1
|
Shariffi B, Harper JL, McMillan NJ, Gonsalves AM, Bond BJ, Pipkins AM, Shoemaker LN, Manrique-Acevedo C, Padilla J, Limberg JK. Effect of insulin on indices of cerebral blood flow and cerebrovascular compliance in young adults. Am J Physiol Heart Circ Physiol 2025; 328:H21-H28. [PMID: 39584591 PMCID: PMC11901331 DOI: 10.1152/ajpheart.00668.2024] [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: 09/25/2024] [Revised: 10/25/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
Insulin has important vasodilatory effects in the peripheral circulation, but less is known about insulin's role in cerebrovascular control. Herein, we hypothesized both systemic (intravenous) and local (intranasal) insulin administration would increase indices of cerebral blood flow and reduce cerebrovascular compliance (Ci) in young adults. Participants were assigned to one of four separate protocols. Middle cerebral artery blood velocity (MCAv, transcranial Doppler ultrasound) and blood pressure (BP, finger photoplethysmography) were measured at baseline and at 1) 2 min of carbon dioxide (CO2) air breathing (high flow control), 2) 60 min of euglycemic intravenous insulin infusion (40 mU/m2 body surface area/min), 3) 60 min following 160 IU of intranasal insulin, 4) 60 minutes of time control. Ci was calculated (modified Windkessel model). Intravenous insulin increased serum insulin (6.0 ± 2.6 to 52.7 ± 12.7 μIU/mL, P < 0.001), whereas serum insulin was reduced following intranasal insulin (6.9 ± 4.5 to 4.9 ± 1.8 μIU/mL, P = 0.030). MCAv increased in response to CO2 (60 ± 13 to 69 ± 11 cm/s, P < 0.001) but was unchanged with time control (50 ± 7 to 49 ± 8, P = 0.658) and both insulin conditions (intravenous: 61 ± 13 to 62 ± 17 cm/s, P = 0.531; intranasal: 57 ± 12 to 51 ± 15 cm/s; p = 0.061). In contrast, Ci remained at baseline levels over time (P = 0.438) and was reduced from baseline under CO2 and both insulin conditions (CO2, P < 0.001; intravenous, P = 0.021; intranasal, P = 0.001). Contrary to our hypothesis, there was no effect of systemic or local insulin administration on resting MCAv in young adults; however, both systemic and local insulin administration reduced Ci. These findings advance our understanding of the cerebrovascular response to acute insulin exposure.NEW & NOTEWORTHY Insulin has important vasodilatory effects in the peripheral circulation, but less is known about the role of insulin in cerebrovascular control. Contrary to our hypothesis, there was no effect of systemic (intravenous) nor local (intranasal) insulin administration on middle cerebral artery blood velocity; however, both systemic and local insulin administration reduced cerebrovascular compliance. Our findings advance our understanding of the cerebrovascular response to insulin and may have implications in the context of known metabolic disturbances.
Collapse
Affiliation(s)
- Brian Shariffi
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Jennifer L Harper
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Neil J McMillan
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | - Anna M Gonsalves
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Braden J Bond
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Aubrey M Pipkins
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Leena N Shoemaker
- School of Kinesiology, Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Camila Manrique-Acevedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Research Services, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Research Services, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
| | - Jacqueline K Limberg
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
2
|
Luchkanych AMS, Morse CJ, Boyes NG, Khan MR, Marshall RA, Morton JS, Tomczak CR, Olver TD. Cerebral sympatholysis: experiments on in vivo cerebrovascular regulation and ex vivo cerebral vasomotor control. Am J Physiol Heart Circ Physiol 2024; 326:H1105-H1116. [PMID: 38391313 DOI: 10.1152/ajpheart.00714.2023] [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: 11/13/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Whether cerebral sympathetic-mediated vasomotor control can be modulated by local brain activity remains unknown. This study tested the hypothesis that the application or removal of a cognitive task during a cold pressor test (CPT) would attenuate and restore decreases in cerebrovascular conductance (CVC), respectively. Middle cerebral artery blood velocity (transcranial Doppler) and mean arterial pressure (finger photoplethysmography) were examined in healthy adults (n = 16; 8 females and 8 males) who completed a control CPT, followed by a CPT coupled with a cognitive task administered either 1) 30 s after the onset of the CPT and for the duration of the CPT or 2) at the onset of the CPT and terminated 30 s before the end of the CPT (condition order was counterbalanced). The major finding was that the CPT decreased the index of CVC, and such decreases were abolished when a cognitive task was completed concurrently and restored when the cognitive task was removed. As a secondary experiment, vasomotor interactions between sympathetic transduction pathways (α1-adrenergic and Y1-peptidergic) and compounds implicated in cerebral blood flow control [adenosine, and adenosine triphosphate (ATP)] were explored in isolated porcine cerebral arteries (wire myography). The data reveal α1-receptor agonism potentiated vasorelaxation modestly in response to adenosine, and preexposure to ATP attenuated contractile responses to α1-agonism. Overall, the data suggest a cognitive task attenuates decreases in CVC during sympathoexcitation, possibly related to an interaction between purinergic and α1-adrenergic signaling pathways.NEW & NOTEWORTHY The present study demonstrates that the cerebrovascular conductance index decreases during sympathoexcitation and this response can be positively and negatively modulated by the application or withdrawal of a nonexercise cognitive task. Furthermore, isolated vessel experiments reveal that cerebral α1-adrenergic agonism potentiates adenosine-mediated vasorelaxation and ATP attenuates α1-adrenergic-mediated vasocontraction.
Collapse
Affiliation(s)
- Adam M S Luchkanych
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cameron J Morse
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Natasha G Boyes
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - M Rafique Khan
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Rory A Marshall
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Jude S Morton
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Corey R Tomczak
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - T Dylan Olver
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
3
|
Kelty TJ, Taylor CL, Wieschhaus NE, Thorne PK, Amin AR, Mueller CM, Olver TD, Tharp DL, Emter CA, Caulk AW, Rector RS. Western diet-induced obesity results in brain mitochondrial dysfunction in female Ossabaw swine. Front Mol Neurosci 2023; 16:1320879. [PMID: 38163062 PMCID: PMC10755880 DOI: 10.3389/fnmol.2023.1320879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
Diet-induced obesity is implicated in the development of a variety of neurodegenerative disorders. Concurrently, the loss of mitochondrial Complex I protein or function is emerging as a key phenotype across an array of neurodegenerative disorders. Therefore, the objective of this study was to determine if Western diet (WD) feeding in swine [carbohydrate = 40.8% kCal (17.8% of total calories from high fructose corn syrup), protein = 16.2% kcal, fat = 42.9% kCal, and 2% cholesterol] would result in Complex I syndrome pathology. To characterize the effects of WD-induced obesity on brain mitochondria in swine, high resolution respirometry measurements from isolated brain mitochondria, oxidative phosphorylation Complex expression, and indices of oxidative stress and mitochondrial biogenesis were assessed in female Ossabaw swine fed a WD for 6-months. In line with Complex I syndrome, WD feeding severely reduced State 3 Complex I, State 3 Complex I and II, and uncoupled mitochondrial respiration in the hippocampus and prefrontal cortex (PFC). State 3 Complex I mitochondrial respiration in the PFC inversely correlated with serum total cholesterol. WD feeding also significantly reduced protein expression of oxidative phosphorylation Complexes I-V in the PFC. WD feeding significantly increased markers of antioxidant defense and mitochondrial biogenesis in the hippocampi and PFC. These data suggest WD-induced obesity may contribute to Complex I syndrome pathology by increasing oxidative stress, decreasing oxidative phosphorylation Complex protein expression, and reducing brain mitochondrial respiration. Furthermore, these findings provide mechanistic insight into the clinical link between obesity and mitochondrial Complex I related neurodegenerative disorders.
Collapse
Affiliation(s)
- Taylor J. Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Chris L. Taylor
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | | | - Pamela K. Thorne
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Amira R. Amin
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Christina M. Mueller
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - T. Dylan Olver
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darla L. Tharp
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Craig A. Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | | | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, United States
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
4
|
Alluri K, Srinivas B, Belmadani S, Matrougui K. Plasmacytoid dendritic cells contribute to vascular endothelial dysfunction in type 2 diabetes. Front Cardiovasc Med 2023; 10:1222243. [PMID: 38094119 PMCID: PMC10716216 DOI: 10.3389/fcvm.2023.1222243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/09/2023] [Indexed: 02/01/2024] Open
Abstract
Objective Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular disease due to macro- and microvascular dysfunction. This study aimed to investigate the potential involvement of plasmacytoid dendritic cells (pDCs) in T2D-related vascular dysfunction. Approach and results pDCs were isolated from db/db and control mice. It was found that pDCs from db/db mice impaired endothelial cell eNOS phosphorylation in response to ATP and decreased vascular endothelium-dependent relaxation compared to pDCs from control mice. Moreover, isolated CD4+ cells from control mice, when stimulated overnight with high glucose and lipids, and isolated pDCs from db/db mice, display elevated levels of ER stress, inflammation, and apoptosis markers. Flow cytometry revealed that pDC frequency was higher in db/db mice than in controls. In vivo, the reduction of pDCs using anti-PDCA-1 antibodies in male and female db/db mice for 4 weeks significantly improved vascular endothelial function and eNOS phosphorylation. Conclusion pDCs may contribute to vascular dysfunction in T2D by impairing endothelial cell function. Targeting pDCs with anti-PDCA-1 antibodies may represent a promising therapeutic strategy for improving vascular endothelial function in T2D patients. This study provides new insights into the pathogenesis of T2D-related vascular dysfunction and highlights the potential of immunomodulatory therapies for treating this complication. Further studies are warranted to explore the clinical potential of this approach.
Collapse
Affiliation(s)
| | | | | | - K. Matrougui
- Department of Physiological Sciences, EVMS, Norfolk, VA, United States
| |
Collapse
|
5
|
Manrique-Acevedo C, Soares RN, Smith JA, Park LK, Burr K, Ramirez-Perez FI, McMillan NJ, Ferreira-Santos L, Sharma N, Olver TD, Emter CA, Parks EJ, Limberg JK, Martinez-Lemus LA, Padilla J. Impact of sex and diet-induced weight loss on vascular insulin sensitivity in type 2 diabetes. Am J Physiol Regul Integr Comp Physiol 2023; 324:R293-R304. [PMID: 36622084 PMCID: PMC9942885 DOI: 10.1152/ajpregu.00249.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/02/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
Vascular insulin resistance, a major characteristic of obesity and type 2 diabetes (T2D), manifests with blunting of insulin-induced vasodilation. Although there is evidence that females are more whole body insulin sensitive than males in the healthy state, whether sex differences exist in vascular insulin sensitivity is unclear. Also uncertain is whether weight loss can reestablish vascular insulin sensitivity in T2D. The purpose of this investigation was to 1) establish if sex differences in vasodilatory responses to insulin exist in absence of disease, 2) determine whether female sex affords protection against the development of vascular insulin resistance with long-term overnutrition and obesity, and 3) examine if diet-induced weight loss can restore vascular insulin sensitivity in men and women with T2D. First, we show in healthy mice and humans that sex does not influence insulin-induced femoral artery dilation and insulin-stimulated leg blood flow, respectively. Second, we provide evidence that female mice are protected against impairments in insulin-induced dilation caused by overnutrition-induced obesity. Third, we show that men and women exhibit comparable levels of vascular insulin resistance when T2D develops but that diet-induced weight loss is effective at improving insulin-stimulated leg blood flow, particularly in women. Finally, we provide indirect evidence that these beneficial effects of weight loss may be mediated by a reduction in endothelin-1. In aggregate, the present data indicate that female sex confers protection against obesity-induced vascular insulin resistance and provide supportive evidence that, in women with T2D, vascular insulin resistance can be remediated with diet-induced weight loss.
Collapse
Affiliation(s)
- Camila Manrique-Acevedo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
| | - Rogerio N Soares
- NextGen Precision Health, University of Missouri, Columbia, Missouri
| | - James A Smith
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Lauren K Park
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Katherine Burr
- NextGen Precision Health, University of Missouri, Columbia, Missouri
| | | | - Neil J McMillan
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Neekun Sharma
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
| | - T Dylan Olver
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Craig A Emter
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Elizabeth J Parks
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
| | - Jacqueline K Limberg
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
6
|
Padilla J, Manrique-Acevedo C, Martinez-Lemus LA. New insights into mechanisms of endothelial insulin resistance in type 2 diabetes. Am J Physiol Heart Circ Physiol 2022; 323:H1231-H1238. [PMID: 36331555 PMCID: PMC9705017 DOI: 10.1152/ajpheart.00537.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Insulin resistance in the vasculature is a hallmark of type 2 diabetes (T2D), and blunting of insulin-induced vasodilation is its primary consequence. Individuals with T2D exhibit a marked impairment in insulin-induced dilation in resistance arteries across vascular beds. Importantly, reduced insulin-stimulated vasodilation and blood flow to skeletal muscle limits glucose uptake and contributes to impaired glucose control in T2D. The study of mechanisms responsible for the suppressed vasodilatory effects of insulin has been a growing topic of interest for not only its association with glucose control and extension to T2D but also its relationship with cardiovascular disease development and progression. In this mini-review, we integrate findings from recent studies by our group with the existing literature focused on the mechanisms underlying endothelial insulin resistance in T2D.
Collapse
Affiliation(s)
- Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
| | - Camila Manrique-Acevedo
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
7
|
Morse CJ, Boerman EM, McDonald MW, Padilla J, Olver TD. The role of nitric oxide in flow-induced and myogenic responses in 1A, 2A, and 3A branches of the porcine middle cerebral artery. J Appl Physiol (1985) 2022; 133:1228-1236. [PMID: 36227166 PMCID: PMC9715271 DOI: 10.1152/japplphysiol.00209.2022] [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: 04/11/2022] [Revised: 09/13/2022] [Accepted: 10/09/2022] [Indexed: 12/15/2022] Open
Abstract
Myogenic and flow-induced reactivity contribute to cerebral autoregulation, with potentially divergent roles for smaller versus larger arteries. The present study tested the hypotheses that compared with first-order (1A) branches of the middle cerebral artery, second- and third-order branches (2A and 3A, respectively) exhibit greater myogenic reactivity but reduced flow-induced constriction. Furthermore, nitric oxide synthase (NOS) inhibition may amplify myogenic reactivity and abolish instances of flow-induced dilation. Isolated porcine cerebral arteries mounted in a pressure myograph were exposed to incremental increases in intraluminal pressure (40-120 mmHg; n = 41) or flow (1-1,170 µL/min; n = 31). Intraluminal flows were adjusted to achieve 5, 10, 20, and 40 dyn/cm2 of wall shear stress at 60 mmHg. Myogenic tone was greater in 3A versus 1A arteries (P < 0.05). There was an inverse relationship between myogenic reactivity and passive arterial diameter (P < 0.01). NOS inhibition increased basal tone to a lesser extent in 3A versus 1A arteries (P < 0.01) but did not influence myogenic reactivity (P = 0.49). Increasing flow decreased luminal diameter (P ≤ 0.01), with increased vasoconstriction at 10-40 dyn/cm2 of shear stress (P < 0.01). However, relative responses were similar between 1A, 2A, and 3A arteries (P = 0.40) with and without NOS inhibition conditions (P ≥ 0.29). Whereas NOS inhibition increases basal myogenic tone, and myogenic reactivity was less in smaller versus larger arteries (range = ∼100-550 µM), neither NOS inhibition nor luminal diameter influences flow-induced constriction in porcine cerebral arteries.NEW & NOTEWORTHY This study demonstrated size-dependent heterogeneity in myogenic reactivity in porcine cerebral arteries. Smaller branches of the middle cerebral artery exhibited increased myogenic reactivity, but attenuated NOS-dependent increases in myogenic tone compared with larger branches. Flow-dependent regulation does not exhibit the same variation; diameter-independent flow-induced vasoconstrictions occur across all branch orders and are not affected by NOS inhibition. Conceptually, flow-induced vasoconstriction contributes to cerebral autoregulation, particularly in larger arteries with low myogenic tone.
Collapse
Affiliation(s)
- Cameron J Morse
- Department Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Erika M Boerman
- Department Medical Physiology and Pharmacology, University of Missouri, Columbia, Missouri
| | - Matthew W McDonald
- Department Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - T Dylan Olver
- Department Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
8
|
Tune JD, Goodwill AG, Baker HE, Dick GM, Warne CM, Tucker SM, Essajee SI, Bailey CA, Klasing JA, Russell JJ, McCallinhart PE, Trask AJ, Bender SB. Chronic high-rate pacing induces heart failure with preserved ejection fraction-like phenotype in Ossabaw swine. Basic Res Cardiol 2022; 117:50. [PMID: 36222894 PMCID: PMC12010922 DOI: 10.1007/s00395-022-00958-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 09/12/2022] [Accepted: 09/28/2022] [Indexed: 01/31/2023]
Abstract
The lack of pre-clinical large animal models of heart failure with preserved ejection fraction (HFpEF) remains a growing, yet unmet obstacle to improving understanding of this complex condition. We examined whether chronic cardiometabolic stress in Ossabaw swine, which possess a genetic propensity for obesity and cardiovascular complications, produces an HFpEF-like phenotype. Swine were fed standard chow (lean; n = 13) or an excess calorie, high-fat, high-fructose diet (obese; n = 16) for ~ 18 weeks with lean (n = 5) and obese (n = 8) swine subjected to right ventricular pacing (180 beats/min for ~ 4 weeks) to induce heart failure (HF). Baseline blood pressure, heart rate, LV end-diastolic volume, and ejection fraction were similar between groups. High-rate pacing increased LV end-diastolic pressure from ~ 11 ± 1 mmHg in lean and obese swine to ~ 26 ± 2 mmHg in lean HF and obese HF swine. Regression analyses revealed an upward shift in LV diastolic pressure vs. diastolic volume in paced swine that was associated with an ~ twofold increase in myocardial fibrosis and an ~ 50% reduction in myocardial capillary density. Hemodynamic responses to graded hemorrhage revealed an ~ 40% decrease in the chronotropic response to reductions in blood pressure in lean HF and obese HF swine without appreciable changes in myocardial oxygen delivery or transmural perfusion. These findings support that high-rate ventricular pacing of lean and obese Ossabaw swine initiates underlying cardiac remodeling accompanied by elevated LV filling pressures with normal ejection fraction. This distinct pre-clinical tool provides a unique platform for further mechanistic and therapeutic studies of this highly complex syndrome.
Collapse
Affiliation(s)
- Johnathan D Tune
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA.
| | - Adam G Goodwill
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Hana E Baker
- Diabetes and Complications Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Gregory M Dick
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Cooper M Warne
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Selina M Tucker
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Salman I Essajee
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Chastidy A Bailey
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Jessica A Klasing
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Jacob J Russell
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Patricia E McCallinhart
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Aaron J Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shawn B Bender
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
9
|
Zhou B, Jin YQ, He LP. Loss of skeletal muscle mass is not specific to type 2 diabetes. World J Diabetes 2022; 13:665-667. [PMID: 36159228 PMCID: PMC9412854 DOI: 10.4239/wjd.v13.i8.665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/30/2022] [Accepted: 07/06/2022] [Indexed: 02/05/2023] Open
Abstract
Skeletal muscle is a massive insulin-sensitive tissue in the body. Loss of muscle mass is associated with mitochondrial dysfunction, and is often a result of diabetes. Insulin deficiency or insulin resistance can only be seen as reduced skeletal muscle mass. Diabetes is caused by insulin deficiency or insulin resistance; however, insulin resistance is not unique to diabetics. Insulin resistance also exists in many diseases.
Collapse
Affiliation(s)
- Bo Zhou
- School of Medicine, Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Ying-Qi Jin
- School of Medicine, Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Lian-Ping He
- School of Medicine, Taizhou University, Taizhou 318000, Zhejiang Province, China
| |
Collapse
|
10
|
Morton JS, Patton B, Morse CJ, El Karsh Z, Rodrigues LA, Mousseau DD, Ferguson DP, Columbus DA, Weber LP, Olver TD. Altered cerebrovascular regulation in low birthweight swine. Comp Biochem Physiol A Mol Integr Physiol 2022; 267:111163. [PMID: 35151870 DOI: 10.1016/j.cbpa.2022.111163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 10/19/2022]
Abstract
Full-term low birthweight (LBW) offspring exhibit peripheral vascular dysfunction in the postnatal period; however, whether such impairments extend to the cerebrovasculature remains to be elucidated. We used a swine model to test the hypothesis that LBW offspring would exhibit cerebrovascular dysfunction at later stages of life. Offspring from 14 sows were identified as normal birthweight (NBW) or LBW and were assessed at 28 (similar to end of infancy) and 56 (similar to childhood) days of age. LBW swine had lower absolute brain mass, but demonstrated evidence of brain sparing (increased brain mass scaled to body mass) at 56 days of age. The cerebral pulsatility index, based on transcranial Doppler, was increased in LBW swine. Moreover, arterial myography of isolated cerebral arteries revealed impaired vasoreactivity to bradykinin and reduced contribution of nitric oxide (NO) to vasorelaxation in the LBW swine. Immunoblotting demonstrated a lower ratio of phosphorylated-to-total endothelial NO synthase in LBW offspring. This impairment in NO signaling was greater at 28 vs. 56 days of age. Vasomotor responses to sodium nitroprusside (NO-donor) were unaltered, while Leu31, Pro34 neuropeptide Y-induced vasoconstriction was enhanced in LBW swine. Increases in total Y1 receptor protein content in the LBW group were not significant. In summary, LBW offspring displayed signs of cerebrovascular dysfunction at 28 and 56 days of age, evidenced by altered cerebral hemodynamics (reflective of increased impedance) coupled with endothelial dysfunction and altered vasomotor control. Overall, the data reveal that normal variance in birthweight of full-term offspring can influence cerebrovascular function later in life.
Collapse
Affiliation(s)
- Jude S Morton
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Breanna Patton
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Cameron J Morse
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zeyad El Karsh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lucas A Rodrigues
- Department of Animal and Poultry Science, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK, Canada; Prairie Swine Center, Inc., Saskatoon, SK, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - David P Ferguson
- Department of Kinesiology, Michigan State University, East Lansing, MI, USA
| | - Daniel A Columbus
- Department of Animal and Poultry Science, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK, Canada; Prairie Swine Center, Inc., Saskatoon, SK, Canada
| | - Lynn P Weber
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - T Dylan Olver
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
11
|
Pettit-Mee RJ, Power G, Cabral-Amador FJ, Ramirez-Perez FI, Nogueira Soares R, Sharma N, Liu Y, Christou DD, Kanaley JA, Martinez-Lemus LA, Manrique-Acevedo CM, Padilla J. Endothelial HSP72 is not reduced in type 2 diabetes nor is it a key determinant of endothelial insulin sensitivity. Am J Physiol Regul Integr Comp Physiol 2022; 323:R43-R58. [PMID: 35470695 DOI: 10.1152/ajpregu.00006.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Impaired endothelial insulin signaling and consequent blunting of insulin-induced vasodilation is a feature of type 2 diabetes (T2D) that contributes to vascular disease and glycemic dysregulation. However, the molecular mechanisms underlying endothelial insulin resistance remain poorly known. Herein, we tested the hypothesis that endothelial insulin resistance in T2D is attributed to reduced expression of heat shock protein 72(HSP72). HSP72 is a cytoprotective chaperone protein that can be upregulated with heating and is reported to promote insulin sensitivity in metabolically active tissues, in part via inhibition of JNK activity. Accordingly, we further hypothesized that, in T2D individuals, seven days of passive heat treatment via hot water immersion to waist-level would improve leg blood flow responses to an oral glucose load (i.e., endogenous insulin stimulation) via induction of endothelial HSP72. In contrast, we found that: 1) endothelial insulin resistance in T2D mice and humans was not associated with reduced HSP72 in aortas and venous endothelial cells, respectively; 2) after passive heat treatment, improved leg blood flow responses to an oral glucose load did not parallel with increased endothelial HSP72; 3) downregulation of HSP72 (via small-interfering RNA) or upregulation of HSP72 (via heating) in cultured endothelial cells did not impair or enhance insulin signaling, respectively, nor was JNK activity altered. Collectively, these findings do not support the hypothesis that reduced HSP72 is a key driver of endothelial insulin resistance in T2D but provide novel evidence that lower-body heating may be an effective strategy for improving leg blood flow responses to glucose ingestion-induced hyperinsulinemia.
Collapse
Affiliation(s)
- Ryan J Pettit-Mee
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Gavin Power
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | | | | | | | - Neekun Sharma
- Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Ying Liu
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Demetra D Christou
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Luis A Martinez-Lemus
- Department of Medicine, University of Missouri, Columbia, MO, United States.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Camila M Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Division of Endocrinology, Diabetes and Metabolism, Department of Medicine University of Missouri, Columbia, MO, United States.,Research Services, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
12
|
Cluzel GL, Ryan PM, Herisson FM, Caplice NM. High-fidelity porcine models of metabolic syndrome: a contemporary synthesis. Am J Physiol Endocrinol Metab 2022; 322:E366-E381. [PMID: 35224983 DOI: 10.1152/ajpendo.00413.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This review aims to describe and compare porcine models of metabolic syndrome. This syndrome and its associated secondary comorbidities are set to become the greatest challenge to healthcare providers and policy makers in the coming century. However, an incomplete understanding of the pathogenesis has left significant knowledge gaps in terms of efficacious therapeutics. To further our comprehension and, in turn, management of metabolic syndrome, appropriate high-fidelity models of the disease complex are of great importance. In this context, our review aims to assess the most promising porcine models of metabolic syndrome currently available for their similarity to the human phenotype. In addition, we aim to highlight the strengths and shortcomings of each model in an attempt to identify the most appropriate application of each. Although no porcine model perfectly recapitulates the human metabolic syndrome, several pose satisfactory approximations. The Ossabaw miniature swine in particular represents a highly translatable model that develops each of the core parameters of the syndrome with many of the associated secondary comorbidities. Future high-fidelity porcine models of metabolic syndrome need to focus on secondary sequelae replication, which may require extended induction period to reveal.
Collapse
Affiliation(s)
- Gaston L Cluzel
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul M Ryan
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Florence M Herisson
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Noel M Caplice
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Vahedi SM, Salek Ardestani S, Karimi K, Banabazi MH. Weighted single-step GWAS for body mass index and scans for recent signatures of selection in Yorkshire pigs. J Hered 2022; 113:325-335. [DOI: 10.1093/jhered/esac004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/24/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Controlling extra fat deposition is economically favorable in modern swine industry. Understanding the genetic architecture of fat deposition traits such as body mass index (BMI) can help in improving genomic selection for such traits. We utilized a weighted single-step genome-wide association study (WssGWAS) to detect genetic regions and candidate genes associated with BMI in a Yorkshire pig population. Three extended haplotype homozygosity (EHH)-related statistics were also incorporated within a de-correlated composite of multiple signals (DCMS) framework to detect recent selection signatures signals. Overall, the full pedigree consisted of 7,016 pigs, of which 5,561 had BMI records and 598 pigs were genotyped with an 80 K single nucleotide polymorphism (SNP) array. Results showed that the most significant windows (top 15) explained 9.35% of BMI genetic variance. Several genes were detected in regions previously associated with pig fat deposition traits and treated as potential candidate genes for BMI in Yorkshire pigs: FTMT, SRFBP1, KHDRBS3, FOXG1, SOD3, LRRC32, TSKU, ACER3, B3GNT6, CCDC201, ADCY1, RAMP3, TBRG4, CCM2. Signature of selection analysis revealed multiple candidate genes previously associated with various economic traits. However, BMI genetic variance explained by regions under selection pressure was minimal (1.31%). In conclusion, candidate genes associated with Yorkshire pigs’ BMI trait were identified using WssGWAS. Gene enrichment analysis indicated that the identified candidate genes were enriched in the insulin secretion pathway. We anticipate that these results further advance our understanding of the genetic architecture of BMI in Yorkshire pigs and provide information for genomic selection for fat deposition in this breed.
Collapse
Affiliation(s)
- Seyed Milad Vahedi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | | | - Karim Karimi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Mohammad Hossein Banabazi
- Department of Biotechnology, Animal Science Research Institute of Iran, Agricultural Research, Education & Extension Organization, Karaj, Iran
- Department of animal breeding and genetics (HGEN), Centre for Veterinary Medicine and Animal Science (VHC), Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| |
Collapse
|
14
|
Sinha NR, Balne PK, Bunyak F, Hofmann AC, Lim RR, Mohan RR, Chaurasia SS. Collagen matrix perturbations in corneal stroma of Ossabaw mini pigs with type 2 diabetes. Mol Vis 2021; 27:666-678. [PMID: 35002212 PMCID: PMC8684810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 12/05/2021] [Indexed: 11/17/2022] Open
Abstract
Purpose Diabetes mellitus (DM) is a metabolic disorder that affects over 450 million people worldwide. DM is characterized by hyperglycemia, causing severe systemic damage to the heart, kidneys, skin, vasculature, nerves, and eye. Type 2 diabetes (T2DM) constitutes 90% of clinical cases and is the most common cause of blindness in working adults. Also, about 70% of T2DM patients show corneal complications including delayed wound healing, often described as diabetic keratopathy (DK). Despite the increasing severity of DM, the research on DK is bleak. This study investigated cellular morphology and collagen matrix alterations of the diabetic and non-diabetic corneas collected from Ossabaw mini pigs, a T2DM animal model with a "thrifty genotype." Methods Pig corneas were collected from six-month-old Ossabaw miniature pigs fed on a western diet (WD) for ten weeks. The tissues were processed for immunohistochemistry and analyzed using hematoxylin and eosin staining, Mason Trichrome staining, Picrosirus Red staining, Collage I staining, and TUNEL assay. mRNA was prepared to quantify fibrotic gene expression using quantitative reverse-transcriptase PCR (qRT-PCR). Transmission electron microscopy (TEM) was performed to evaluate stromal fibril arrangements to compare collagen dynamics in WD vs. standard diet (SD) fed Ossabaw pig corneas. Results Ossabaw mini pigs fed on a WD for 10 weeks exhibit classic symptoms of metabolic syndrome and hyperglycemia seen in T2DM patients. We observed significant disarray in cornea stromal collagen matrix in Ossabaw mini pigs fed on WD compared to the age-matched mini pigs fed on a standard chow diet using Masson Trichome and Picrosirius Red staining. Furthermore, ultrastructure evaluation using TEM showed alterations in stromal collagen fibril size and organization in diabetic corneas compared to healthy age-matched corneas. These changes were accompanied by significantly decreased levels of Collagen IV and increased expression of matrix metallopeptidase 9 in WD-fed pigs. Conclusions This pilot study indicates that Ossabaw mini pigs fed on WD showed collagen disarray and altered gene expression involved in wound healing, suggesting that corneal stromal collagens are vulnerable to diabetic conditions.
Collapse
Affiliation(s)
- Nishant R. Sinha
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- One-Health Vision Research Program, Departments of Ophthalmology and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Praveen K. Balne
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- One-Health Vision Research Program, Departments of Ophthalmology and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Filiz Bunyak
- Department of Computer Science, University of Missouri, Columbia, MO
| | - Alexandria C. Hofmann
- One-Health Vision Research Program, Departments of Ophthalmology and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Rayne R. Lim
- Department of Ophthalmology, University of Washington, Seattle, WA
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- One-Health Vision Research Program, Departments of Ophthalmology and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO
| | - Shyam S. Chaurasia
- One-Health Vision Research Program, Departments of Ophthalmology and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
- Department of Ophthalmology and Visual Sciences, The Eye Institute, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
15
|
Spooner HC, Derrick SA, Maj M, Manjarín R, Hernandez GV, Tailor DS, Bastani PS, Fanter RK, Fiorotto ML, Burrin DG, La Frano MR, Sikalidis AK, Blank JM. High-Fructose, High-Fat Diet Alters Muscle Composition and Fuel Utilization in a Juvenile Iberian Pig Model of Non-Alcoholic Fatty Liver Disease. Nutrients 2021; 13:nu13124195. [PMID: 34959747 PMCID: PMC8705774 DOI: 10.3390/nu13124195] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a serious metabolic condition affecting millions of people worldwide. A “Western-style diet” has been shown to induce pediatric NAFLD with the potential disruption of skeletal muscle composition and metabolism. To determine the in vivo effect of a “Western-style diet” on pediatric skeletal muscle fiber type and fuel utilization, 28 juvenile Iberian pigs were fed either a control diet (CON) or a high-fructose, high-fat diet (HFF), with or without probiotic supplementation, for 10 weeks. The HFF diets increased the total triacylglycerol content of muscle tissue but decreased intramyocellular lipid (IMCL) content and the number of type I (slow oxidative) muscle fibers. HFF diets induced autophagy as assessed by LC3I and LC3II, and inflammation, as assessed by IL-1α. No differences in body composition were observed, and there was no change in insulin sensitivity, but HFF diets increased several plasma acylcarnitines and decreased expression of lipid oxidation regulators PGC1α and CPT1, suggesting disruption of skeletal muscle metabolism. Our results show that an HFF diet fed to juvenile Iberian pigs produces a less oxidative skeletal muscle phenotype, similar to a detraining effect, and reduces the capacity to use lipid as fuel, even in the absence of insulin resistance and obesity.
Collapse
Affiliation(s)
- Heather C. Spooner
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (H.C.S.); (M.M.); (D.S.T.); (P.S.B.)
| | - Stefani A. Derrick
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (S.A.D.); (M.R.L.F.); (A.K.S.)
| | - Magdalena Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (H.C.S.); (M.M.); (D.S.T.); (P.S.B.)
| | - Rodrigo Manjarín
- Department of Animal Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (R.M.); (G.V.H.)
| | - Gabriella V. Hernandez
- Department of Animal Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (R.M.); (G.V.H.)
| | - Deepali S. Tailor
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (H.C.S.); (M.M.); (D.S.T.); (P.S.B.)
| | - Parisa S. Bastani
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (H.C.S.); (M.M.); (D.S.T.); (P.S.B.)
| | - Rob K. Fanter
- College of Agriculture Food and Environmental Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA;
- Cal Poly Metabolomics Service Center, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Marta L. Fiorotto
- United States Department of Agriculture-Agricultural Research Services, Children’s Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (M.L.F.); (D.G.B.)
| | - Douglas G. Burrin
- United States Department of Agriculture-Agricultural Research Services, Children’s Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Houston, TX 77030, USA; (M.L.F.); (D.G.B.)
| | - Michael R. La Frano
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (S.A.D.); (M.R.L.F.); (A.K.S.)
- Cal Poly Metabolomics Service Center, California Polytechnic State University, San Luis Obispo, CA 93407, USA
- Center for Health Research, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Angelos K. Sikalidis
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (S.A.D.); (M.R.L.F.); (A.K.S.)
| | - Jason M. Blank
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (H.C.S.); (M.M.); (D.S.T.); (P.S.B.)
- Correspondence: ; Tel.: +1-805-756-5629
| |
Collapse
|
16
|
Das UN. Molecular biochemical aspects of salt (sodium chloride) in inflammation and immune response with reference to hypertension and type 2 diabetes mellitus. Lipids Health Dis 2021; 20:83. [PMID: 34334139 PMCID: PMC8327432 DOI: 10.1186/s12944-021-01507-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity, insulin resistance, type 2 diabetes mellitus (T2DM) and hypertension (HTN) are common that are associated with low-grade systemic inflammation. Diet, genetic factors, inflammation, and immunocytes and their cytokines play a role in their pathobiology. But the exact role of sodium, potassium, magnesium and other minerals, trace elements and vitamins in the pathogenesis of HTN and T2DM is not known. Recent studies showed that sodium and potassium can modulate oxidative stress, inflammation, alter the autonomic nervous system and induce dysfunction of the innate and adaptive immune responses in addition to their action on renin-angiotensin-aldosterone system. These actions of sodium, potassium and magnesium and other minerals, trace elements and vitamins are likely to be secondary to their action on pro-inflammatory cytokines IL-6, TNF-α and IL-17 and metabolism of essential fatty acids that may account for their involvement in the pathobiology of insulin resistance, T2DM, HTN and autoimmune diseases.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA, 98604, USA.
| |
Collapse
|
17
|
Baranowski BJ, Allen MD, Nyarko JN, Rector RS, Padilla J, Mousseau DD, Rau CD, Wang Y, Laughlin MH, Emter CA, MacPherson RE, Olver TD. Cerebrovascular insufficiency and amyloidogenic signaling in Ossabaw swine with cardiometabolic heart failure. JCI Insight 2021; 6:143141. [PMID: 34027891 PMCID: PMC8262360 DOI: 10.1172/jci.insight.143141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 04/14/2021] [Indexed: 12/26/2022] Open
Abstract
Individuals with heart failure (HF) frequently present with comorbidities, including obesity, insulin resistance, hypertension, and dyslipidemia. Many patients with HF experience cardiogenic dementia, yet the pathophysiology of this disease remains poorly understood. Using a swine model of cardiometabolic HF (Western diet+aortic banding; WD-AB), we tested the hypothesis that WD-AB would promote a multidementia phenotype involving cerebrovascular dysfunction alongside evidence of Alzheimer’s disease (AD) pathology. The results provide evidence of cerebrovascular insufficiency coupled with neuroinflammation and amyloidosis in swine with experimental cardiometabolic HF. Although cardiac ejection fraction was normal, indices of arterial compliance and cerebral blood flow were reduced, and cerebrovascular regulation was impaired in the WD-AB group. Cerebrovascular dysfunction occurred concomitantly with increased MAPK signaling and amyloidogenic processing (i.e., increased APP, BACE1, CTF, and Aβ40 in the prefrontal cortex and hippocampus) in the WD-AB group. Transcriptomic profiles of the stellate ganglia revealed the WD-AB group displayed an enrichment of gene networks associated with MAPK/ERK signaling, AD, frontotemporal dementia, and a number of behavioral phenotypes implicated in cognitive impairment. These provide potentially novel evidence from a swine model that cerebrovascular and neuronal pathologies likely both contribute to the dementia profile in a setting of cardiometabolic HF.
Collapse
Affiliation(s)
- Bradley J Baranowski
- Department of Health Sciences and.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| | - Matti D Allen
- Department of Physical Medicine and Rehabilitation, School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jennifer Nk Nyarko
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, USA
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Darrell D Mousseau
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Christoph D Rau
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yibin Wang
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Rebecca Ek MacPherson
- Department of Health Sciences and.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
18
|
Baranowski BJ, Hayward GC, Marko DM, MacPherson REK. Examination of BDNF Treatment on BACE1 Activity and Acute Exercise on Brain BDNF Signaling. Front Cell Neurosci 2021; 15:665867. [PMID: 34017238 PMCID: PMC8129185 DOI: 10.3389/fncel.2021.665867] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/09/2021] [Indexed: 11/23/2022] Open
Abstract
Perturbations in metabolism results in the accumulation of beta-amyloid peptides, which is a pathological feature of Alzheimer’s disease. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is the rate limiting enzyme responsible for beta-amyloid production. Obesogenic diets increase BACE1 while exercise reduces BACE1 activity, although the mechanisms are unknown. Brain-derived neurotropic factor (BDNF) is an exercise inducible neurotrophic factor, however, it is unknown if BDNF is related to the effects of exercise on BACE1. The purpose of this study was to determine the direct effect of BDNF on BACE1 activity and to examine neuronal pathways induced by exercise. C57BL/6J male mice were assigned to either a low (n = 36) or high fat diet (n = 36) for 10 weeks. To determine the direct effect of BDNF on BACE1, a subset of mice (low fat diet = 12 and high fat diet n = 12) were used for an explant experiment where the brain tissue was directly treated with BDNF (100 ng/ml) for 30 min. To examine neuronal pathways activated with exercise, mice remained sedentary (n = 12) or underwent an acute bout of treadmill running at 15 m/min with a 5% incline for 120 min (n = 12). The prefrontal cortex and hippocampus were collected 2-h post-exercise. Direct treatment with BDNF resulted in reductions in BACE1 activity in the prefrontal cortex (p < 0.05), but not the hippocampus. The high fat diet reduced BDNF content in the hippocampus; however, the acute bout of exercise increased BDNF in the prefrontal cortex (p < 0.05). These novel findings demonstrate the region specific differences in exercise induced BDNF in lean and obese mice and show that BDNF can reduce BACE1 activity, independent of other exercise-induced alterations. This work demonstrates a previously unknown link between BDNF and BACE1 regulation.
Collapse
Affiliation(s)
| | - Grant C Hayward
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Daniel M Marko
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
19
|
Kelly SC, Rau CD, Ouyang A, Thorne PK, Olver TD, Edwards JC, Domeier TL, Padilla J, Grisanti LA, Fleenor BS, Wang Y, Rector RS, Emter CA. The right ventricular transcriptome signature in Ossabaw swine with cardiometabolic heart failure: implications for the coronary vasculature. Physiol Genomics 2021; 53:99-115. [PMID: 33491589 PMCID: PMC7988741 DOI: 10.1152/physiolgenomics.00093.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) patients with deteriorating right ventricular (RV) structure and function have a nearly twofold increased risk of death compared with those without. Despite the well-established clinical risk, few studies have examined the molecular signature associated with this HF condition. The purpose of this study was to integrate morphological, molecular, and functional data with the transcriptome data set in the RV of a preclinical model of cardiometabolic HF. Ossabaw swine were fed either normal diet without surgery (lean control, n = 5) or Western diet and aortic-banding (WD-AB; n = 4). Postmortem RV weight was increased and positively correlated with lung weight in the WD-AB group compared with CON. Total RNA-seq was performed and gene expression profiles were compared and analyzed using principal component analysis, weighted gene co-expression network analysis, module enrichment analysis, and ingenuity pathway analysis. Gene networks specifically associated with RV hypertrophic remodeling identified a hub gene in MAPK8 (or JNK1) that was associated with the selective induction of the extracellular matrix (ECM) component fibronectin. JNK1 and fibronectin protein were increased in the right coronary artery (RCA) of WD-AB animals and associated with a decrease in matrix metalloproteinase 14 protein, which specifically degrades fibronectin. RCA fibronectin content was correlated with increased vascular stiffness evident as a decreased elastin elastic modulus in WD-AB animals. In conclusion, this study establishes a molecular and transcriptome signature in the RV using Ossabaw swine with cardiometabolic HF. This signature was associated with altered ECM regulation and increased vascular stiffness in the RCA, with selective dysregulation of fibronectin.
Collapse
Affiliation(s)
- Shannon C Kelly
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Christoph D Rau
- Department of Computational Medicine and Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - An Ouyang
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pamela K Thorne
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jenna C Edwards
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Laurel A Grisanti
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Bradley S Fleenor
- Human Performance Laboratory, School of Kinesiology, Ball State University, Muncie, Indiana
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, California
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Department of Medicine-Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial VA Hospital, University of Missouri, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
20
|
Zhao Q, Zhang Y, Liao X, Wang W. Executive Function and Diabetes: A Clinical Neuropsychology Perspective. Front Psychol 2020; 11:2112. [PMID: 32973635 PMCID: PMC7468478 DOI: 10.3389/fpsyg.2020.02112] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/29/2020] [Indexed: 02/05/2023] Open
Abstract
Objective Diabetes is a global public health concern. Management of diabetes depends on successful implementation of strategies to alleviate decline in executive functions (EFs), a characteristic of diabetes progression. In this review, we describe recent research on the relationship between diabetes and EF, summarize the existing evidence, and put forward future research directions and applications. Methods Herein, we provide an overview of recent studies, to elucidate the relationship between DM and EF. We identified new screening objectives, management tools, and intervention targets for diabetes management. We also discuss the implications for clinical practice. Results In both types 1 and 2 diabetes mellitus (DM), hyperglycemia substantially impairs EF in people of all age groups and ethnicities. Hypoglycemia can similarly impair EF. Interestingly, a decline in EF contributes to DM progression. Glucose dysregulation and EF decline exacerbate each other in a vicious cycle: poor blood glucose control, impaired EF, diabetes management task failure, then back to poor blood glucose control. Many pathophysiological indexes (e.g., obesity, metabolic index, inflammatory and immune factors), neuropsychological indexes (e.g., compliance, eating habits, physical exercise, sleep, and depression), and genetic factors are changed by this pathological interaction between DM and EF. These changes can provide insight into the pathophysiological mechanisms of diabetes-related EF decline. Conclusion Further studies, including large-scale prospective and randomized controlled trials, are needed to elucidate the mechanism of the interaction between diabetes and EF and to develop novel strategies for breaking this cycle.
Collapse
Affiliation(s)
- Qian Zhao
- International Medical Center/Ward of General Practice and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyang Liao
- International Medical Center/Ward of General Practice and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Weiwen Wang
- Department of Neurology, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
21
|
Silva KAS, Emter CA. Large Animal Models of Heart Failure: A Translational Bridge to Clinical Success. JACC Basic Transl Sci 2020; 5:840-856. [PMID: 32875172 PMCID: PMC7452204 DOI: 10.1016/j.jacbts.2020.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022]
Abstract
Preclinical large animal models play a critical and expanding role in translating basic science findings to the development and clinical approval of novel cardiovascular therapeutics. This state-of-the-art review outlines existing methodologies and physiological phenotypes of several HF models developed in large animals. A comprehensive list of porcine, ovine, and canine models of disease are presented, and the translational importance of these studies to clinical success is highlighted through a brief overview of recent devices approved by the FDA alongside associated clinical trials and preclinical animal reports. Increasing the use of large animal models of HF holds significant potential for identifying new mechanisms underlying this disease and providing valuable information regarding the safety and efficacy of new therapies, thus, improving physiological and economical translation of animal research to the successful treatment of human HF.
Preclinical large animal models of heart failure (HF) play a critical and expanding role in translating basic science findings to the development and clinical approval of novel therapeutics and devices. The complex combination of cardiovascular events and risk factors leading to HF has proved challenging for the development of new treatments for these patients. This state-of-the-art review presents historical and recent studies in porcine, ovine, and canine models of HF and outlines existing methodologies and physiological phenotypes. The translational importance of large animal studies to clinical success is also highlighted with an overview of recent devices approved by the Food and Drug Administration, together with preclinical HF animal studies used to aid both development and safety and/or efficacy testing. Increasing the use of large animal models of HF holds significant potential for identifying the novel mechanisms underlying the clinical condition and to improving physiological and economical translation of animal research to successfully treat human HF.
Collapse
Key Words
- AF, atrial fibrillation
- ECM, extracellular matrix
- EDP, end-diastolic pressure
- EF, ejection fraction
- FDA, Food and Drug Administration
- HF, heart failure
- HFpEF
- HFpEF, heart failure with preserved ejection fraction
- HFrEF
- HFrEF, heart failure with reduced ejection fraction
- I/R, ischemia/reperfusion
- IABP, intra-aortic balloon pump
- LAD, left anterior descending
- LCx, left circumflex
- LV, left ventricular
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- RV, right ventricular
- heart failure
- large animal model
- preclinical
Collapse
Affiliation(s)
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
22
|
Grunewald ZI, Ramirez-Perez FI, Woodford ML, Morales-Quinones M, Mejia S, Manrique-Acevedo C, Siebenlist U, Martinez-Lemus LA, Chandrasekar B, Padilla J. TRAF3IP2 (TRAF3 Interacting Protein 2) Mediates Obesity-Associated Vascular Insulin Resistance and Dysfunction in Male Mice. Hypertension 2020; 76:1319-1329. [PMID: 32829657 DOI: 10.1161/hypertensionaha.120.15262] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Insulin resistance in the vasculature is a characteristic feature of obesity and contributes to the pathogenesis of vascular dysfunction and disease. However, the molecular mechanisms underlying obesity-associated vascular insulin resistance and dysfunction remain poorly understood. We hypothesized that TRAF3IP2 (TRAF3 interacting protein 2), a proinflammatory adaptor molecule known to activate pathological stress pathways and implicated in cardiovascular diseases, plays a causal role in obesity-associated vascular insulin resistance and dysfunction. We tested this hypothesis by employing genetic-manipulation in endothelial cells in vitro, in isolated arteries ex vivo, and diet-induced obesity in a mouse model of TRAF3IP2 ablation in vivo. We show that ectopic expression of TRAF3IP2 blunts insulin signaling in endothelial cells and diminishes endothelium-dependent vasorelaxation in isolated aortic rings. Further, 16 weeks of high fat/high sucrose feeding impaired glucose tolerance, aortic insulin-induced vasorelaxation, and hindlimb postocclusive reactive hyperemia, while increasing blood pressure and arterial stiffness in wild-type male mice. Notably, TRAF3IP2 ablation protected mice from such high fat/high sucrose feeding-induced metabolic and vascular defects. Interestingly, wild-type female mice expressed markedly reduced levels of TRAF3IP2 mRNA independent of diet and were protected against high fat/high sucrose diet-induced vascular dysfunction. These data indicate that TRAF3IP2 plays a causal role in vascular insulin resistance and dysfunction. Specifically, the present findings highlight a sexual dimorphic role of TRAF3IP2 in vascular control and identify it as a promising therapeutic target in vasculometabolic derangements associated with obesity, particularly in males.
Collapse
Affiliation(s)
- Zachary I Grunewald
- From the Department of Nutrition and Exercise Physiology (Z.I.G., M.L.W., J.P.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia
| | - Makenzie L Woodford
- From the Department of Nutrition and Exercise Physiology (Z.I.G., M.L.W., J.P.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Mariana Morales-Quinones
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Salvador Mejia
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Division of Endocrinology and Metabolism, Department of Medicine (C.M.-A.), University of Missouri, Columbia.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (C.M.-A., B.C.)
| | | | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Department of Biological Engineering (F.I.R.-P., L.A.M.-L.), University of Missouri, Columbia.,Department of Medical Pharmacology and Physiology (L.A.M.-L., B.C.), University of Missouri, Columbia
| | - Bysani Chandrasekar
- Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia.,Division of Cardiovascular Medicine, Department of Medicine (B.C.), University of Missouri, Columbia.,Department of Medical Pharmacology and Physiology (L.A.M.-L., B.C.), University of Missouri, Columbia.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (C.M.-A., B.C.)
| | - Jaume Padilla
- From the Department of Nutrition and Exercise Physiology (Z.I.G., M.L.W., J.P.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (Z.I.G., F.I.R.-P., M.L.W., M.M.-Q., S.M., C.M.-A., L.A.M.-L., B.C., J.P.), University of Missouri, Columbia
| |
Collapse
|
23
|
Smith JR, Hayman GT, Wang SJ, Laulederkind SJF, Hoffman MJ, Kaldunski ML, Tutaj M, Thota J, Nalabolu HS, Ellanki SLR, Tutaj MA, De Pons JL, Kwitek AE, Dwinell MR, Shimoyama ME. The Year of the Rat: The Rat Genome Database at 20: a multi-species knowledgebase and analysis platform. Nucleic Acids Res 2020; 48:D731-D742. [PMID: 31713623 PMCID: PMC7145519 DOI: 10.1093/nar/gkz1041] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022] Open
Abstract
Formed in late 1999, the Rat Genome Database (RGD, https://rgd.mcw.edu) will be 20 in 2020, the Year of the Rat. Because the laboratory rat, Rattus norvegicus, has been used as a model for complex human diseases such as cardiovascular disease, diabetes, cancer, neurological disorders and arthritis, among others, for >150 years, RGD has always been disease-focused and committed to providing data and tools for researchers doing comparative genomics and translational studies. At its inception, before the sequencing of the rat genome, RGD started with only a few data types localized on genetic and radiation hybrid (RH) maps and offered only a few tools for querying and consolidating that data. Since that time, RGD has expanded to include a wealth of structured and standardized genetic, genomic, phenotypic, and disease-related data for eight species, and a suite of innovative tools for querying, analyzing and visualizing this data. This article provides an overview of recent substantial additions and improvements to RGD's data and tools that can assist researchers in finding and utilizing the data they need, whether their goal is to develop new precision models of disease or to more fully explore emerging details within a system or across multiple systems.
Collapse
Affiliation(s)
- Jennifer R Smith
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- To whom correspondence should be addressed. Tel: +1 414 955 8871; Fax: +1 414 955 6595;
| | - G Thomas Hayman
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shur-Jen Wang
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stanley J F Laulederkind
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew J Hoffman
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Genomic Sciences and Precision Medicine Center and Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary L Kaldunski
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monika Tutaj
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jyothi Thota
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Harika S Nalabolu
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Santoshi L R Ellanki
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Marek A Tutaj
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jeffrey L De Pons
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anne E Kwitek
- Genomic Sciences and Precision Medicine Center and Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Melinda R Dwinell
- Genomic Sciences and Precision Medicine Center and Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary E Shimoyama
- Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
24
|
Parry SA, Turner MC, Woods RM, James LJ, Ferguson RA, Cocks M, Whytock KL, Strauss JA, Shepherd SO, Wagenmakers AJM, van Hall G, Hulston CJ. High-Fat Overfeeding Impairs Peripheral Glucose Metabolism and Muscle Microvascular eNOS Ser1177 Phosphorylation. J Clin Endocrinol Metab 2020; 105:5568321. [PMID: 31513265 DOI: 10.1210/clinem/dgz018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/06/2019] [Indexed: 11/19/2022]
Abstract
CONTEXT The mechanisms responsible for dietary fat-induced insulin resistance of skeletal muscle and its microvasculature are only partially understood. OBJECTIVE To determine the impact of high-fat overfeeding on postprandial glucose fluxes, muscle insulin signaling, and muscle microvascular endothelial nitric oxide synthase (eNOS) content and activation. DESIGN Fifteen non-obese volunteers consumed a high-fat (64%) high-energy (+47%) diet for 7 days. Experiments were performed before and after the diet. Stable isotope tracers were used to determine glucose fluxes in response to carbohydrate plus protein ingestion. Muscle insulin signaling was determined as well as the content and activation state of muscle microvascular eNOS. RESULTS High-fat overfeeding impaired postprandial glycemic control as demonstrated by higher concentrations of glucose (+11%; P = 0.004) and insulin (+19%; P = 0.035). Carbohydrate plus protein ingestion suppressed endogenous glucose production to a similar extent before and after the diet. Conversely, high-fat overfeeding reduced whole-body glucose clearance (-16%; P = 0.021) and peripheral insulin sensitivity (-26%; P = 0.006). This occurred despite only minor alterations in skeletal muscle insulin signaling. High-fat overfeeding reduced eNOS content in terminal arterioles (P = 0.017) and abolished the increase in eNOS Ser1177 phosphorylation that was seen after carbohydrate plus protein ingestion. CONCLUSION High-fat overfeeding impaired whole-body glycemic control due to reduced glucose clearance, not elevated endogenous glucose production. The finding that high-fat overfeeding abolished insulin-mediated eNOS Ser1177 phosphorylation in the terminal arterioles suggests that impairments in the vasodilatory capacity of the skeletal muscle microvasculature may contribute to early dietary fat-induced impairments in glycemic control.
Collapse
Affiliation(s)
- Siôn A Parry
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, UK
| | - Mark C Turner
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, UK
- University Hospitals of Leicester NHS Trust, Infirmary Square, Leicester, UK
| | - Rachel M Woods
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, UK
| | - Lewis J James
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, UK
| | - Richard A Ferguson
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, UK
| | - Matthew Cocks
- School of Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Katie L Whytock
- School of Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Juliette A Strauss
- School of Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Sam O Shepherd
- School of Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Anton J M Wagenmakers
- School of Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Gerrit van Hall
- Clinical Metabolomics Core Facility, Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carl J Hulston
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
25
|
Olver TD, Grunewald ZI, Ghiarone T, Restaino RM, Sales ARK, Park LK, Thorne PK, Ganga RR, Emter CA, Lemon PWR, Shoemaker JK, Manrique-Acevedo C, Martinez-Lemus LA, Padilla J. Persistent insulin signaling coupled with restricted PI3K activation causes insulin-induced vasoconstriction. Am J Physiol Heart Circ Physiol 2019; 317:H1166-H1172. [PMID: 31603345 DOI: 10.1152/ajpheart.00464.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Insulin modulates vasomotor tone through vasodilator and vasoconstrictor signaling pathways. The purpose of the present work was to determine whether insulin-stimulated vasoconstriction is a pathophysiological phenomenon that can result from a combination of persistent insulin signaling, suppressed phosphatidylinositol-3 kinase (PI3K) activation, and an ensuing relative increase in MAPK/endothelin-1 (ET-1) activity. First, we examined previously published work from our group where we assessed changes in lower-limb blood flow in response to an oral glucose tolerance test (endogenous insulin stimulation) in lean and obese subjects. The new analyses showed that the peak rise in vascular resistance during the postprandial state was greater in obese compared with lean subjects. We next extended on these findings by demonstrating that insulin-induced vasoconstriction in isolated resistance arteries from obese subjects was attenuated with ET-1 receptor antagonism, thus implicating ET-1 signaling in this constriction response. Last, we examined in isolated resistance arteries from pigs the dual roles of persistent insulin signaling and blunted PI3K activation in modulating vasomotor responses to insulin. We found that prolonged insulin stimulation did not alter vasomotor responses to insulin when insulin-signaling pathways remained unrestricted. However, prolonged insulinization along with pharmacological suppression of PI3K activity resulted in insulin-induced vasoconstriction, rather than vasodilation. Notably, such aberrant vascular response was rescued with either MAPK inhibition or ET-1 receptor antagonism. In summary, we demonstrate that insulin-induced vasoconstriction is a pathophysiological phenomenon that can be recapitulated when sustained insulin signaling is coupled with depressed PI3K activation and the concomitant relative increase in MAPK/ET-1 activity.NEW & NOTEWORTHY This study reveals that insulin-induced vasoconstriction is a pathophysiological phenomenon. We also provide evidence that in the setting of persistent insulin signaling, impaired phosphatidylinositol-3 kinase activation appears to be a requisite feature precipitating MAPK/endothelin 1-dependent insulin-induced vasoconstriction.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatchewan, Canada
| | - Zachary I Grunewald
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Robert M Restaino
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, New York
| | - Allan R K Sales
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil.,D'Or Institute for Research and Education, São Paulo, Brazil
| | - Lauren K Park
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Pamela K Thorne
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Rama Rao Ganga
- Department of Surgery, University of Missouri, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Peter W R Lemon
- School of Kinesiology, The University of Western Ontario, London, Ontario, Canada
| | - J Kevin Shoemaker
- School of Kinesiology, The University of Western Ontario, London, Ontario, Canada
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri.,Research Services, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
26
|
Hayward GC, LeBlanc PJ, Emter CA, Nyarko JNK, Mousseau DD, MacPherson REK, Olver TD. Female Sex Hormones and Cardiac Pressure Overload Independently Contribute to the Cardiogenic Dementia Profile in Yucatan Miniature Swine. Front Cardiovasc Med 2019; 6:129. [PMID: 31552273 PMCID: PMC6746895 DOI: 10.3389/fcvm.2019.00129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/19/2019] [Indexed: 01/29/2023] Open
Abstract
Post-menopausal women with heart failure (HF) frequently exhibit cardiogenic dementia. Using a pre-clinical swine model of post-menopausal HF, we recently demonstrated that experimental menopause (ovariectomy; OVX) and HF (6-month cardiac pressure overload/aortic banding; AB) independently altered cerebral vasomotor control and together impaired cognitive function. The purpose of this study was to examine the prefrontal cortex and hippocampus tissues from these animals to assess whether OVX and HF are associated with neurologic alterations that may contribute to cardiogenic dementia. We hypothesized that OVX and HF would independently alter neuronal cell signaling in swine with post-menopausal cardiogenic dementia. Immunoblot analyses revealed OVX was associated with reduced estrogen receptor-α in both brain regions and HF tended to exacerbate OVX-induced deficits in the hippocampus. Further, OVX was associated with a reduction in the ratio of phosphorylated:total Akt and ERK in the hippocampus as well as decreased total Akt and synaptophysin in the prefrontal cortex. In contrast, HF was associated with a trend toward reduced phosphorylated:total ERK in the prefrontal cortex. In addition, HF was associated with decreased β-amyloid (1-38) in the prefrontal cortex and increased β-amyloid (1-38) in the hippocampus. Regional brain lipid analysis revealed OVX tended to increase total, saturated, and monounsaturated fatty acid content in the prefrontal cortex, with the greatest magnitude of change occurring in the AB-OVX group. The data from this study suggest that OVX and HF are independently associated with regional-specific neurologic changes in the brain that contribute to the cardiogenic dementia profile in this model. This pre-clinical swine model may be a useful tool for better understanding post-menopausal cardiogenic dementia pathology and developing novel therapies.
Collapse
Affiliation(s)
- Grant C. Hayward
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| | - Paul J. LeBlanc
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Craig A. Emter
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Jennifer N. K. Nyarko
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darrell D. Mousseau
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Rebecca E. K. MacPherson
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| | - T. Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
27
|
The Postprandial Appearance of Features of Cardiometabolic Risk: Acute Induction and Prevention by Nutrients and Other Dietary Substances. Nutrients 2019; 11:nu11091963. [PMID: 31438565 PMCID: PMC6770341 DOI: 10.3390/nu11091963] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
The purpose of this review is to provide an overview of diets, food, and food components that affect postprandial inflammation, endothelial function, and oxidative stress, which are related to cardiometabolic risk. A high-energy meal, rich in saturated fat and sugars, induces the transient appearance of a series of metabolic, signaling and physiological dysregulations or dysfunctions, including oxidative stress, low-grade inflammation, and endothelial dysfunction, which are directly related to the amplitude of postprandial plasma triglycerides and glucose. Low-grade inflammation and endothelial dysfunction are also known to cluster together with insulin resistance, a third risk factor for cardiovascular diseases (CVD) and type-II diabetes, thus making a considerable contribution to cardiometabolic risk. Because of the marked relevance of the postprandial model to nutritional pathophysiology, many studies have investigated whether adding various nutrients and other substances to such a challenge meal might mitigate the onset of these adverse effects. Some foods (e.g., nuts, berries, and citrus), nutrients (e.g., l-arginine), and other substances (various polyphenols) have been widely studied. Reports of favorable effects in the postprandial state have concerned plasma markers for systemic or vascular pro-inflammatory conditions, the activation of inflammatory pathways in plasma monocytes, vascular endothelial function (mostly assessed using physiological criteria), and postprandial oxidative stress. Although the literature is fragmented, this topic warrants further study using multiple endpoints and markers to investigate whether the interesting candidates identified might prevent or limit the postprandial appearance of critical features of cardiometabolic risk.
Collapse
|
28
|
Increased coronary arteriolar contraction to serotonin in juvenile pigs with metabolic syndrome. Mol Cell Biochem 2019; 461:57-64. [PMID: 31352610 PMCID: PMC6790190 DOI: 10.1007/s11010-019-03589-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/13/2019] [Indexed: 02/03/2023]
Abstract
Metabolic syndrome (MetS) is associated with alterations in coronary vascular smooth muscle and endothelial function. The current study examined the contractile response of the isolated coronary arterioles to serotonin in pigs with and without MetS and investigated the signaling pathways responsible for serotonin-induced vasomotor tone. The MetS pigs (8-weeks old) were fed with a hyper-caloric, fat/cholesterol diet and the control animals (lean) were fed with a regular diet for 12 weeks (n = 6/group). The coronary arterioles (90–180 μm in diameter) were dissected from the harvested pig myocardial tissues and the in vitro coronary arteriolar response to serotonin was measured in the presence of pharmacological inhibitors. The protein expressions of phospholipase A2 (PLA2), TXA2 synthase, and the thromboxane-prostanoid (TP) receptor in the pigs’ left ventricular tissue samples were measured using Western blotting. Serotonin (10−9–10−5 M) induced dose-dependent contractions of coronary-resistant arterioles in both non-MetS control (lean) and MetS pigs. This effect was more pronounced in the MetS vessels compared with those of non-MetS controls (lean, P < 0.05]. Serotonin-induced contraction of the MetS vessels was significantly inhibited in the presence of the selective PLA2 inhibitor quinacrine (10−6 M), the COX inhibitor indomethacin (10−5 M), and the TP receptor antagonist SQ29548 (10−6 M), respectively (P < 0.05). MetS exhibited significant increases in tissue levels of TXA2 synthase and TP receptors (P < 0.05 vs. lean), respectively. MetS is associated with increased contractile response of porcine coronary arterioles to serotonin, which is in part via upregulation/activation of PLA2, COX, and subsequent TXA2, suggesting that alteration of vasomotor function may occur at an early stage of MetS and juvenile obesity.
Collapse
|
29
|
Affiliation(s)
- Thomas E. Sharp
- Cardiovascular Center of Excellence, School of Medicine, LSU Health Science Center, New Orleans, Louisiana
| | - David J. Lefer
- Cardiovascular Center of Excellence, School of Medicine, LSU Health Science Center, New Orleans, Louisiana
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Steven R. Houser
- Department of Physiology and Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Pennsylvania
| |
Collapse
|
30
|
Olver TD, Edwards JC, Jurrissen TJ, Veteto AB, Jones JL, Gao C, Rau C, Warren CM, Klutho PJ, Alex L, Ferreira-Nichols SC, Ivey JR, Thorne PK, McDonald KS, Krenz M, Baines CP, Solaro RJ, Wang Y, Ford DA, Domeier TL, Padilla J, Rector RS, Emter CA. Western Diet-Fed, Aortic-Banded Ossabaw Swine: A Preclinical Model of Cardio-Metabolic Heart Failure. JACC Basic Transl Sci 2019; 4:404-421. [PMID: 31312763 PMCID: PMC6610000 DOI: 10.1016/j.jacbts.2019.02.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
The development of new treatments for heart failure lack animal models that encompass the increasingly heterogeneous disease profile of this patient population. This report provides evidence supporting the hypothesis that Western Diet-fed, aortic-banded Ossabaw swine display an integrated physiological, morphological, and genetic phenotype evocative of cardio-metabolic heart failure. This new preclinical animal model displays a distinctive constellation of findings that are conceivably useful to extending the understanding of how pre-existing cardio-metabolic syndrome can contribute to developing HF.
Collapse
Key Words
- AB, aortic-banded
- CON, control
- EDPVR, end-diastolic pressure−volume relationship
- EF, ejection fraction
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IL1RL1, interleukin 1 receptor-like 1
- LV, left ventricle
- NF, nuclear factor
- PTX3, pentraxin-3
- WD, Western Diet
- cardio-metabolic disease
- heart failure
- integrative pathophysiology
- preclinical model of cardiovascular disease
Collapse
Affiliation(s)
- T. Dylan Olver
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Jenna C. Edwards
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Thomas J. Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Adam B. Veteto
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - John L. Jones
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Chen Gao
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Christoph Rau
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chad M. Warren
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Paula J. Klutho
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Linda Alex
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | | | - Jan R. Ivey
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Pamela K. Thorne
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Christopher P. Baines
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David A. Ford
- Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University- School of Medicine, St. Louis, Missouri
| | - Timothy L. Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
- Department of Child Health, University of Missouri-Columbia, Columbia, Missouri
| | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Department of Medicine – University of Missouri-Columbia, Columbia, Missouri
- Research Service, Harry S Truman Memorial VA Hospital, University of Missouri-Columbia, Columbia, Missouri
| | - Craig A. Emter
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
31
|
Olver TD, Laughlin MH, Padilla J. Exercise and Vascular Insulin Sensitivity in the Skeletal Muscle and Brain. Exerc Sport Sci Rev 2019; 47:66-74. [PMID: 30883470 DOI: 10.1249/jes.0000000000000182] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We present the hypothesis that exercise-induced hyperemia, perhaps through vascular shear stress, represents an important factor responsible for the effects of physical activity (PA) on vascular insulin sensitivity. Specifically, we postulate PA involving the greatest amount of skeletal muscle mass and the greatest central neural recruitment maximizes perfusion and consequently enhances vascular insulin sensitivity in the skeletal muscle and brain.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - M Harold Laughlin
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center
| | - Jaume Padilla
- Dalton Cardiovascular Research Center.,Department of Nutrition and Exercise Physiology, and.,Department of Child Health, University of Missouri, Columbia, MO
| |
Collapse
|
32
|
Lim RR, Grant DG, Olver TD, Padilla J, Czajkowski AM, Schnurbusch TR, Mohan RR, Hainsworth DP, Walters EM, Chaurasia SS. Young Ossabaw Pigs Fed a Western Diet Exhibit Early Signs of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2019; 59:2325-2338. [PMID: 29847637 PMCID: PMC5937800 DOI: 10.1167/iovs.17-23616] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Recent clinical data suggest an increasing prevalence of obesity and type 2 diabetes in adolescents, placing them at high risk of developing diabetic retinopathy during adult working years. The present study was designed to characterize the early retinal and microvascular alterations in young Ossabaw pigs fed a Western diet, described as a model of metabolic syndrome genetically predisposed to type 2 diabetes. Methods Four-month-old Ossabaw miniature pigs were divided into two groups, lean and diet-induced obesity. Obese pigs were fed a Western diet with high-fat/high-fructose corn syrup/high-choleric content for 10 weeks. Blood and retina were collected for biochemical profiling, trypsin digest, flatmounts, Fluoro-Jade C staining, electron microscopy, quantitative PCR, immunohistochemistry, and Western blots. Results Young Ossabaw pigs had elevated fasting blood glucose after feeding on a Western diet for 10 weeks. Their retina showed disrupted cellular architecture across neural layers, with numerous large vacuoles seen in cell bodies of the inner nuclear layer. Microvessels in the obese animals exhibited thickened basement membrane, along with pericyte ghosts and acellular capillaries. The pericyte to endothelial ratio decreased significantly. Retina flatmounts from obese pigs displayed reduced capillary density, numerous terminal capillary loops, and string vessels, which stained collagen IV but not isolectin IB4. Quantitative PCR and Western blots showed significantly high levels of basement membrane proteins collagen IV and fibronectin in obese pigs. Conclusions This is the first study to describe the ultrastructural neuronal and vascular changes in the retina of young Ossabaw pigs fed a Western diet, simulating early signs of diabetic retinopathy pathogenesis.
Collapse
Affiliation(s)
- Rayne R Lim
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, Missouri, United States.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States.,Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States
| | - DeAna G Grant
- Electron Microscopy Core, University of Missouri, Columbia, Missouri, United States
| | - T Dylan Olver
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States.,Child Health, University of Missouri, Columbia, Missouri, United States
| | - Alana M Czajkowski
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri, United States
| | - Teagan R Schnurbusch
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri, United States
| | - Rajiv R Mohan
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, Missouri, United States.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States.,Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States.,Mason Eye Institute, University of Missouri, Columbia, Missouri, United States
| | - Dean P Hainsworth
- Mason Eye Institute, University of Missouri, Columbia, Missouri, United States
| | - Eric M Walters
- National Swine Resource and Research Center, University of Missouri, Columbia, Missouri, United States
| | - Shyam S Chaurasia
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, Missouri, United States.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States.,Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States
| |
Collapse
|
33
|
Winn NC, Jurrissen TJ, Grunewald ZI, Cunningham RP, Woodford ML, Kanaley JA, Lubahn DB, Manrique-Acevedo C, Rector RS, Vieira-Potter VJ, Padilla J. Estrogen receptor-α signaling maintains immunometabolic function in males and is obligatory for exercise-induced amelioration of nonalcoholic fatty liver. Am J Physiol Endocrinol Metab 2019; 316:E156-E167. [PMID: 30512987 PMCID: PMC6397364 DOI: 10.1152/ajpendo.00259.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The role of estrogen receptor-α (ERα) signaling in immunometabolic function is established in females. However, its necessity in males, while appreciated, requires further study. Accordingly, we first determined whether lower metabolic function in male mice compared with females is related to reduced ERα expression. ERα protein expression in metabolically active tissues was lower in males than in females, and this lower expression was associated with worse glucose tolerance. Second, we determined whether ERα is required for optimal immunometabolic function in male mice consuming a chow diet. Despite lower expression of ERα in males, its genetic ablation (KO) caused an insulin-resistant phenotype characterized by enhanced adiposity, glucose intolerance, hepatic steatosis, and metaflammation in adipose tissue and liver. Last, we determined whether ERα is essential for exercise-induced metabolic adaptations. Twelve-week-old wild-type (WT) and ERα KO mice either remained sedentary (SED) or were given access to running wheels (WR) for 10 wk while fed an obesogenic diet. Body weight and fat mass were lower in WR mice regardless of genotype. Daily exercise obliterated immune cell infiltration and inflammatory gene transcripts in adipose tissue in both genotypes. In the liver, however, wheel running suppressed hepatic steatosis and inflammatory gene transcripts in WT but not in KO mice. In conclusion, the present findings indicate that ERα is required for optimal immunometabolic function in male mice despite their reduced ERα protein expression in metabolically active tissues. Furthermore, for the first time, we show that ERα signaling appears to be obligatory for exercise-induced prevention of hepatic steatosis.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Thomas J Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Zachary I Grunewald
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Rory P Cunningham
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Makenzie L Woodford
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
| | - Dennis B Lubahn
- Department of Biochemistry, University of Missouri , Columbia, Missouri
| | - Camila Manrique-Acevedo
- Department of Medicine, Division of Endocrinology, University of Missouri , Columbia, Missouri
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri , Columbia, Missouri
- Research Service, Harry S. Truman Memorial Hospital, University of Missouri , Columbia, Missouri
| | | | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri , Columbia, Missouri
- Department of Child Health, University of Missouri , Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri
| |
Collapse
|