mm2 in the sub-basal neural plexus (SBNP) were significantly higher in T1D and T2D compared to non-diabetic mice. DK clinical manifestations were observed four weeks post-induction, including significantly lower tear production, corneal sensitivity, and epitheliopathy. Nerve density in the SBNP and intraepithelial terminal endings per 40x field were lower in both models compared to the normal controls. The findings of this study indicate that DM alters the immune quiescent state of the cornea during disease onset, which may be associated with the progressive development of the clinical manifestations of DK.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Akitomo Narimatsu
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Katayoon Forouzanfar
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Sara Shoushtari
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Tomas Blanco
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
11
|
McCurry CM, Sunilkumar S, Subrahmanian SM, Yerlikaya EI, Toro AL, VanCleave AM, Stevens SA, Barber AJ, Sundstrom JM, Dennis MD. NLRP3 Inflammasome Priming in the Retina of Diabetic Mice Requires REDD1-Dependent Activation of GSK3β. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 38546584 PMCID: PMC10981446 DOI: 10.1167/iovs.65.3.34] [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: 09/27/2023] [Accepted: 03/04/2024] [Indexed: 04/01/2024] Open
Abstract
Purpose Inflammasome activation has been implicated in the development of retinal complications caused by diabetes. This study was designed to identify signaling events that promote retinal NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation in response to diabetes. Methods Diabetes was induced in mice by streptozotocin administration. Retinas were examined after 16 weeks of diabetes. Human MIO-M1 Müller cells were exposed to hyperglycemic culture conditions. Genetic and pharmacological interventions were used to interrogate signaling pathways. Visual function was assessed in mice using a virtual optomotor system. Results In the retina of diabetic mice and in Müller cell cultures, NLRP3 and interleukin-1β (IL-1β) were increased in response to hyperglycemic conditions and the stress response protein Regulated in Development and DNA damage 1 (REDD1) was required for the effect. REDD1 deletion prevented caspase-1 activation in Müller cells exposed to hyperglycemic conditions and reduced IL-1β release. REDD1 promoted nuclear factor κB signaling in cells exposed to hyperglycemic conditions, which was necessary for an increase in NLRP3. Expression of a constitutively active GSK3β variant restored NLRP3 expression in REDD1-deficient cells exposed to hyperglycemic conditions. GSK3 activity was necessary for increased NLRP3 expression in the retina of diabetic mice and in cells exposed to hyperglycemic conditions. Müller glia-specific REDD1 deletion prevented increased retinal NLRP3 levels and deficits in contrast sensitivity in diabetic mice. Conclusions The data support a role for REDD1-dependent activation of GSK3β in NLRP3 inflammasome transcriptional priming and in the production of IL-1β by Müller glia in response to diabetes.
Collapse
Affiliation(s)
- Christopher M. McCurry
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Sandeep M. Subrahmanian
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Esma I. Yerlikaya
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Allyson L. Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Ashley M. VanCleave
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Shaunaci A. Stevens
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Alistair J. Barber
- Department of Ophthalmology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Jeffery M. Sundstrom
- Department of Ophthalmology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Michael D. Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
- Department of Ophthalmology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| |
Collapse
|
12
|
Pinho ACO, Santos D, Oliveira PJ, Leal EC, Carvalho E. Real-time OXPHOS capacity analysis in wounded skin from diabetic mice: A pilot study. Eur J Clin Invest 2024; 54:e14128. [PMID: 37975307 DOI: 10.1111/eci.14128] [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: 07/26/2023] [Revised: 10/25/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Diabetes mellitus (DM) impairs wound healing. The aim was to determine whether DM influences mitochondrial respiration in wounded skin (WS) and non-wounded skin (NWS), in a pre-clinical wound healing model of streptozotocin (STZ)-induced diabetes. METHODS Six weeks after diabetes induction, two wounds were created in the back of C57BL/J6 mice. Using high-resolution respirometry (HRR), oxygen flux was measured, in WS and NWS, using two substrate-uncoupler-inhibitor titration protocols, at baseline (day 0), day 3 and 10 post-wounding, in STZ-DM and non-diabetic (NDM) mice. Flux control ratios for the oxidative phosphorylation (OXPHOS) capacity were calculated. RESULTS A significant increase in mitochondrial respiration was observed in STZ-DM skin compared to control skin at baseline. The OXPHOS capacity was decreased in WS under diabetes at day 3 post-wounding (inflammation phase). However, at day 10 post-wounding (remodeling phase), the OXPHOS capacity was higher in WS from STZ-DM compared to NDM mice, and compared to NWS from STZ-DM mice. A significant relative contribution of pyruvate, malate and glutamate (PMG) oxidation to the OXPHOS capacity was observed in WS compared to NWS from STZ-DM mice, at day 10, while the relative contribution of fatty acid oxidation to the OXPHOS capacity was higher in NWS. The OXPHOS capacity is altered in WS from STZ-DM compared to NDM mice across the healing process, and so is the substrate contribution in WS and NWS from STZ-DM mice, at each time point. CONCLUSION HRR may be a sensitive tool to evaluate the underlying mechanisms of tissue repair during wound healing.
Collapse
Grants
- DL57/2016/CP1448/ CT0024 Fundação para a Ciência e a Tecnologia (FCT), I. P, Portugal
- POCI-01-0145-FEDER-007440 Fundação para a Ciência e a Tecnologia (FCT), I. P, Portugal
- SFRH/BD/144199/2019 Fundação para a Ciência e a Tecnologia (FCT), I. P, Portugal
- SFRH/BD/145054/2019 Fundação para a Ciência e a Tecnologia (FCT), I. P, Portugal
- UIDB/04539/2020 Fundação para a Ciência e a Tecnologia (FCT), I. P, Portugal
Collapse
Affiliation(s)
- Aryane Cruz Oliveira Pinho
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Inovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, PhD Programme in Biosciences, University of Coimbra, Coimbra, Portugal
| | - Diana Santos
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Inovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Paulo J Oliveira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Inovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ermelindo Carreira Leal
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Inovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Eugenia Carvalho
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Inovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- APDP-Portuguese Diabetes Association, Lisbon, Portugal
| |
Collapse
|
13
|
Chaudhary S, Pahwa F, Nanda RK. Dysregulated cysteine metabolism leads to worsened liver pathology in diabetes-tuberculosis comorbid condition. J Biol Chem 2024; 300:105634. [PMID: 38199571 PMCID: PMC10850780 DOI: 10.1016/j.jbc.2024.105634] [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: 08/27/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetes mellitus (DM) is a risk factor for developing active tuberculosis (TB) with a 3-fold increase in susceptibility and a 4-fold higher relapse rate. With increasing DM prevalence in TB endemic regions, understanding pathophysiological changes associated with DM-TB comorbidity is imperative. In this study, streptozotocin (STZ)-induced DM C57BL/6 mice were aerosol infected with low dose (100-120 CFU) Mycobacterium tuberculosis H37Rv. At 3 weeks post infection (w.p.i.), multiple tissue mycobacterial load and metabolites were profiled. The liver proteome of DM-TB and controls were analyzed using quantitative proteomics, and multi-omics data were integrated. DM-TB mice showed dysregulated multi-tissue (lungs, liver, brain, kidney and thigh muscle) metabolism. In contrast, the mycobacterial burden in the lung, spleen and liver was similar at 3 w.p.i. in DM-TB and TB groups. Enrichment analysis of deregulated liver metabolites (n = 20; log2DM-TB/TB>±1.0) showed significant perturbation in cysteine-methionine, glycine-serine, BCAA and fatty acid metabolism. 60 out of 1660 identified liver proteins showed deregulation (log2DM-TB/TB>±1.0) and contributed from perturbed cysteine-methionine metabolism corroborating metabolomics data. In addition, amino acid biosynthesis, retinol metabolism and polyol biosynthetic process were also differentially enriched in the livers of DM-TB groups. Global correlation analysis of liver metabolome and proteome data showed a strong association between aspartic acid, pyruvic acid, leucine and isoleucine with CYP450 enzymes involved in retinol metabolism, while iminodiacetic acid, isoleucine and γ-aminobutyric acid (GABA) strong positive correlation involved in cysteine metabolism. Targeting perturbed cysteine metabolism using micro molecules, like DL-Propargylglycine, might help prevent liver damage in DM-TB comorbid conditions.
Collapse
Affiliation(s)
- Shweta Chaudhary
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Falak Pahwa
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ranjan K Nanda
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.
| |
Collapse
|
14
|
Cubillos S, Kazlauskas A. Manifestation of Pathology in Animal Models of Diabetic Retinopathy Is Delayed from the Onset of Diabetes. Int J Mol Sci 2024; 25:1610. [PMID: 38338889 PMCID: PMC10855501 DOI: 10.3390/ijms25031610] [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: 12/18/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Diabetic retinopathy (DR) is the most common complication that develops in patients with diabetes mellitus (DM) and is the leading cause of blindness worldwide. Fortunately, sight-threatening forms of DR develop only after several decades of DM. This well-documented resilience to DR suggests that the retina is capable of protecting itself from DM-related damage and also that accumulation of such damage occurs only after deterioration of this resilience. Despite the enormous translational significance of this phenomenon, very little is known regarding the nature of resilience to DR. Rodent models of DR have been used extensively to study the nature of the DM-induced damage, i.e., cardinal features of DR. Many of these same animal models can be used to investigate resilience because DR is delayed from the onset of DM by several weeks or months. The purpose of this review is to provide a comprehensive overview of the literature describing the use of rodent models of DR in type-1 and type-2 diabetic animals, which most clearly document the delay between the onset of DM and the appearance of DR. These readily available experimental settings can be used to advance our current understanding of resilience to DR and thereby identify biomarkers and targets for novel, prevention-based approaches to manage patients at risk for developing DR.
Collapse
Affiliation(s)
- Samuel Cubillos
- University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA;
| | | |
Collapse
|
15
|
Meda Venkata SP, Li H, Xu L, Wang JM. The Impact of Obesity on Diabetes Onset and Neovascularization in Mouse Models of Metabolic Stress. Int J Mol Sci 2024; 25:1214. [PMID: 38279212 PMCID: PMC10816627 DOI: 10.3390/ijms25021214] [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: 11/14/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Animal models of metabolic disorders are essential to studying pathogenic mechanisms and developing therapies for diabetes, but the induction protocols vary, and sexual dimorphism often exists. In a chronic diabetic model of diet-induced obesity (DIO) and low-dose streptozotocin (STZ)-induced hyperglycemia, blood glucose and lipid profiles were measured. The high-fat (HF) diet damaged insulin sensitivity and increased triglycerides, total cholesterol, LDL-cholesterol, HDL-cholesterol, and liver lipid deposition. STZ increased blood glucose and liver fibrosis with less effects on blood lipids or liver lipid deposition. The combination of DIO and STZ treatments led to significant liver lipid deposition and fibrosis. Female mice showed delayed body weight gain on HF diet and resisted STZ-induced hyperglycemia. However, once they developed DIO, which occurs around 26 weeks of HF diet, the female mice were prone to STZ-induced hyperglycemia. In hindlimb ischemia, male mice in the DIO-STZ group showed significantly worse neovascularization compared with DIO or STZ groups. The DIO-STZ females showed significantly worse recovery than the DIO-STZ males. Our observations suggest that DIO-STZ is a plausible model for studying metabolic and cardiovascular disorders in obesity and diabetes. Moreover, the findings in female animals stress the need to assess sexual dimorphism and investigate the underlying mechanisms that contribute to the worse vasculopathy manifestations in females in metabolic models.
Collapse
Affiliation(s)
- Sai Pranathi Meda Venkata
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
| | - Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
| | - Liping Xu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
- Centers for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Karmanos Cancer Institute, Detroit, MI 48201, USA
| |
Collapse
|
16
|
Weeks KL, Kiriazis H, Wadley GD, Masterman EI, Sergienko NM, Raaijmakers AJA, Trewin AJ, Harmawan CA, Yildiz GS, Liu Y, Drew BG, Gregorevic P, Delbridge LMD, McMullen JR, Bernardo BC. A gene therapy targeting medium-chain acyl-CoA dehydrogenase (MCAD) did not protect against diabetes-induced cardiac pathology. J Mol Med (Berl) 2024; 102:95-111. [PMID: 37987775 DOI: 10.1007/s00109-023-02397-2] [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: 03/20/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Diabetic cardiomyopathy describes heart disease in patients with diabetes who have no other cardiac conditions but have a higher risk of developing heart failure. Specific therapies to treat the diabetic heart are limited. A key mechanism involved in the progression of diabetic cardiomyopathy is dysregulation of cardiac energy metabolism. The aim of this study was to determine if increasing the expression of medium-chain acyl-coenzyme A dehydrogenase (MCAD; encoded by Acadm), a key regulator of fatty acid oxidation, could improve the function of the diabetic heart. Male mice were administered streptozotocin to induce diabetes, which led to diastolic dysfunction 8 weeks post-injection. Mice then received cardiac-selective adeno-associated viral vectors encoding MCAD (rAAV6:MCAD) or control AAV and were followed for 8 weeks. In the non-diabetic heart, rAAV6:MCAD increased MCAD expression (mRNA and protein) and increased Acadl and Acadvl, but an increase in MCAD enzyme activity was not detectable. rAAV6:MCAD delivery in the diabetic heart increased MCAD mRNA expression but did not significantly increase protein, activity, or improve diabetes-induced cardiac pathology or molecular metabolic and lipid markers. The uptake of AAV viral vectors was reduced in the diabetic versus non-diabetic heart, which may have implications for the translation of AAV therapies into the clinic. KEY MESSAGES: The effects of increasing MCAD in the diabetic heart are unknown. Delivery of rAAV6:MCAD increased MCAD mRNA and protein, but not enzyme activity, in the non-diabetic heart. Independent of MCAD enzyme activity, rAAV6:MCAD increased Acadl and Acadvl in the non-diabetic heart. Increasing MCAD cardiac gene expression alone was not sufficient to protect against diabetes-induced cardiac pathology. AAV transduction efficiency was reduced in the diabetic heart, which has clinical implications.
Collapse
Affiliation(s)
- Kate L Weeks
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
| | - Helen Kiriazis
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Emma I Masterman
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Nicola M Sergienko
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Antonia J A Raaijmakers
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Adam J Trewin
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Claudia A Harmawan
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Gunes S Yildiz
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Yingying Liu
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Brian G Drew
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Paul Gregorevic
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
- Centre for Muscle Research, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Julie R McMullen
- Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Bianca C Bernardo
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia.
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia.
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
17
|
Trentin-Sonoda M, Cheff V, Gutsol A, Hébert RL. Sex-dependent effects of Canagliflozin on kidney protection in mice with combined hypertension-type 1 diabetes. PLoS One 2023; 18:e0295284. [PMID: 38055691 DOI: 10.1371/journal.pone.0295284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
Canagliflozin (CANA) is a sodium-glucose cotransporter 2 (SGLT2) inhibitor with blood glucose lowering effects. CANA also promotes kidney protection in patients with cardiovascular diseases and type 2 diabetes (T2D), as well as in normoglycemic patients with hypertension or heart failure. Clinical studies, although conduct in both sexes, do not report sex-dependent differences in T2DM treated with CANA. However, the impact of CANA in type 1 diabetes, as well in sex-dependent outcomes in such cohort needs further understanding. To analyze the effects of CANA in mice with combined hypertension and type 1 diabetes, diabetes was induced by STZ injection (5 days, 50mg/kg/day) in both male and female 8 weeks old genetic hypertensive mice (Lin), whereas the control (Lin) received 0.1M sodium citrate injections. 8 weeks after STZ. Mice were fed either regular or CANA-infused diet for 4 weeks. 8 weeks after STZ, hyperglycemia was present in both male and female mice. CANA reversed BG increase mice fed regular diet. Male LinSTZ mice had elevated water intake, urine output, urinary albumin to creatinine ratio (ACR), kidney lesion score, and creatinine clearance compared to the Lin control group. Kidney injury was improved in male LinSTZ + CANA group in male mice. Water intake and urine output were not statistically significantly different in female LinSTZ compared to female LinSTZ+ CANA. Moreover, CANA did not improve kidney injury in female mice, showing no effect in creatinine clearance, lesion score and fibrosis when compared to LinSTZ fed regular diet. Here we show that Canagliflozin might exert different kidney protection effects in male compared to female mice with hypertension and type 1 diabetes. Sex-dimorphisms were previously found in the pathophysiology of diabetes induced by STZ. Therefore, we highlight the importance of in-depth investigation on sex-dependent effects of CANA, taking in consideration the unique characteristics of disease progression for each sex.
Collapse
Affiliation(s)
- Mayra Trentin-Sonoda
- Kidney Research Centre, Division of Nephrology, Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Véronique Cheff
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alex Gutsol
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Ganesh GV, Ramkumar KM. Pterostilbene accelerates wound healing response in diabetic mice through Nrf2 regulation. Mol Immunol 2023; 164:17-27. [PMID: 37926050 DOI: 10.1016/j.molimm.2023.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023]
Abstract
Pterostilbene (PTS), known for its diverse beneficial effects via Nuclear factor erythroid-2 related factor (Nrf2) activation, holds potential for Diabetic Foot Ulcer (DFU) treatment. However, PTS-mediated Nrf2 regulation in diabetic wounds has yet to be elucidated. We used IC21 macrophage-conditioned media to simulate complex events that can influence the fibroblast phenotype using L929 cells during the wound healing process under a hyperglycemic microenvironment. We found that PTS attenuated fibroblast migration and alpha-smooth muscle actin (α-SMA) levels and hypoxia-inducible factor- 1 alpha (HIF1α). Furthermore, we demonstrated that wounds in diabetic mice characterized by impaired wound closure in a heightened inflammatory milieu, such as the NOD-like receptor P3 (NLRP3) and intercellular adhesion molecule 1 (ICAM1), and deficient Nrf2 response accompanying lowered Akt signaling and heme oxygenase1 (HO1) expression along with the impaired macrophage M2 marker CD206 expression, was rescued by administration of PTS. Such an elicited response was also compared favorably with the standard treatment using Regranex, a commercially available topical formulation for treating DFUs. Our findings suggest that PTS regulates Nrf2 in diabetic wounds, triggering a pro-wound healing response mediated by macrophages. This insight holds the potential for developing targeted therapies to heal chronic wounds, including DFUs.
Collapse
Affiliation(s)
- Goutham V Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
19
|
Zhang S, Wei X, Bowers M, Jessberger S, Golczak M, Semenkovich CF, Rajagopal R. Increasing Energetic Demands on Photoreceptors in Diabetes Corrects Retinal Lipid Dysmetabolism and Reduces Subsequent Microvascular Damage. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2144-2155. [PMID: 37741454 PMCID: PMC10777362 DOI: 10.1016/j.ajpath.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/14/2023] [Accepted: 09/01/2023] [Indexed: 09/25/2023]
Abstract
Mechanisms responsible for the pathogenesis of diabetic retinal disease remain incompletely understood, but they likely involve multiple cellular targets, including photoreceptors. Evidence suggests that dysregulated de novo lipogenesis in photoreceptors is a critical early target of diabetes. Following on this observation, the present study aimed to determine whether two interventions shown to improve diabetic retinopathy in mice-pharmacologic visual cycle inhibition and prolonged dark adaptation-reduce photoreceptor anabolic lipid metabolism. Elevated retinal lipid biosynthetic signaling was observed in two mouse models of diabetes, with both models showing reduced retinal AMP-activated kinase (AMPK) signaling, elevated acetyl CoA carboxylase (ACC) signaling, and increased activity of fatty acid synthase, which promotes lipotoxicity in photoreceptors. Although retinal AMPK-ACC axis signaling was dependent on systemic glucose fluctuations in healthy animals, mice with diabetes lacked such regulation. Visual cycle inhibition and prolonged dark adaptation reversed abnormal retinal AMPK-ACC signaling in mice with diabetes. Although visual cycle inhibition reduced the severity of diabetic retinopathy in control mice, as assessed by retinal capillary atrophy, this intervention was ineffective in fatty acid synthase gain-of-function mice. These results suggest that early diabetic retinopathy is characterized by glucose-driven elevations in retinal lipid biosynthetic activity, and that two interventions known to increase photoreceptor glucose demands alleviate disease by reversing these signals.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri
| | - Xiaochao Wei
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Megan Bowers
- Faculties of Medicine and Science, Laboratory of Neural Plasticity, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Sebastian Jessberger
- Faculties of Medicine and Science, Laboratory of Neural Plasticity, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Clay F Semenkovich
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Rithwick Rajagopal
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
20
|
McCready RG, Gilley KR, Kusumo LE, Hall GM, Vichaya EG. Chronic Stress Exacerbates Hyperglycemia-Induced Affective Symptoms in Male Mice. Neuroimmunomodulation 2023; 30:302-314. [PMID: 37852199 PMCID: PMC10641805 DOI: 10.1159/000534669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023] Open
Abstract
INTRODUCTION Among chronically ill populations, affective disorders remain underdiagnosed and undertreated. A high degree of comorbidity exists between diabetes and affective disorders, particularly depression and anxiety. The mechanisms underlying stress-induced affective dysregulation are likely distinct from those induced by diabetes. A direct comparison between stress- and hyperglycemia-induced affective dysregulation could provide insight into distinct mechanistic targets for depression/anxiety associated with these different conditions. METHODS To this end, the present study used male C57BL/6J mice to compare the independent and combined behavioral and neuroinflammatory effects of two models: (1) unpredictable chronic mild stress and (2) pharmacologically induced hyperglycemia. RESULTS Streptozotocin-induced hyperglycemia was associated with a set of behavioral changes reflective of the neurovegetative symptoms of depression (i.e., reduced open field activity, reduced grooming, increased immobility in the forced swim task, and decreased marble burying), increased hippocampal Bdnf and Tnf expression, and elevations in frontal cortex Il1b expression. Our chronic stress protocol produced alterations in anxiety-like behavior and decreased frontal cortex Il1b expression. DISCUSSION While the combination of chronic stress and hyperglycemia produced limited additive effects, their combination exacerbated total symptom burden. Overall, the data indicate that stress and hyperglycemia induce different symptom profiles via distinct mechanisms.
Collapse
Affiliation(s)
- Riley G McCready
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Kayla R Gilley
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Laura E Kusumo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Grace M Hall
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Elisabeth G Vichaya
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| |
Collapse
|
21
|
Park SS, Mai M, Ploszaj M, Cai H, McGarvey L, Mueller C, Garcia-Arcos I, Geraghty P. Type 1 diabetes contributes to combined pulmonary fibrosis and emphysema in male alpha 1 antitrypsin deficient mice. PLoS One 2023; 18:e0291948. [PMID: 37819895 PMCID: PMC10566687 DOI: 10.1371/journal.pone.0291948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/09/2023] [Indexed: 10/13/2023] Open
Abstract
Type 1 diabetes (T1D) is a metabolic disease characterized by hyperglycemia and can affect multiple organs, leading to life-threatening complications. Increased prevalence of pulmonary disease is observed in T1D patients, and diabetes is a leading cause of comorbidity in several lung pathologies. A deficiency of alpha-1 antitrypsin (AAT) can lead to the development of emphysema. Decreased AAT plasma concentrations and anti-protease activity are documented in T1D patients. The objective of this study was to determine whether T1D exacerbates the progression of lung damage in AAT deficiency. First, pulmonary function testing (PFT) and histopathological changes in the lungs of C57BL/6J streptozotocin (STZ)-induced T1D mice were investigated 3 and 6 months after the onset of hyperglycemia. PFT demonstrated a restrictive pulmonary pattern in the lungs of STZ-injected mice, along with upregulation of mRNA expression of pro-fibrotic markers Acta2, Ccn2, and Fn1. Increased collagen deposition was observed 6 months after the onset of hyperglycemia. To study the effect of T1D on the progression of lung damage in AAT deficiency background, C57BL/6J AAT knockout (KO) mice were used. Control and STZ-challenged AAT KO mice did not show significant changes in lung function 3 months after the onset of hyperglycemia. However, histological examination of the lung demonstrated increased collagen accumulation and alveolar space enlargement in STZ-induced AAT KO mice. AAT pretreatment on TGF-β-stimulated primary lung fibroblasts reduced mRNA expression of pro-fibrotic markers ACTA2, CCN2, and FN1. Induction of T1D in AAT deficiency leads to a combined pulmonary fibrosis and emphysema (CPFE) phenotype in male mice.
Collapse
Affiliation(s)
- Sangmi S. Park
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
| | - Michelle Mai
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
| | - Magdalena Ploszaj
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
| | - Huchong Cai
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
| | - Lucas McGarvey
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
| | - Christian Mueller
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, Worcester, Massachusetts, United States of America
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Itsaso Garcia-Arcos
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
| | - Patrick Geraghty
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, United States of America
| |
Collapse
|
22
|
Elzinga SE, Eid SA, McGregor BA, Jang DG, Hinder LM, Dauch JR, Hayes JM, Zhang H, Guo K, Pennathur S, Kretzler M, Brosius FC, Koubek EJ, Feldman EL, Hur J. Transcriptomic analysis of diabetic kidney disease and neuropathy in mouse models of type 1 and type 2 diabetes. Dis Model Mech 2023; 16:dmm050080. [PMID: 37791586 PMCID: PMC10565109 DOI: 10.1242/dmm.050080] [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: 01/13/2023] [Accepted: 04/26/2023] [Indexed: 10/05/2023] Open
Abstract
Diabetic kidney disease (DKD) and diabetic peripheral neuropathy (DPN) are common complications of type 1 (T1D) and type 2 (T2D) diabetes. However, the mechanisms underlying pathogenesis of these complications are unclear. In this study, we optimized a streptozotocin-induced db/+ murine model of T1D and compared it to our established db/db T2D mouse model of the same C57BLKS/J background. Glomeruli and sciatic nerve transcriptomic data from T1D and T2D mice were analyzed by self-organizing map and differential gene expression analysis. Consistent with prior literature, pathways related to immune function and inflammation were dysregulated in both complications in T1D and T2D mice. Gene-level analysis identified a high degree of concordance in shared differentially expressed genes (DEGs) in both complications and across diabetes type when using mice from the same cohort and genetic background. As we have previously shown a low concordance of shared DEGs in DPN when using mice from different cohorts and genetic backgrounds, this suggests that genetic background may influence diabetic complications. Collectively, these findings support the role of inflammation and indicate that genetic background is important in complications of both T1D and T2D.
Collapse
Affiliation(s)
- Sarah E. Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephanie A. Eid
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brett A. McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Dae-Gyu Jang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lucy M. Hinder
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hongyu Zhang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Frank C. Brosius
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - Emily J. Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| |
Collapse
|
23
|
Yang W, Xia F, Mei F, Shi S, Robichaux WG, Lin W, Zhang W, Liu H, Cheng X. Upregulation of Epac1 Promotes Pericyte Loss by Inducing Mitochondrial Fission, Reactive Oxygen Species Production, and Apoptosis. Invest Ophthalmol Vis Sci 2023; 64:34. [PMID: 37651112 PMCID: PMC10476449 DOI: 10.1167/iovs.64.11.34] [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: 03/30/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Purpose The pathogenic mechanisms behind the development of ischemic retinopathy are complex and poorly understood. This study investigates the involvement of exchange protein directly activated by cAMP (Epac)1 signaling in pericyte injury during ischemic retinopathy, including diabetic retinopathy, a disease that threatens vision. Methods Mouse models of retinal ischemia-reperfusion injury and type 1 diabetes induced by streptozotocin were used to investigate the pathogenesis of these diseases. The roles of Epac1 signaling in the pathogenesis of ischemic retinopathy were determined by an Epac1 knockout mouse model. The cellular and molecular mechanisms of Epac1-mediated pericyte dysfunction in response to high glucose were investigated by specific modulation of Epac1 activity in primary human retinal pericytes using Epac1-specific RNA interference and a pharmacological inhibitor. Results Ischemic injury or diabetes-induced retinal capillary degeneration were associated with an increased expression of Epac1 in the mouse retinal vasculature, including both endothelial cells and pericytes. Genetic deletion of Epac1 protected ischemic injury-induced pericyte loss and capillary degeneration in the mouse retina. Furthermore, high glucose-induced Epac1 expression in retinal pericytes was accompanied by increased Drp1 phosphorylation, mitochondrial fission, reactive oxygen species production, and caspase 3 activation. Inhibition of Epac1 via RNA interference or pharmacological approaches blocked high glucose-mediated mitochondrial dysfunction and caspase 3 activation. Conclusions Our study reveals an important role of Epac1 signaling in mitochondrial dynamics, reactive oxygen species production, and apoptosis in retinal pericytes and identifies Epac1 as a therapeutic target for treating ischemic retinopathy.
Collapse
Affiliation(s)
- Wenli Yang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Fang Mei
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - William G. Robichaux
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Wei Lin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
- Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas, United States
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| |
Collapse
|
24
|
Li Y, Baccouche B, Del-Risco N, Park J, Song A, McAnany JJ, Kazlauskas A. The Slow Progression of Diabetic Retinopathy Is Associated with Transient Protection of Retinal Vessels from Death. Int J Mol Sci 2023; 24:10869. [PMID: 37446043 PMCID: PMC10341443 DOI: 10.3390/ijms241310869] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
The purpose of this study was to investigate the reason that diabetic retinopathy (DR) is delayed from the onset of diabetes (DM) in diabetic mice. To this end, we tested the hypothesis that the deleterious effects of DM are initially tolerated because endogenous antioxidative defense is elevated and thereby confers resistance to oxidative stress-induced death. We found that this was indeed the case in both type 1 DM (T1D) and type 2 DM (T2D) mouse models. The retinal expression of antioxidant defense genes was increased soon after the onset of DM. In addition, ischemia/oxidative stress caused less death in the retinal vasculature of DM versus non-DM mice. Further investigation with T1D mice revealed that protection was transient; it waned as the duration of DM was prolonged. Finally, a loss of protection was associated with the manifestation of both neural and vascular abnormalities that are diagnostic of DR in mice. These observations demonstrate that DM can transiently activate protection from oxidative stress, which is a plausible explanation for the delay in the development of DR from the onset of DM.
Collapse
Affiliation(s)
- Yanliang Li
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.L.); (B.B.); (N.D.-R.); (J.P.); (A.S.); (J.J.M.)
| | - Basma Baccouche
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.L.); (B.B.); (N.D.-R.); (J.P.); (A.S.); (J.J.M.)
| | - Norma Del-Risco
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.L.); (B.B.); (N.D.-R.); (J.P.); (A.S.); (J.J.M.)
| | - Jason Park
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.L.); (B.B.); (N.D.-R.); (J.P.); (A.S.); (J.J.M.)
| | - Amy Song
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.L.); (B.B.); (N.D.-R.); (J.P.); (A.S.); (J.J.M.)
| | - J. Jason McAnany
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.L.); (B.B.); (N.D.-R.); (J.P.); (A.S.); (J.J.M.)
| | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.L.); (B.B.); (N.D.-R.); (J.P.); (A.S.); (J.J.M.)
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
25
|
Sharma A, Choi JSY, Watson AMD, Li L, Sonntag T, Lee MKS, Murphy AJ, De Blasio M, Head GA, Ritchie RH, de Haan JB. Cardiovascular characterisation of a novel mouse model that combines hypertension and diabetes co-morbidities. Sci Rep 2023; 13:8741. [PMID: 37253814 DOI: 10.1038/s41598-023-35680-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/22/2023] [Indexed: 06/01/2023] Open
Abstract
Epidemiologic data suggest that the prevalence of hypertension in patients with diabetes mellitus is ∼1.5-2.0 times greater than in matched non-diabetic patients. This co-existent disease burden exacerbates cardiac and vascular injury, leading to structural and functional changes to the myocardium, impaired cardiac function and heart failure. Oxidative stress and persistent low-grade inflammation underlie both conditions, and are identified as major contributors to pathological cardiac remodelling. There is an urgent need for effective therapies that specifically target oxidative stress and inflammation to protect against cardiac remodelling. Animal models are a valuable tool for testing emerging therapeutics, however, there is a notable lack of appropriate animal models of co-morbid diabetes and hypertension. In this study, we describe a novel preclinical mouse model combining diabetes and hypertension to investigate cardiac and vascular pathology of co-morbid disease. Type 1 diabetes was induced in spontaneously hypertensive, 8-week old, male Schlager (BPH/2) mice via 5 consecutive, daily injections of streptozotocin (55 mg/kg in citrate buffer; i.p.). Non-diabetic mice received citrate buffer only. After 10 weeks of diabetes induction, cardiac function was assessed by echocardiography prior to post-mortem evaluation of cardiomyocyte hypertrophy, interstitial fibrosis and inflammation by histology, RT-PCR and flow cytometry. We focussed on the oxidative and inflammatory stress pathways that contribute to cardiovascular remodelling. In particular, we demonstrate that markers of inflammation (monocyte chemoattractant protein; MCP-1), oxidative stress (urinary 8-isoprostanes) and fibrosis (connective tissue growth factor; CTGF) are significantly increased, whilst diastolic dysfunction, as indicated by prolonged isovolumic relaxation time (IVRT), is elevated in this diabetic and hypertensive mouse model. In summary, this pre-clinical mouse model provides researchers with a tool to test therapeutic strategies unique to co-morbid diabetes and hypertension, thereby facilitating the emergence of novel therapeutics to combat the cardiovascular consequences of these debilitating co-morbidities.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Judy S Y Choi
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Anna M D Watson
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, Australia
| | - Leila Li
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Thomas Sonntag
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Man K S Lee
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Andrew J Murphy
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Miles De Blasio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Geoffrey A Head
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Rebecca H Ritchie
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Judy B de Haan
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Australia
- Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
26
|
Choi S, Hong SP, Bae JH, Suh SH, Bae H, Kang KP, Lee HJ, Koh GY. Hyperactivation of YAP/TAZ Drives Alterations in Mesangial Cells through Stabilization of N-Myc in Diabetic Nephropathy. J Am Soc Nephrol 2023; 34:809-828. [PMID: 36724799 PMCID: PMC10125647 DOI: 10.1681/asn.0000000000000075] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 12/18/2022] [Indexed: 01/26/2023] Open
Abstract
SIGNIFICANCE STATEMENT Mesangial cells (MCs) in the kidney are essential to maintaining glomerular integrity, and their impairment leads to major glomerular diseases including diabetic nephropathy (DN). Although high blood glucose elicits abnormal alterations in MCs, the underlying mechanism is poorly understood. We show that YAP/TAZ are increased in MCs of patients with DN and two animal models of DN. High glucose directly induces activation of YAP/TAZ through the canonical Hippo pathway in cultured MCs. Hyperactivation of YAP/TAZ in mouse MCs recapitulates the hallmarks of DN. Activated YAP/TAZ bind and stabilize N-Myc, one of the Myc family. N-Myc stabilization leads to aberrant enhancement of its transcriptional activity and to MC impairments. Our findings shed light on how high blood glucose in diabetes mellitus leads to DN and support a rationale that lowering blood glucose in diabetes mellitus could delay DN pathogenesis. BACKGROUND Mesangial cells (MCs) in the kidney are central to maintaining glomerular integrity, and their impairment leads to major glomerular diseases, including diabetic nephropathy (DN). Although high blood glucose elicits abnormal alterations in MCs, the underlying molecular mechanism is poorly understood. METHODS Immunolocalization of YAP/TAZ and pathological features of PDGFRβ + MCs were analyzed in the glomeruli of patients with DN, in Zucker diabetic fatty rats, and in Lats1/2i ΔPβ mice. RiboTag bulk-RNA sequencing and transcriptomic analysis of gene expression profiles of the isolated MCs from control and Lats1/2iΔPβ mice were performed. Immunoprecipitation analysis and protein stability of N-Myc were performed by the standard protocols. RESULTS YAP and TAZ, the final effectors of the Hippo pathway, are highly increased in MCs of patients with DN and in Zucker diabetic fatty rats. Moreover, high glucose directly induces activation of YAP/TAZ through the canonical Hippo pathway in cultured MCs. Hyperactivation of YAP/TAZ in mouse model MCs recapitulates the hallmarks of DN, including excessive proliferation of MCs and extracellular matrix deposition, endothelial cell impairment, glomerular sclerosis, albuminuria, and reduced glomerular filtration rate. Mechanistically, activated YAP/TAZ bind and stabilize N-Myc protein, one of the Myc family of oncogenes. N-Myc stabilization leads to aberrant enhancement of its transcriptional activity and eventually to MC impairments and DN pathogenesis. CONCLUSIONS Our findings shed light on how high blood glucose in diabetes mellitus leads to DN and support a rationale that lowering blood glucose in diabetes mellitus could delay DN pathogenesis.
Collapse
Affiliation(s)
- Seunghyeok Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Seon Pyo Hong
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jung Hyun Bae
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Sang Heon Suh
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hosung Bae
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Research Institute of Clinical Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Hyuek Jong Lee
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| |
Collapse
|
27
|
Mariadoss AVA, Park S, Saravanakumar K, Sathiyaseelan A, Wang MH. Phytochemical profiling, in vitro antioxidants, and antidiabetic efficacy of ethyl acetate fraction of Lespedeza cuneata on streptozotocin-induced diabetic rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:60976-60993. [PMID: 37042914 DOI: 10.1007/s11356-023-26412-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/08/2023] [Indexed: 05/10/2023]
Abstract
In the recent past, phytomolecules are exponentially applied in discovering the antidiabetic drug due to less adverse effects. This work screened the active solvent fraction of Lespedeza cuneata based on the phytochemical, enzyme inhibition, and antioxidant properties. The antioxidant efficacy of the different fractions of the L. cuneata was assessed by 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), ferric reducing power, hydrogen peroxide, and hydroxyl radical scavenging assays. The digestive enzyme (α-amylase and α-glucosidase) inhibitory activity was also evaluated. The phytochemical composition of ethyl acetate fraction of L. cuneata (Lc-EAF) was studied by UHPLC-QTOF-MS/MS. The effect of Lc-EAF treatments on glucose uptake was studied in insulin resistance HepG2 cells (IR-HepG2). Further, the antidiabetic effect of Lc-EAF in streptozotocin (STZ)-induced diabetic mice were demonstrated. Ethyl acetate, hexane, and methanol fractions of the L. cuneata showed notable antioxidant, α-amylase, and α-glucosidase inhibitory properties. Among the fractions, Lc-EAF was found to be the most potent. The Lc-EAF exhibited an IC50 of 205.32 ± 23.47 µg/mL and 105.32 ± 13.93 µg/mL for α-amylase and α-glucosidase inhibition, respectively. In addition, 75 µg/mL of Lc-EAF exposure enhanced glucose uptake (68.23%) in IR-HepG2 cells. In vivo study indicated that treatment of Lc-EAF (100 mg/kg b.wt) maintained the blood glucose level through reduced insulin level while improving the lipid profile, hepatic, and renal markers. These findings suggest that Lc-EAF could be considered a prominent source for antidiabetic, anti-hyperlipidemic, and anti-ROS potentials.
Collapse
Affiliation(s)
- Arokia Vijaya Anand Mariadoss
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 200-701, Republic of Korea
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, 18450, Republic of Korea
| | - SeonJu Park
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon, 24341, Republic of Korea
| | - Kandasamy Saravanakumar
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 200-701, Republic of Korea
| | - Anbazhagan Sathiyaseelan
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 200-701, Republic of Korea
| | - Myeong-Hyeon Wang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 200-701, Republic of Korea.
| |
Collapse
|
28
|
Dodson TA, Nieuwoudt S, Morse CN, Pierre V, Liu C, Senyo SE, Prestwich EG. Ribonucleosides from tRNA in hyperglycemic mammalian cells and diabetic murine cardiac models. Life Sci 2023; 318:121462. [PMID: 36736767 PMCID: PMC9992345 DOI: 10.1016/j.lfs.2023.121462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
AIMS Cardiomyopathy is a diabetic comorbidity with few molecular targets. To address this, we evaluated transfer RNA (tRNA) modifications in the diabetic heart because tRNA modifications have been implicated in diabetic etiologies. MAIN METHODS tRNA was isolated from aorta, apex, and atrial tissue of healthy and diabetic murine hearts and related hyperglycemic cell models. tRNA modifications and canonical ribonucleosides were quantified by liquid-chromatography tandem mass spectrometry (LC-MS/MS) using stable isotope dilution. Correlations between ribonucleosides and diabetic comorbidity pathology were assessed using statistical analyses. KEY FINDINGS Total tRNA ribonucleoside levels were analyzed from cell types and healthy and diabetic murine heart tissue. Each heart structure had characteristic ribonucleoside profiles and quantities. Several ribonucleosides were observed as significantly different in hyperglycemic cells and diabetic tissues. In hyperglycemic models, ribonucleosides N4-acetylcytidine (ac4C), 5-methoxycarbonylmethyl-2-thiouridine (mcm5s2U), 5-methylcytidine (m5C), and N1-methylguanosine (m1G) were anomalous. Specific tRNA modifications known to be on murine tRNAIni(CAU) were higher in diabetic heart tissue which suggests that tRNA modifications could be regulating translation in diabetes. SIGNIFICANCE We identified tRNA ribonucleosides and tRNA species associated with hyperglycemia and diabetic etiology.
Collapse
Affiliation(s)
- Taylor A Dodson
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Stephan Nieuwoudt
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Chase N Morse
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Valinteshley Pierre
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Chao Liu
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Samuel E Senyo
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Erin G Prestwich
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States.
| |
Collapse
|
29
|
Saadane A, Veenstra AA, Minns MS, Tang J, Du Y, Abubakr Elghazali F, Lessieur EM, Pearlman E, Kern TS. CCR2-positive monocytes contribute to the pathogenesis of early diabetic retinopathy in mice. Diabetologia 2023; 66:590-602. [PMID: 36698021 PMCID: PMC9892100 DOI: 10.1007/s00125-022-05860-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 09/13/2022] [Indexed: 01/27/2023]
Abstract
AIMS/HYPOTHESIS Accumulating evidence suggests that leucocytes play a critical role in diabetes-induced vascular lesions and other abnormalities that characterise the early stages of diabetic retinopathy. However, the role of monocytes has yet to be fully investigated; therefore, we used Ccr2-/- mice to study the role of CCR2+ inflammatory monocytes in the pathogenesis of diabetes-induced degeneration of retinal capillaries. METHODS Experimental diabetes was induced in wild-type and Ccr2-/- mice using streptozotocin. After 2 months, superoxide levels, expression of inflammatory genes, leucostasis, leucocyte- and monocyte-mediated cytotoxicity against retinal endothelial cell death, retinal thickness and visual function were evaluated. Retinal capillary degeneration was determined after 8 months of diabetes. Flow cytometry of peripheral blood for differential expression of CCR2 in monocytes was assessed. RESULTS In nondiabetic mice, CCR2 was highly expressed on monocytes, and Ccr2-/- mice lack CCR2+ monocytes in the peripheral blood. Diabetes-induced retinal superoxide, expression of proinflammatory genes Inos and Icam1, leucostasis and leucocyte-mediated cytotoxicity against retinal endothelial cells were inhibited in diabetic Ccr2-deficient mice and in chimeric mice lacking Ccr2 only from myeloid cells. In order to focus on monocytes, these cells were immuno-isolated after 2 months of diabetes, and they significantly increased monocyte-mediated endothelial cell cytotoxicity ex vivo. Monocytes from Ccr2-deficient mice caused significantly less endothelial cell death. The diabetes-induced retinal capillary degeneration was inhibited in Ccr2-/- mice and in chimeric mice lacking Ccr2 only from myeloid cells. CONCLUSIONS/INTERPRETATION CCR2+ inflammatory monocytes contribute to the pathogenesis of early lesions of diabetic retinopathy.
Collapse
Affiliation(s)
- Aicha Saadane
- Department of Ophthalmology, University of California-Irvine, Irvine, CA, USA.
| | | | - Martin S Minns
- Institute for Immunology, University of California-Irvine, Irvine, CA, USA
| | - Jie Tang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Yunpeng Du
- Department of Ophthalmology, University of California-Irvine, Irvine, CA, USA
| | | | - Emma M Lessieur
- Department of Ophthalmology, University of California-Irvine, Irvine, CA, USA
| | - Eric Pearlman
- Institute for Immunology, University of California-Irvine, Irvine, CA, USA
| | - Timothy S Kern
- Department of Ophthalmology, University of California-Irvine, Irvine, CA, USA
- Veterans Administration Medical Center Research Service, Long Beach, CA, USA
| |
Collapse
|
30
|
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
|
31
|
Parlar Köprülü RE. How to create an experimental diabetes mellitus model? PHARMACIA 2023. [DOI: 10.3897/pharmacia.70.e96028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Diabetes is a metabolic disorder characterized by chronic hyperglycemia. Early treatment is very important in terms of preventing diabetes-related late complications with high treatment costs and increasing the patient’s quality of life. In addition to investigating the pathophysiology of the disease studied, animal experiments pave the way for new approaches in treatments. Although there are many methods that can be used when creating a diabetes model, induction of diabetes with alloxan and streptozotocin are the most preferred ones. The aim of this article is to review the available information on diabetes-related methods, common problems and solutions, with known mechanisms of action, dose and time-determined methods.
Collapse
|
32
|
High fat diet is protective against kidney injury in hypertensive-diabetic mice, but leads to liver injury. PLoS One 2023; 18:e0281123. [PMID: 36730247 PMCID: PMC9894391 DOI: 10.1371/journal.pone.0281123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/13/2023] [Indexed: 02/03/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health burden with increases risk of end-stage renal function if left untreated. CKD induced in the context of metabolic syndrome (MS) increases risks of hypertension, hyperglycemia, excess body fat and dyslipidemia. To test if combining a high-fat diet (HFD) regimen onto the hypertensive/ diabetic phenotype would mimic features of MS induced-CKD in mice, hyperglycemia was induced in genetically hypertensive mice (Lin), followed by HFD regimen. For that, 8-week-old male were subjected to streptozotocin (STZ) intraperitoneal (i.p.) injections (50 mg/kg, 5 days consecutive). LinSTZ were fed a 60% kCal HFD for 8 weeks. Lin mice treated with STZ developed polydipsia, became hypertensive and hyperglycemic. HFD induced weight gain, protected against glomerular hypertrophy, scarring, and albuminuria at endpoint compared to regular diet fed LinSTZ. On the other hand, HFD induced steatosis, liver fibrosis, inflammation, and increase in AST/ALT ratio, characteristics of non-alcoholic liver disease. Taken together, our results show that LinSTZ mice fed a HFD did not lead to a more robust model of MS-induced CKD, protected against kidney injury, but inducing liver damage. More studies are necessary to understand the kidney protective mechanisms of HFD when superimposed with hypertension and type 1 diabetes.
Collapse
|
33
|
Yun J, Park S, Park HY, Lee KA. Efficacy of Polydeoxyribonucleotide in Promoting the Healing of Diabetic Wounds in a Murine Model of Streptozotocin-Induced Diabetes: A Pilot Experiment. Int J Mol Sci 2023; 24:ijms24031932. [PMID: 36768255 PMCID: PMC9916466 DOI: 10.3390/ijms24031932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
We assessed the efficacy of polydeoxyribonucleotide (PDRN) in accelerating the healing of diabetic wounds in a murine model of streptozotocin (STZ)-induced diabetes. After the creation of diabetic wounds, the mice of the PDRN SC, PDRN IP and PBS groups received a subcutaneous, an intra-peritoneal injection of PDRN and a subcutaneous injection of PBS, respectively. After euthanasia, time-dependent changes in the wound diameter and histologic scores were measured and vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1) and collagen types I and III were assessed for their expression levels. The PDRN SC and the PDRN IP groups showed a significantly smaller diameter of diabetic wounds, significantly higher histologic scores, a significantly greater expression of VEGF, a significantly lower expression of TGF-β1 and a significantly greater expression of collagen types I and III as compared with the PBS group (p < 0.05 or 0.0001). In conclusion, PDRN might be effective in promoting the healing of diabetic wounds in a murine model of STZ-induced diabetes.
Collapse
Affiliation(s)
- Jiyoung Yun
- Department of Plastic and Reconstructive Surgery, Inje University Busan Paik Hospital, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - SaeGwang Park
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
- Innovative Therapeutic Research Institute, Inje University Busan Paik Hospital, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Ha Young Park
- Department of Pathology, Inje University Busan Paik Hospital, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Kyung Ah Lee
- Department of Plastic and Reconstructive Surgery, Inje University Haeundae Paik Hospital, College of Medicine, Inje University, Busan 48108, Republic of Korea
- Correspondence:
| |
Collapse
|
34
|
Ghasemi A, Jeddi S. Streptozotocin as a tool for induction of rat models of diabetes: a practical guide. EXCLI JOURNAL 2023; 22:274-294. [PMID: 36998708 PMCID: PMC10043433 DOI: 10.17179/excli2022-5720] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/09/2023] [Indexed: 04/01/2023]
Abstract
Streptozotocin (STZ) is the most used diabetogenic chemical for creating rat models of type 1 and type 2 diabetes. Despite ~60 years of using STZ in animal diabetes research, some prevailing views about STZ preparation and use are not supported by evidence. Here, we provide practical guides for using STZ to induce diabetes in rats. Susceptibility to the diabetogenic effect of STZ is inversely related to age, and males are more susceptible to STZ than females. Wistar and Sprague-Dawley rats, the most commonly-used rat strains, are sensitive to STZ, but some strains (e.g., Wistar-Kyoto rats) are less sensitive. STZ is mostly injected intravenously or intraperitoneally, but its intravenous injection produces more stable hyperglycemia. Despite the prevailing view, no fasting is necessary before STZ injection, and injection of its anomer-equilibrated solutions (i.e., more than 2 hours of dissolving) is recommended. Mortality following the injection of diabetogenic doses of STZ is due to severe hypoglycemia (during the first 24 h) or severe hyperglycemia (24 h after the injection and onwards). Some measures to prevent hypoglycemia-related mortality in rats include providing access to food soon after the injection, administration of glucose/sucrose solutions during the first 24-48 h after the injection, administration of STZ to fed animals, and using anomer-equilibrated solutions of STZ. Hyperglycemia-related mortality following injection of high doses of STZ can be overcome with insulin administration. In conclusion, STZ is a valuable chemical for inducing diabetes in rats, but some practical guides should be considered to perform well-conducted and ethical studies.
Collapse
Affiliation(s)
- Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *To whom correspondence should be addressed: Sajad Jeddi, Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Arabi Street, Daneshjoo Blvd, Velenjak, P.O. Box: 19395-4763, Tehran, Iran, E-mail:
| |
Collapse
|
35
|
Rai V, Agrawal DK. Male or female sex: considerations and translational aspects in diabetic foot ulcer research using rodent models. Mol Cell Biochem 2022. [PMID: 36574098 DOI: 10.1007/s11010-022-04642-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
36
|
Damrath JG, Metzger CE, Allen MR, Wallace JM. A novel murine model of combined insulin-dependent diabetes and chronic kidney disease has greater skeletal detriments than either disease individually. Bone 2022; 165:116559. [PMID: 36116758 PMCID: PMC9798592 DOI: 10.1016/j.bone.2022.116559] [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: 06/24/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 12/31/2022]
Abstract
Diabetes and chronic kidney disease (CKD) consistently rank among the top ten conditions in prevalence and mortality in the United States. Insulin-dependent diabetes (IDD) and CKD each increase the risk of skeletal fractures and fracture-related mortality. However, it remains unknown whether these conditions have interactive end-organ effects on the skeleton. We hypothesized that combining IDD and CKD in mice would cause structural and mechanical bone alterations that are more deleterious compared to the single disease states. Female C57BL6/J mice were divided into four groups: 1) N = 12 Control (CTRL), 2) N = 10 Streptozotocin-induced IDD (STZ), 3) N = 10 Adenine diet-induced CKD (AD), and 4) N = 9 Combination (STZ+AD). STZ administration resulted in significantly higher blood glucose, HbA1c (p < 0.0001), and glucose intolerance (p < 0.0001). AD resulted in higher blood urea nitrogen (p = 0.0002) while AD, but not STZ+AD mice, had high serum parathyroid hormone (p < 0.0001) and phosphorus (p = 0.0005). STZ lowered bone turnover (p = 0.001). Trabecular bone volume was lowered by STZ (p < 0.0001) and increased by AD (p = 0.003). Tissue mineral density was lowered by STZ (p < 0.0001) and AD (p = 0.02) in trabecular bone but only lowered by STZ in cortical bone (p = 0.002). Cortical porosity of the proximal tibia was increased by AD, moment of inertia was lower in both disease groups, and most cortical properties were lower in all groups vs CTRL. Ultimate force, stiffness, toughness, and total displacement/strain were lowered by STZ and AD. Fracture toughness was lower by AD (p = 0.003). Importantly, Cohen's D indicated that STZ+AD most strongly lowered bone turnover and mechanical properties. Taken together, structural and material-level bone properties are altered by STZ and AD while their combination resulted in greater detriments, indicating that improving bone health in the combined disease state may require novel interventions.
Collapse
Affiliation(s)
- John G Damrath
- Purdue University, Weldon School of Biomedical Engineering, West Lafayette, IN, United States
| | - Corinne E Metzger
- Indiana University School of Medicine, Department of Anatomy and Cell Biology, Indianapolis, IN, United States
| | - Matthew R Allen
- Indiana University School of Medicine, Department of Anatomy and Cell Biology, Indianapolis, IN, United States
| | - Joseph M Wallace
- Indiana University-Purdue University at Indianapolis, Department of Biomedical Engineering, Indianapolis, IN, United States.
| |
Collapse
|
37
|
Hughes FM, Odom MR, Cervantes A, Purves J. Inflammation triggered by the NLRP3 inflammasome is a critical driver of diabetic bladder dysfunction. Front Physiol 2022; 13:920487. [PMID: 36505062 PMCID: PMC9733912 DOI: 10.3389/fphys.2022.920487] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
Diabetes is a rapidly expanding epidemic projected to affect as many as 1 in 3 Americans by 2050. This disease is characterized by devastating complications brought about high glucose and metabolic derangement. The most common of these complications is diabetic bladder dysfunction (DBD) and estimates suggest that 50-80% of patients experience this disorder. Unfortunately, the Epidemiology of Diabetes Interventions and Complications Study suggests that strict glucose control does not decrease ones risk for incontinence, although it does decrease the risk of other complications such as retinopathy, nephropathy and neuropathy. Thus, there is a significant unmet need to better understand DBD in order to develop targeted therapies to alleviate patient suffering. Recently, the research community has come to understand that diabetes produces a systemic state of low-level inflammation known as meta-inflammation and attention has focused on a role for the sterile inflammation-inducing structure known as the NLRP3 inflammasome. In this review, we will examine the evidence that NLRP3 plays a central role in inducing DBD and driving its progression towards an underactive phenotype.
Collapse
Affiliation(s)
- Francis M. Hughes
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | | | | | | |
Collapse
|
38
|
Wang W, Hapach LA, Griggs L, Smart K, Wu Y, Taufalele PV, Rowe MM, Young KM, Bates ME, Johnson AC, Ferrell NJ, Pozzi A, Reinhart-King CA. Diabetic hyperglycemia promotes primary tumor progression through glycation-induced tumor extracellular matrix stiffening. SCIENCE ADVANCES 2022; 8:eabo1673. [PMID: 36399580 PMCID: PMC9674287 DOI: 10.1126/sciadv.abo1673] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 09/29/2022] [Indexed: 05/31/2023]
Abstract
Diabetes mellitus is a complex metabolic disorder that is associated with an increased risk of breast cancer. Despite this correlation, the interplay between tumor progression and diabetes, particularly with regard to stiffening of the extracellular matrix, is still mechanistically unclear. Here, we established a murine model where hyperglycemia was induced before breast tumor development. Using the murine model, in vitro systems, and patient samples, we show that hyperglycemia increases tumor growth, extracellular matrix stiffness, glycation, and epithelial-mesenchymal transition of tumor cells. Upon inhibition of glycation or mechanotransduction in diabetic mice, these same metrics are reduced to levels comparable with nondiabetic tumors. Together, our study describes a novel biomechanical mechanism by which diabetic hyperglycemia promotes breast tumor progression via glycating the extracellular matrix. In addition, our work provides evidence that glycation inhibition is a potential adjuvant therapy for diabetic cancer patients due to the key role of matrix stiffening in both diseases.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lauren A. Hapach
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Lauren Griggs
- College of Engineering, Pennsylvania State University, State College, PA 16802, USA
| | - Kyra Smart
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Yusheng Wu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Paul V. Taufalele
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Matthew M. Rowe
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine M. Young
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Madison E. Bates
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Andrew C. Johnson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Nicholas J. Ferrell
- Department of Internal Medicine, Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Veterans Affairs Hospitals, Nashville, TN 37684, USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
39
|
O’Neill CC, Locke EJ, Sipf DA, Thompson JH, Drebushenko EK, Berger NS, Segich BS, Kolwicz SC. The Effects of Exercise Training on Glucose Homeostasis and Muscle Metabolism in Type 1 Diabetic Female Mice. Metabolites 2022; 12:metabo12100948. [PMID: 36295850 PMCID: PMC9608674 DOI: 10.3390/metabo12100948] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 10/01/2022] [Indexed: 01/13/2023] Open
Abstract
Although exercise training is an important recommendation for the management of type 1 diabetes (T1D), most of the available research studies predominantly focus on male subjects. Given the importance of sex as a biological variable, additional studies are required to improve the knowledge gap regarding sex differences in T1D research. Therefore, the purpose of this study was to examine the role of exercise training in mediating changes in glucose homeostasis and skeletal muscle metabolism in T1D female mice. Female mice were injected with streptozotocin (STZ) to induce T1D. Two weeks after STZ injection, control (CON) and STZ mice were exercise trained on a treadmill for 4 weeks. Aerobic exercise training failed to improve glucose tolerance, prevent the decrease in body weight and adipose tissue mass, or attenuate muscle atrophy in T1D female mice. However, insulin sensitivity was improved in T1D female mice after exercise training. Aerobic exercise training maintained skeletal muscle triglyceride content but did not prevent depletion of skeletal muscle or liver glycogen in T1D mice. Gene expression analysis suggested that T1D resulted in decreased glucose transport, decreased ketone body oxidation, and increased fatty acid metabolism in the skeletal muscle, which was not altered by exercise training. These data demonstrate that 4 weeks of aerobic exercise training of a moderate intensity is insufficient to counteract the negative effects of T1D in female mice, but does lead to an improvement in insulin sensitivity.
Collapse
|
40
|
Lessieur EM, Liu H, Saadane A, Du Y, Kiser J, Kern TS. ICAM-1 on the luminal surface of endothelial cells is induced to a greater extent in mouse retina than in other tissues in diabetes. Diabetologia 2022; 65:1734-1744. [PMID: 35852587 PMCID: PMC9481679 DOI: 10.1007/s00125-022-05719-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/16/2022] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Induction of intercellular adhesion molecule-1 (ICAM-1) has been implicated in the development of macrovascular and microvascular diseases such as diabetic retinopathy. Lesions of diabetic retinopathy are unique to the retina but the reason for this is unclear, as all tissues are exposed to the same hyperglycaemic insult. We tested whether diabetes induces ICAM-1 on the luminal surface of endothelial cells to a greater extent in the retina than in other tissues and the role of vision itself in that induction. METHODS Experimental diabetes was induced in C57Bl/6J, P23H opsin mutant and Gnat1-/- × Gnat2-/- double knockout mice using streptozotocin. The relative abundance of ICAM-1 on the luminal surface of endothelial cells in retina and other tissues was determined by conjugating anti-ICAM-1 antibodies to fluorescent microspheres (2 μm), injecting them intravenously and allowing them to circulate for 30 min. After transcardial perfusion, quantification of microspheres adherent to the endothelium in tissues throughout the body was carried out by fluorescent microscopy or flow cytometry. Mice injected with lipopolysaccharide (LPS) were used as positive controls. The difference in leucostasis between retinal and non-retinal vasculature was evaluated. RESULTS Diabetes significantly increased ICAM-1-mediated adherence of microspheres to retinal microvessels by almost threefold, independent of sex. In contrast, diabetes had a much smaller effect on endothelial ICAM-1 in other tissues, and more tissues showed a significant induction of endothelial ICAM-1 with LPS than with diabetes. The diabetes-induced increase in endothelial ICAM-1 in retinal vasculature was inhibited by blocking phototransduction in photoreceptor cells. Diabetes significantly increased leucostasis in the retina by threefold compared with a non-ocular tissue (cremaster). CONCLUSIONS/INTERPRETATION The diabetes-induced upregulation of ICAM-1 on the luminal surface of the vascular endothelium varies considerably among tissues and is highest in the retina. Induction of ICAM-1 on retinal vascular endothelial cells in diabetes is influenced by vision-related processes in photoreceptor cells. The unique presence of photoreceptors in the retina might contribute to the greater susceptibility of this tissue to vascular disease in diabetes.
Collapse
Affiliation(s)
- Emma M Lessieur
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA.
| | - Haitao Liu
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA
- Glia Research Laboratory, Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aicha Saadane
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
| | - Yunpeng Du
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
| | - Jianying Kiser
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
| | - Timothy S Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
- Veterans Administration Medical Center Research Service, Long Beach, CA, USA
| |
Collapse
|
41
|
Chen F, Ge L, Jiang X, Lai Y, Huang P, Hua J, Lin Y, Lin Y, Jiang X. Construction of the experimental rat model of gestational diabetes. PLoS One 2022; 17:e0273703. [PMID: 36107823 PMCID: PMC9477341 DOI: 10.1371/journal.pone.0273703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 08/14/2022] [Indexed: 11/27/2022] Open
Abstract
Objective Numerous methods for modeling gestational diabetes mellitus (GDM) in rats exist. However, their repeatability and stability are unclear. This study aimed to compare the effects of high-fat and high-sugar (HFHS) diet, HFHS diet combined with streptozotocin (STZ) administration, and HFHS diet combined with movement restriction (MR) modeling methods on rat models to confirm the best method for constructing a rat model of GDM. Method Forty female Sprague-Dawley rats were randomly divided into four groups (n = 10): the normal control (NC), HFHS, HFHS+STZ, and HFHS+MR groups. The rats in the NC group were fed with a standard diet, and those in the remaining groups were fed with a HFHS diet. The rats in the HFHS+STZ group received 25 mg/kg STZ on their first day of pregnancy, and those in the HFHS+MR group were subjected to MR during pregnancy. Bodyweight, food intake, water intake, fasting blood glucose (FBG), fasting insulin (FINS), homeostasis model assessment of insulin resistance (HOMA-IR), homeostasis model assessment of insulin sensitivity (HOMA-IS), homeostasis model assessment of β-cell function, pancreatic and placental morphology, and the expression levels of glucose transporter 1 (GLUT1) and glucose transporter 3 (GLUT3) in placentas were then quantified. Moreover, iTRAQ was used to identify placental proteomics. Results During pregnancy, the rats in the HFHS+STZ group showed FBG levels that were kept stable in a state of moderate hyperglycemia; the typical GDM symptoms of polydipsia, polyphagia, polyuria, and increased body weight; and the modeling rate of 87.5%. On the first and 19th days of pregnancy, the rats in the HFHS group showed higher FBG than that of the NC group, increasing body weight and food intake and the modeling rate of 50%. On the 19th day of pregnancy, the FBG of the rats in the HFHS+MR group was higher than that of the rats in the NC group, and the modeling rate of 42.9%. Comparison with the NC group revealed that the three modeling groups exhibited increased FINS and HOMA-IR, decreased HOMA-IS, and different degrees of pathological changes in pancreases and placentas. Among the groups, the HFHS+STZ group displayed the greatest changes with significant reductions in the numbers of pancreatic and placental cells and appeared cavitation. The expression levels of GLUT1 and GLUT3 in the placentas of the HFHS+STZ and HFHS+MR groups were higher than those in the placentas of the NC and HFHS groups. The above results indicated that the rats in the HFHS+STZ group showed the best performance in terms of modeling indicators. After the changes in placental proteomics in the HFHS+STZ group were compared with those in the NC group, we found that in the HFHS+STZ group, five proteins were up-regulated and 18 were down-regulated; these proteins were enriched in estrogen signaling pathways. Conclusion HFHS combined with the intraperitoneal injection of 25 mg/kg STZ was the best modeling method for the nonspontaneous model of experimentally induced GDM, and its modeling rate was high. The pathological characteristics of the constructed GDM rat model were similar to those of human patients with GDM. Moreover, the model was stable and reliable. The modeling method can provide a basis for constructing a GDM rat model for subsequent research on the prevention and treatment of GDM.
Collapse
Affiliation(s)
- Fan Chen
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Li Ge
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- * E-mail:
| | - Xinyong Jiang
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuting Lai
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Pingping Huang
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jinghe Hua
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuzheng Lin
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yan Lin
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiumin Jiang
- Fujian Maternity and Child Health Hospital Affiliated to Fujian Medical University, Fuzhou, China
| |
Collapse
|
42
|
Abstract
Diabetes is a chronic metabolic disease affecting an increasing number of people. Although diabetes has negative health outcomes for diagnosed individuals, a population at particular risk are pregnant women, as diabetes impacts not only a pregnant woman's health but that of her child. In this review, we cover the current knowledge and unanswered questions on diabetes affecting an expectant mother, focusing on maternal and fetal outcomes.
Collapse
Affiliation(s)
- Cecilia González Corona
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA
| | - Ronald J. Parchem
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA,Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
43
|
Antioxidant and anti-apoptotic effects of tocotrienol-rich fraction against streptozotocin-induced diabetic retinopathy in rats. Biomed Pharmacother 2022; 153:113533. [DOI: 10.1016/j.biopha.2022.113533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
|
44
|
Heather LC, Hafstad AD, Halade GV, Harmancey R, Mellor KM, Mishra PK, Mulvihill EE, Nabben M, Nakamura M, Rider OJ, Ruiz M, Wende AR, Ussher JR. Guidelines on Models of Diabetic Heart Disease. Am J Physiol Heart Circ Physiol 2022; 323:H176-H200. [PMID: 35657616 PMCID: PMC9273269 DOI: 10.1152/ajpheart.00058.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Diabetes is a major risk factor for cardiovascular diseases, including diabetic cardiomyopathy, atherosclerosis, myocardial infarction, and heart failure. As cardiovascular disease represents the number one cause of death in people with diabetes, there has been a major emphasis on understanding the mechanisms by which diabetes promotes cardiovascular disease, and how antidiabetic therapies impact diabetic heart disease. With a wide array of models to study diabetes (both type 1 and type 2), the field has made major progress in answering these questions. However, each model has its own inherent limitations. Therefore, the purpose of this guidelines document is to provide the field with information on which aspects of cardiovascular disease in the human diabetic population are most accurately reproduced by the available models. This review aims to emphasize the advantages and disadvantages of each model, and to highlight the practical challenges and technical considerations involved. We will review the preclinical animal models of diabetes (based on their method of induction), appraise models of diabetes-related atherosclerosis and heart failure, and discuss in vitro models of diabetic heart disease. These guidelines will allow researchers to select the appropriate model of diabetic heart disease, depending on the specific research question being addressed.
Collapse
Affiliation(s)
- Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anne D Hafstad
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ganesh V Halade
- Department of Medicine, The University of Alabama at Birmingham, Tampa, Florida, United States
| | - Romain Harmancey
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Erin E Mulvihill
- University of Ottawa Heart Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Miranda Nabben
- Departments of Genetics and Cell Biology, and Clinical Genetics, Maastricht University Medical Center, CARIM School of Cardiovascular Diseases, Maastricht, the Netherlands
| | - Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Oliver J Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matthieu Ruiz
- Montreal Heart Institute, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
45
|
Miller WP, Toro AL, Sunilkumar S, Stevens SA, VanCleave AM, Williamson DL, Barber AJ, Dennis MD. Müller Glial Expression of REDD1 Is Required for Retinal Neurodegeneration and Visual Dysfunction in Diabetic Mice. Diabetes 2022; 71:1051-1062. [PMID: 35167652 PMCID: PMC9074768 DOI: 10.2337/db21-0853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022]
Abstract
Clinical studies support a role for the protein regulated in development and DNA damage response 1 (REDD1) in ischemic retinal complications. To better understand how REDD1 contributes to retinal pathology, we examined human single-cell sequencing data sets and found specificity of REDD1 expression that was consistent with markers of retinal Müller glia. Thus, we investigated the hypothesis that REDD1 expression specifically in Müller glia contributes to diabetes-induced retinal pathology. The retina of Müller glia-specific REDD1 knockout (REDD1-mgKO) mice exhibited dramatic attenuation of REDD1 transcript and protein expression. In the retina of streptozotocin-induced diabetic control mice, REDD1 protein expression was enhanced coincident with an increase in oxidative stress. In the retina of diabetic REDD1-mgKO mice, there was no increase in REDD1 protein expression, and oxidative stress was reduced compared with diabetic control mice. In both Müller glia within the retina of diabetic mice and human Müller cell cultures exposed to hyperglycemic conditions, REDD1 was necessary for increased expression of the gliosis marker glial fibrillary acidic protein. The effect of REDD1 deletion in preventing gliosis was associated with suppression of oxidative stress and required the antioxidant transcription factor nuclear factor erythroid-2-related factor 2 (Nrf2). In contrast to diabetic control mice, diabetic REDD1-mgKO mice did not exhibit retinal thinning, increased markers of neurodegeneration within the retinal ganglion cell layer, or deficits in visual function. Overall, the findings support a key role for Müller glial REDD1 in the failed adaptive response of the retina to diabetes that includes gliosis, neurodegeneration, and impaired vision.
Collapse
Affiliation(s)
- William P. Miller
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Allyson L. Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Shaunaci A. Stevens
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Ashley M. VanCleave
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - David L. Williamson
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
- Kinesiology Program, Penn State Harrisburg, Middletown, PA
| | - Alistair J. Barber
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
- Department of Ophthalmology, Penn State College of Medicine, Hershey, PA
| | - Michael D. Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
- Department of Ophthalmology, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
46
|
Balakrishnan R, Thurmond DC. Mechanisms by Which Skeletal Muscle Myokines Ameliorate Insulin Resistance. Int J Mol Sci 2022; 23:4636. [PMID: 35563026 PMCID: PMC9102915 DOI: 10.3390/ijms23094636] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 12/17/2022] Open
Abstract
The skeletal muscle is the largest organ in the body and secretes circulating factors, including myokines, which are involved in various cellular signaling processes. Skeletal muscle is vital for metabolism and physiology and plays a crucial role in insulin-mediated glucose disposal. Myokines have autocrine, paracrine, and endocrine functions, serving as critical regulators of myogenic differentiation, fiber-type switching, and maintaining muscle mass. Myokines have profound effects on energy metabolism and inflammation, contributing to the pathophysiology of type 2 diabetes (T2D) and other metabolic diseases. Myokines have been shown to increase insulin sensitivity, thereby improving glucose disposal and regulating glucose and lipid metabolism. Many myokines have now been identified, and research on myokine signaling mechanisms and functions is rapidly emerging. This review summarizes the current state of the field regarding the role of myokines in tissue cross-talk, including their molecular mechanisms, and their potential as therapeutic targets for T2D.
Collapse
Affiliation(s)
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA;
| |
Collapse
|
47
|
Pitale PM, Gorbatyuk MS. Diabetic Retinopathy: From Animal Models to Cellular Signaling. Int J Mol Sci 2022; 23:ijms23031487. [PMID: 35163410 PMCID: PMC8835767 DOI: 10.3390/ijms23031487] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/24/2022] Open
Abstract
Diabetic retinopathy (DR) is an ocular complication of diabetes mellitus (DM), a metabolic disorder characterized by elevation in blood glucose level. The pathogenesis of DR includes vascular, neuronal, and inflammatory components leading to activation of complex cellular molecular signaling. If untreated, the disease can culminate in vision loss that eventually leads to blindness. Animal models mimicking different aspects of DM complications have been developed to study the development and progression of DR. Despite the significant contribution of the developed DR models to discovering the mechanisms of DR and the recent achievements in the research field, the sequence of cellular events in diabetic retinas is still under investigation. Partially, this is due to the complexity of molecular mechanisms, although the lack of availability of models that adequately mimic all the neurovascular pathobiological features observed in patients has also contributed to the delay in determining a precise molecular trigger. In this review, we provide an update on the status of animal models of DR to help investigators choose an appropriate system to validate their hypothesis. We also discuss the key cellular and physiological events of DR in these models.
Collapse
Affiliation(s)
- Priyamvada M. Pitale
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Marina S. Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Correspondence: ; Tel.: +1-205-934-6762; Fax: +1-205-934-3425
| |
Collapse
|
48
|
Daniels Gatward LF, Kennard MR, Smith LIF, King AJF. The use of mice in diabetes research: The impact of physiological characteristics, choice of model and husbandry practices. Diabet Med 2021; 38:e14711. [PMID: 34614258 DOI: 10.1111/dme.14711] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus is characterised by hyperglycaemia, which results from an absolute or relative lack of insulin. Chronic and acute hyperglycaemia are associated with a range of health complications and an overall increased risk of mortality. Mouse models are vital in understanding the pathogenesis of this disease and its complications, as well as for developing new diabetes therapeutics. However, for experimental questions to be suitably tested, it is critical that factors inherent to the animal model are considered, as these can have profound impacts on experimental outcome, data reproducibility and robustness. In this review, we discuss key considerations relating to model choice, physiological characteristics (such as age, sex and genetic background) and husbandry practices and explore the impact of these on common experimental readouts used in preclinical diabetes research.
Collapse
|
49
|
Fu SH, Lai MC, Zheng YY, Sun YW, Qiu JJ, Gui F, Zhang Q, Liu F. MiR-195 inhibits the ubiquitination and degradation of YY1 by Smurf2, and induces EMT and cell permeability of retinal pigment epithelial cells. Cell Death Dis 2021; 12:708. [PMID: 34267179 PMCID: PMC8282777 DOI: 10.1038/s41419-021-03956-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022]
Abstract
The dysregulated microRNAs (miRNAs) are involved in diabetic retinopathy progression. Epithelial mesenchymal transition (EMT) and cell permeability are important events in diabetic retinopathy. However, the function and mechanism of miR-195 in EMT and cell permeability in diabetic retinopathy remain largely unclear. Diabetic retinopathy models were established using streptozotocin (STZ)-induced diabetic mice and high glucose (HG)-stimulated ARPE-19 cells. Retina injury was investigated by hematoxylin-eosin (HE) staining. EMT and cell permeability were analyzed by western blotting, immunofluorescence, wound healing, and FITC-dextran assays. MiR-195 expression was detected via qRT-PCR. YY1, VEGFA, Snail1, and Smurf2 levels were detected via western blotting. The interaction relationship was analyzed via ChIP, Co-IP, or dual-luciferase reporter assay. The retina injury, EMT, and cell permeability were induced in STZ-induced diabetic mice. HG induced EMT and cell permeability in ARPE-19 cells. MiR-195, YY1, VEGFA, and Snail1 levels were enhanced, but Smurf2 abundance was reduced in STZ-induced diabetic mice and HG-stimulated ARPE-19 cells. VEGFA knockdown decreased Snail1 expression and attenuated HG-induced EMT and cell permeability. YY1 silence reduced VEGFA and Snail1 expression, and mitigated HG-induced EMT and cell permeability. YY1 could bind with VEGFA and Snail1, and it was degraded via Smurf2-mediated ubiquitination. MiR-195 knockdown upregulated Smurf2 to decrease YY1 expression and inhibited HG-induced EMT and cell permeability. MiR-195 targeted Smurf2, increased expression of YY1, VEGFA, and Snail1, and promoted HG-induced EMT and cell permeability. MiR-195 promotes EMT and cell permeability of HG-stimulated ARPE-19 cells by increasing VEGFA/Snail1 via inhibiting the Smurf2-mediated ubiquitination of YY1.
Collapse
Affiliation(s)
- Shu-Hua Fu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China
| | - Mei-Chen Lai
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China
| | - Yun-Yao Zheng
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China
| | - Ya-Wen Sun
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China
| | - Jing-Jing Qiu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China
| | - Fu Gui
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China
| | - Qian Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China
| | - Fei Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, P. R. China.
| |
Collapse
|
50
|
Choi JSY, de Haan JB, Sharma A. Animal models of diabetes-associated vascular diseases: an update on available models and experimental analysis. Br J Pharmacol 2021; 179:748-769. [PMID: 34131901 DOI: 10.1111/bph.15591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/08/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetes is a chronic metabolic disorder associated with the accelerated development of macrovascular (atherosclerosis and coronary artery disease) and microvascular complications (nephropathy, retinopathy and neuropathy), which remain the principal cause of mortality and morbidity in this population. Current understanding of cellular and molecular pathways of diabetes-driven vascular complications, as well as therapeutic interventions has arisen from studying disease pathogenesis in animal models. Diabetes-associated vascular complications are multi-faceted, involving the interaction between various cellular and molecular pathways. Thus, the choice of an appropriate animal model to study vascular pathogenesis is important in our quest to identify innovative and mechanism-based targeted therapies to reduce the burden of diabetic complications. Herein, we provide up-to-date information on available mouse models of both Type 1 and Type 2 diabetic vascular complications as well as experimental analysis and research outputs.
Collapse
Affiliation(s)
- Judy S Y Choi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Judy B de Haan
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia.,Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Arpeeta Sharma
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Diabetes, Monash University, Central Clinical School, Melbourne, Victoria, Australia
| |
Collapse
|