1
|
Ferreira-Santos L, Ramirez-Perez FI, Foote CA, Augenreich MA, McMillan NJ, Williams MB, Gonzalez-Vallejo JD, Power G, Wheeler AA, Manrique-Acevedo C, Martinez-Lemus LA, Padilla J. Neuraminidase-induced externalization of phosphatidylserine activates ADAM17 and impairs insulin signaling in endothelial cells. Am J Physiol Heart Circ Physiol 2024; 326:H270-H277. [PMID: 37999645 PMCID: PMC11219045 DOI: 10.1152/ajpheart.00638.2023] [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: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023]
Abstract
Endothelial insulin resistance represents a causal factor in the pathogenesis of type 2 diabetes (T2D) and vascular disease, thus the need to identify molecular mechanisms underlying defects in endothelial insulin signaling. We previously have shown that a disintegrin and metalloproteinase-17 (ADAM17) is increased while insulin receptor α-subunit (IRα) is decreased in the vasculature of patients with T2D, leading to impaired insulin-induced vasodilation. We have also demonstrated that ADAM17 sheddase activity targets IRα; however, the mechanisms driving endothelial ADAM17 activity in T2D are largely unknown. Herein, we report that externalization of phosphatidylserine (PS) to the outer leaflet of the plasma membrane causes ADAM17-mediated shedding of IRα and blunting of insulin signaling in endothelial cells. Furthermore, we demonstrate that endothelial PS externalization is mediated by the phospholipid scramblase anoctamin-6 (ANO6) and that this process can be stimulated by neuraminidase, a soluble enzyme that cleaves sialic acid residues. Of note, we demonstrate that men and women with T2D display increased levels of neuraminidase activity in plasma, relative to age-matched healthy individuals, and this occurs in conjunction with increased ADAM17 activity and impaired leg blood flow responses to endogenous insulin. Collectively, this work reveals the neuraminidase-ANO6-ADAM17 axis as a novel potential target for restoring endothelial insulin sensitivity in T2D.NEW & NOTEWORTHY This work provides the first evidence that neuraminidase, an enzyme increased in the circulation of men and women with type 2 diabetes (T2D), promotes anoctamin-6 (ANO6)-dependent externalization of phosphatidylserine in endothelial cells, which in turn leads to activation of a disintegrin and metalloproteinase-17 (ADAM17) and consequent shedding of the insulin receptor-α from the cell surface. Hence, this work supports that consideration should be given to the neuraminidase-ANO6-ADAM17 axis as a novel potential target for restoring endothelial insulin sensitivity in T2D.
Collapse
Affiliation(s)
| | | | - Christopher A Foote
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
| | - Marc A Augenreich
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Neil J McMillan
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Morgan B Williams
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | | | - Gavin Power
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Andrew A Wheeler
- Department of Surgery, University of Missouri, Columbia, Missouri, United States
| | - Camila Manrique-Acevedo
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| |
Collapse
|
2
|
Foote CA, Ramirez-Perez FI, Smith JA, Ghiarone T, Morales-Quinones M, McMillan NJ, Augenreich MA, Power G, Burr K, Aroor AR, Bender SB, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. Neuraminidase inhibition improves endothelial function in diabetic mice. Am J Physiol Heart Circ Physiol 2023; 325:H1337-H1353. [PMID: 37801046 PMCID: PMC10908409 DOI: 10.1152/ajpheart.00337.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023]
Abstract
Neuraminidases cleave sialic acids from glycocalyx structures and plasma neuraminidase activity is elevated in type 2 diabetes (T2D). Therefore, we hypothesize circulating neuraminidase degrades the endothelial glycocalyx and diminishes flow-mediated dilation (FMD), whereas its inhibition restores shear mechanosensation and endothelial function in T2D settings. We found that compared with controls, subjects with T2D have higher plasma neuraminidase activity, reduced plasma nitrite concentrations, and diminished FMD. Ex vivo and in vivo neuraminidase exposure diminished FMD and reduced endothelial glycocalyx presence in mouse arteries. In cultured endothelial cells, neuraminidase reduced glycocalyx coverage. Inhalation of the neuraminidase inhibitor, zanamivir, reduced plasma neuraminidase activity, enhanced endothelial glycocalyx length, and improved FMD in diabetic mice. In humans, a single-arm trial (NCT04867707) of zanamivir inhalation did not reduce plasma neuraminidase activity, improved glycocalyx length, or enhanced FMD. Although zanamivir plasma concentrations in mice reached 225.8 ± 22.0 ng/mL, in humans were only 40.0 ± 7.2 ng/mL. These results highlight the potential of neuraminidase inhibition for ameliorating endothelial dysfunction in T2D and suggest the current Food and Drug Administration-approved inhaled dosage of zanamivir is insufficient to achieve desired outcomes in humans.NEW & NOTEWORTHY This work identifies neuraminidase as a key mediator of endothelial dysfunction in type 2 diabetes that may serve as a biomarker for impaired endothelial function and predictive of development and progression of cardiovascular pathologies associated with type 2 diabetes (T2D). Data show that intervention with the neuraminidase inhibitor zanamivir at effective plasma concentrations may represent a novel pharmacological strategy for restoring the glycocalyx and ameliorating endothelial dysfunction.
Collapse
Affiliation(s)
- Christopher A Foote
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
| | | | - James A Smith
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Thaysa Ghiarone
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | | | - Neil J McMillan
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Marc A Augenreich
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Gavin Power
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Katherine Burr
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | - Annayya R Aroor
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
| | - Shawn B Bender
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Camila Manrique-Acevedo
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
3
|
Zheng X, Li Z, Berg Sen J, Samarah L, Deacon CS, Bernardo J, Machin DR. Western diet augments metabolic and arterial dysfunction in a sex-specific manner in outbred, genetically diverse mice. Front Nutr 2023; 9:1090023. [PMID: 36687716 PMCID: PMC9853899 DOI: 10.3389/fnut.2022.1090023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/23/2022] [Indexed: 01/07/2023] Open
Abstract
Western diet (WD), characterized by excess saturated fat and sugar intake, is a major contributor to obesity and metabolic and arterial dysfunction in humans. However, these phenotypes are not consistently observed in traditional inbred, genetically identical mice. Therefore, we sought to determine the effects of WD on visceral adiposity and metabolic/arterial function in UM-HET3 mice, an outbred, genetically diverse strain of mice. Male and female UM-HET3 mice underwent normal chow (NC) or WD for 12 weeks. Body mass and visceral adiposity were higher in WD compared to NC (P < 0.05). Female WD mice had greater visceral adiposity than male WD mice (P < 0.05). The results of glucose and insulin tolerance tests demonstrated that metabolic function was lower in WD compared to NC mice (P < 0.05). Metabolic dysfunction in WD as was driven by male mice, as metabolic function in female WD mice was unchanged (P > 0.05). Systolic blood pressure (BP) and aortic stiffness were increased in WD after 2 weeks compared to baseline and continued to increase through week 12 (P < 0.05). Systolic BP and aortic stiffness were higher from weeks 2-12 in WD compared to NC (P < 0.05). Aortic collagen content was higher in WD compared to NC (P < 0.05). Carotid artery endothelium-dependent dilation was lower in WD compared to NC (P < 0.05). These data suggest sex-related differences in visceral adiposity and metabolic dysfunction in response to WD. Despite this, arterial dysfunction was similar in male and female WD mice, indicating this model may provide unique translational insight into similar sex-related observations in humans that consume WD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniel R. Machin
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
4
|
Foote CA, Soares RN, Ramirez-Perez FI, Ghiarone T, Aroor A, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. Endothelial Glycocalyx. Compr Physiol 2022; 12:3781-3811. [PMID: 35997082 PMCID: PMC10214841 DOI: 10.1002/cphy.c210029] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The glycocalyx is a polysaccharide structure that protrudes from the body of a cell. It is primarily conformed of glycoproteins and proteoglycans, which provide communication, electrostatic charge, ionic buffering, permeability, and mechanosensation-mechanotransduction capabilities to cells. In blood vessels, the endothelial glycocalyx that projects into the vascular lumen separates the vascular wall from the circulating blood. Such a physical location allows a number of its components, including sialic acid, glypican-1, heparan sulfate, and hyaluronan, to participate in the mechanosensation-mechanotransduction of blood flow-dependent shear stress, which results in the synthesis of nitric oxide and flow-mediated vasodilation. The endothelial glycocalyx also participates in the regulation of vascular permeability and the modulation of inflammatory responses, including the processes of leukocyte rolling and extravasation. Its structural architecture and negative charge work to prevent macromolecules greater than approximately 70 kDa and cationic molecules from binding and flowing out of the vasculature. This also prevents the extravasation of pathogens such as bacteria and virus, as well as that of tumor cells. Due to its constant exposure to shear and circulating enzymes such as neuraminidase, heparanase, hyaluronidase, and matrix metalloproteinases, the endothelial glycocalyx is in a continuous process of degradation and renovation. A balance favoring degradation is associated with a variety of pathologies including atherosclerosis, hypertension, vascular aging, metastatic cancer, and diabetic vasculopathies. Consequently, ongoing research efforts are focused on deciphering the mechanisms that promote glycocalyx degradation or limit its syntheses, as well as on therapeutic approaches to improve glycocalyx integrity with the goal of reducing vascular disease. © 2022 American Physiological Society. Compr Physiol 12: 1-31, 2022.
Collapse
Affiliation(s)
- Christopher A. Foote
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Rogerio N. Soares
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | | | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Annayya Aroor
- Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Luis A. Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
5
|
The Gut Microbiota and Vascular Aging: A State-of-the-Art and Systematic Review of the Literature. J Clin Med 2022; 11:jcm11123557. [PMID: 35743626 PMCID: PMC9224769 DOI: 10.3390/jcm11123557] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota is a critical regulator of human physiology, deleterious changes to its composition and function (dysbiosis) have been linked to the development and progression of cardiovascular diseases. Vascular ageing (VA) is a process of progressive stiffening of the arterial tree associated with arterial wall remodeling, which can precede hypertension and organ damage, and is associated with cardiovascular risk. Arterial stiffness has become the preferred marker of VA. In our systematic review, we found an association between gut microbiota composition and arterial stiffness, with two patterns, in most animal and human studies: a direct correlation between arterial stiffness and abundances of bacteria associated with altered gut permeability and inflammation; an inverse relationship between arterial stiffness, microbiota diversity, and abundances of bacteria associated with most fit microbiota composition. Interventional studies were able to show a stable link between microbiota modification and arterial stiffness only in animals. None of the human interventional trials was able to demonstrate this relationship, and very few adjusted the analyses for determinants of arterial stiffness. We observed a lack of large randomized interventional trials in humans that test the role of gut microbiota modifications on arterial stiffness, and take into account BP and hemodynamic alterations.
Collapse
|
6
|
Castorena-Gonzalez JA. Lymphatic Valve Dysfunction in Western Diet-Fed Mice: New Insights Into Obesity-Induced Lymphedema. Front Pharmacol 2022; 13:823266. [PMID: 35308249 PMCID: PMC8931217 DOI: 10.3389/fphar.2022.823266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
A two-way connection between obesity and lymphatic dysfunction has now been established. Clinical studies have demonstrated that obesity significantly increases the risk for developing secondary lymphedema. Using animal-models, obesity and metabolic syndrome have been linked to different aspects of lymphatic structural abnormalities and lymphatic dysfunction, including impaired contractility, impaired flow-mediated responses, impaired fluid transport, as well as increased permeability, and abnormal dendritic cell migration among others. Dysfunction of lymphatic valves is a main form of lymphatic dysfunction, known to result in severe edematous phenotypes; however, the extent of lymphatic valve deficiency in secondary lymphedema, including obesity-induced lymphedema, remains unknown. Therefore, the aims of the present study were 1) to determine whether western diet-induced obesity results in lymphatic valve dysfunction, and 2) to determine whether lymphatic valve dysfunction in western diet-induced obesity results from the diet itself, or as a consequence of the metabolic alterations induced by the diet. First, we quantitatively assessed and compared valve function in isolated popliteal and mesenteric collecting lymphatic vessels from control and western diet-induced obese C57BL/6J (WT) mice. Feeding a western diet for 14 weeks induced obesity and elevated plasma glucose and cholesterol levels when compared to controls. The function of lymphatic valves in popliteal lymphatics was not affected by diet-induced obesity; however, significant back-leak of pressure was observed in mesenteric lymphatic valves. Dysfunctional, leaky valves from obese animals also required significantly higher adverse pressure to trigger valve closure. Importantly, when subjected to treatment with a western diet, globally deficient PAI-1 mice were significantly protected against metabolic dysfunction and displayed fully functional, competent mesenteric lymphatic valves. In conclusion, our findings show for the first time that, in association with the metabolic alterations induced by the western diet, lymphatic valve dysfunction can be a critical component of obesity-induced lymphedema.
Collapse
|
7
|
Ramirez-Perez FI, Cabral-Amador FJ, Whaley-Connell AT, Aroor AR, Morales-Quinones M, Woodford ML, Ghiarone T, Ferreira-Santos L, Jurrissen TJ, Manrique-Acevedo CM, Jia G, DeMarco VG, Padilla J, Martinez-Lemus LA, Lastra G. Cystamine reduces vascular stiffness in Western diet-fed female mice. Am J Physiol Heart Circ Physiol 2022; 322:H167-H180. [PMID: 34890280 PMCID: PMC8742720 DOI: 10.1152/ajpheart.00431.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Consumption of diets high in fat, sugar, and salt (Western diet, WD) is associated with accelerated arterial stiffening, a major independent risk factor for cardiovascular disease (CVD). Women with obesity are more prone to develop arterial stiffening leading to more frequent and severe CVD compared with men. As tissue transglutaminase (TG2) has been implicated in vascular stiffening, our goal herein was to determine the efficacy of cystamine, a nonspecific TG2 inhibitor, at reducing vascular stiffness in female mice chronically fed a WD. Three experimental groups of female mice were created. One was fed regular chow diet (CD) for 43 wk starting at 4 wk of age. The second was fed a WD for the same 43 wk, whereas a third cohort was fed WD, but also received cystamine (216 mg/kg/day) in the drinking water during the last 8 wk on the diet (WD + C). All vascular stiffness parameters assessed, including aortic pulse wave velocity and the incremental modulus of elasticity of isolated femoral and mesenteric arteries, were significantly increased in WD- versus CD-fed mice, and reduced in WD + C versus WD-fed mice. These changes coincided with respectively augmented and diminished vascular wall collagen and F-actin content, with no associated effect in blood pressure. In cultured human vascular smooth muscle cells, cystamine reduced TG2 activity, F-actin:G-actin ratio, collagen compaction capacity, and cellular stiffness. We conclude that cystamine treatment represents an effective approach to reduce vascular stiffness in female mice in the setting of WD consumption, likely because of its TG2 inhibitory capacity.NEW & NOTEWORTHY This study evaluates the novel role of transglutaminase 2 (TG2) inhibition to directly treat vascular stiffness. Our data demonstrate that cystamine, a nonspecific TG2 inhibitor, improves vascular stiffness induced by a diet rich in fat, fructose, and salt. This research suggests that TG2 inhibition might bear therapeutic potential to reduce the disproportionate burden of cardiovascular disease in females in conditions of chronic overnutrition.
Collapse
Affiliation(s)
- Francisco I. Ramirez-Perez
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Biomedical, Biological, and Chemical Engineering Department, University of Missouri, Columbia, Missouri
| | | | - Adam T. Whaley-Connell
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,4Division of Nephrology and Hypertension, Department of Medicine, University of Missouri, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | - Annayya R. Aroor
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | | | - Makenzie L. Woodford
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Thaysa Ghiarone
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Larissa Ferreira-Santos
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,6Instituto do Coracao, Hospital das Clínicas da Faculdade de
Medicina da Universidade de São Paulo, Faculdade de Medicina, Universidade
de São Paulo, São Paulo, Brazil
| | - Thomas J. Jurrissen
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,7Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Camila M. Manrique-Acevedo
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | - GuangHong Jia
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| | - Vincent G. DeMarco
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,4Division of Nephrology and Hypertension, Department of Medicine, University of Missouri, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri,8Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,7Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Luis A. Martinez-Lemus
- 1Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Biomedical, Biological, and Chemical Engineering Department, University of Missouri, Columbia, Missouri,8Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Guido Lastra
- 3Research Service, Harry S. Truman Memorial
Veterans’ Hospital, Columbia, Missouri,5Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
8
|
González-Blázquez R, Alcalá M, Cárdenas-Rebollo JM, Viana M, Steckelings UM, Boisvert WA, Unger T, Fernández-Alfonso MS, Somoza B, Gil-Ortega M. AT2R stimulation with C21 prevents arterial stiffening and endothelial dysfunction in the abdominal aorta from mice fed a high-fat diet. Clin Sci (Lond) 2021; 135:2763-2780. [PMID: 34854902 DOI: 10.1042/cs20210971] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to evaluate the effect of Compound 21 (C21), a selective AT2R agonist, on the prevention of endothelial dysfunction, extracellular matrix (ECM) remodeling and arterial stiffness associated with diet-induced obesity (DIO). Five-week-old male C57BL/6J mice were fed a standard (Chow) or high-fat diet (HF) for 6 weeks. Half of the animals of each group were simultaneously treated with C21 (1 mg/kg/day, in the drinking water), generating four groups: Chow C, Chow C21, HF C, and HF C21. Vascular function and mechanical properties were determined in the abdominal aorta. To evaluate ECM remodeling, collagen deposition and TGF-β1 concentrations were determined in the abdominal aorta and the activity of metalloproteinases (MMP) 2 and 9 was analyzed in the plasma. Abdominal aortas from HF C mice showed endothelial dysfunction as well as enhanced contractile but reduced relaxant responses to Ang II. This effect was abrogated with C21 treatment by preserving NO availability. A left-shift in the tension-stretch relationship, paralleled by an augmented β-index (marker of intrinsic arterial stiffness), and enhanced collagen deposition and MMP-2/-9 activities were also detected in HF mice. However, when treated with C21, HF mice exhibited lower TGF-β1 levels in abdominal aortas together with reduced MMP activities and collagen deposition compared with HF C mice. In conclusion, these data demonstrate that AT2R stimulation by C21 in obesity preserves NO availability and prevents unhealthy vascular remodeling, thus protecting the abdominal aorta in HF mice against the development of endothelial dysfunction, ECM remodeling and arterial stiffness.
Collapse
Affiliation(s)
- Raquel González-Blázquez
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Martín Alcalá
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, 28925, Madrid, Spain
| | - José Miguel Cárdenas-Rebollo
- Departamento de Matemática Aplicada y Estadística. Facultad de Ciencias Económicas y Empresariales. Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Marta Viana
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, 28925, Madrid, Spain
| | - Ulrike Muscha Steckelings
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, BSB311, Honolulu, HI 96813, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Str., Kazan 420008, Russia
| | - Thomas Unger
- CARIM - School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - María S Fernández-Alfonso
- Instituto Pluridisciplinar, Unidad de Cartografía Cerebral, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| |
Collapse
|
9
|
Boateng SY, Olfert IM, Chantler PD. Role of Perivascular Adipose Tissue and Exercise on Arterial Function with Obesity. Exerc Sport Sci Rev 2021; 49:188-196. [PMID: 33831902 PMCID: PMC8195847 DOI: 10.1249/jes.0000000000000251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Adipose tissue and arterial dysfunction are common in the obese state. Perivascular adipose tissue (PVAT) plays an important role in mediating arterial health, and with obesity, the PVAT dysfunction negatively affects arterial health. Exercise training exerts direct and beneficial effects on PVAT, providing an additional and novel pathway by which exercise can improve arterial health in diseased populations.
Collapse
Affiliation(s)
- Samuel Y Boateng
- Biological Sciences, School of Biological Sciences, University of Reading, UK
| | - I. Mark Olfert
- Department of Human Performance, Division of Exercise Physiology, School of Medicine, West Virginia University, USA
- West Virginia Clinical and Translational Science Institute (WVCTSI), Morgantown, WV
| | - Paul D Chantler
- Department of Human Performance, Division of Exercise Physiology, School of Medicine, West Virginia University, USA
- West Virginia Clinical and Translational Science Institute (WVCTSI), Morgantown, WV
| |
Collapse
|
10
|
Ogola BO, Clark GL, Abshire CM, Harris NR, Gentry KL, Gunda SS, Kilanowski-Doroh I, Wong TJ, Visniauskas B, Lawrence DJ, Zimmerman MA, Bayer CL, Groban L, Miller KS, Lindsey SH. Sex and the G Protein-Coupled Estrogen Receptor Impact Vascular Stiffness. Hypertension 2021; 78:e1-e14. [PMID: 34024124 PMCID: PMC8192475 DOI: 10.1161/hypertensionaha.120.16915] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Benard O. Ogola
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | - Gabrielle L. Clark
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | - Caleb M. Abshire
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | | | - Kaylee L. Gentry
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | - Shreya S. Gunda
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | | | - Tristen J. Wong
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| | | | - Dylan J. Lawrence
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | | | - Carolyn L. Bayer
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kristin S. Miller
- Tulane University, Department of Biomedical Engineering, New Orleans, LA, USA
| | - Sarah H. Lindsey
- Tulane University, Department of Pharmacology, New Orleans, LA, USA
| |
Collapse
|
11
|
Wijesekara P, Liu Y, Wang W, Johnston EK, Sullivan MLG, Taylor RE, Ren X. Accessing and Assessing the Cell-Surface Glycocalyx Using DNA Origami. NANO LETTERS 2021; 21:4765-4773. [PMID: 34030445 PMCID: PMC8193633 DOI: 10.1021/acs.nanolett.1c01236] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Indexed: 05/30/2023]
Abstract
The cell-surface glycocalyx serves as a physiological barrier regulating cellular accessibility to macromolecules and other cells. Conventional glycocalyx characterization has largely been morphological rather than functional. Here, we demonstrated direct glycocalyx anchoring of DNA origami nanotiles and performed a comprehensive comparison with traditional origami targeting to the phospholipid bilayer (PLB) using cholesterol. While DNA nanotiles effectively accessed single-stranded DNA initiators anchored on the glycocalyx, their accessibility to the underlying PLB was only permitted by extended nanotile-to-initiator spacing or by enzymatic glycocalyx degradation using trypsin or pathogenic neuraminidase. Thus, the DNA nanotiles, being expelled by the physiologic glycocalyx, provide an effective functional measure of the glycocalyx barrier integrity and faithfully predict cell-to-cell accessibility during DNA-guided multicellular assembly. Lastly, the glycocalyx-anchoring mechanism enabled enhanced cell-surface stability and cellular uptake of nanotiles compared to PLB anchoring. This research lays the foundation for future development of DNA nanodevices to access the cell surface.
Collapse
Affiliation(s)
- Piyumi Wijesekara
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Ying Liu
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Weitao Wang
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Elizabeth K. Johnston
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Mara L. G. Sullivan
- Center
for Biologic Imaging, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, Pennsylvania, United States
| | - Rebecca E. Taylor
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
- Department
of Electrical and Computer Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| | - Xi Ren
- Department
of Biomedical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
- Department
of Mechanical Engineering, Carnegie Mellon
University, 5000 Forbes Avenue, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
12
|
Clayton ZS, Brunt VE, Hutton DA, Casso AG, Ziemba BP, Melov S, Campisi J, Seals DR. Tumor Necrosis Factor Alpha-Mediated Inflammation and Remodeling of the Extracellular Matrix Underlies Aortic Stiffening Induced by the Common Chemotherapeutic Agent Doxorubicin. Hypertension 2021; 77:1581-1590. [PMID: 33719511 DOI: 10.1161/hypertensionaha.120.16759] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - David A Hutton
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Abigail G Casso
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Brian P Ziemba
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA (S.M., J.C.)
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA (S.M., J.C.).,Lawrence Berkeley National Laboratory, CA (J.C.)
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| |
Collapse
|
13
|
Toyama K, Igase M, Spin JM, Abe Y, Javkhlant A, Okada Y, Wagenhäuser MU, Schelzig H, Tsao PS, Mogi M. Exosome miR-501-3p Elevation Contributes to Progression of Vascular Stiffness. Circ Rep 2021; 3:170-177. [PMID: 33738350 PMCID: PMC7956882 DOI: 10.1253/circrep.cr-20-0135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Tight junction (TJ) disruption and dysfunction are involved in the progression of arteriosclerosis. miR-501-3p regulates endothelial TJ protein-1, resulting in TJ disruption. Because exosomal microRNAs can travel to distant tissues and influence cell behavior, patients with elevated miR-501-3p may experience accelerated vascular disease progression secondary to miR-501-3p-induced reductions in TJ. This study investigated whether plasma exosome miR-501-3p levels are associated with vascular stiffness, an indicator for arteriosclerotic changes. Methods and Results: Fifty-one subjects (mean [±SD] age 70±8 years, 37% male) enrolled in a medical checkup program were recruited to the study. Brachial-ankle arterial pulse wave velocity (baPWV) and plasma exosome miR-501-3p expression were measured. Patients were divided into 2 groups depending on whether their miR-501-3p ∆Ct values were above ("High"; n=24) or below ("Low"; n=27) the cut-off levels determined by receiver operating characteristic (ROC) curve analysis. Median (interquartile range) baPWV levels were significantly higher in the miR-501-3p High than Low group (1,664 [1,496-1,859] vs. 1,450 [1,353-1,686] cm/s, respectively; P<0.05). Multivariate logistic regression analysis showed a significant association between increased baPWV and High miR-501-3p expression (odds ratio 4.66). At follow-up visits (mean 62 months later), baPWV remained significantly higher in the miR-501-3p High than Low group (1,830 [1,624-2,056] vs. 1,620 [1,377-1,816] cm/s, respectively; P<0.05). Conclusions: High expression levels of exosome miR-501-3p contribute to arteriosclerotic changes.
Collapse
Affiliation(s)
- Kensuke Toyama
- Department of Pharmacology, Ehime University Graduate School of Medicine Toon Japan
| | - Michiya Igase
- Department of Anti-aging Medicine, Ehime University Graduate School of Medicine Toon Japan
| | - Joshua M Spin
- VA Palo Alto Health Care System Palo Alto, CA USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine Stanford, CA USA
| | - Yasunori Abe
- Department of Pharmacology, Ehime University Graduate School of Medicine Toon Japan
| | - Amarsanaa Javkhlant
- Department of Pharmacology, Ehime University Graduate School of Medicine Toon Japan
| | - Yoko Okada
- Department of Anti-aging Medicine, Ehime University Graduate School of Medicine Toon Japan
| | - Markus U Wagenhäuser
- Department of Vascular and Endovascular Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf Germany
| | - Hubert Schelzig
- Department of Vascular and Endovascular Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf Germany
| | - Philip S Tsao
- VA Palo Alto Health Care System Palo Alto, CA USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine Stanford, CA USA
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine Toon Japan
| |
Collapse
|
14
|
Dunn S, Hilgers RH, Das KC. Thioredoxin deficiency exacerbates vascular dysfunction during diet-induced obesity in small mesenteric artery in mice. Microcirculation 2020; 28:e12674. [PMID: 33316843 DOI: 10.1111/micc.12674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/07/2020] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Thioredoxin (Trx) is a small cellular redox protein with established antioxidant and disulfide reductase properties. We hypothesized that Trx deficiency in mice would cause increased oxidative stress with consequent redox imbalance that would exacerbate obesity-induced vascular dysfunction. METHODS Non-transgenic (NT, C57BL/6) and dominant-negative Trx (dnTrx-Tg, low levels of redox-active protein) mice were either fed a normal diet (NC) or high fat diet plus sucrose (HFS) diet for 4 months (3-month HFD+ 1-month HFS). Weight gain, glucose tolerance test (GTT), insulin tolerance test (ITT), and other metabolic parameters were performed following NC or HFS diet. Arterial structural remodeling and functional parameters were assessed by myography. RESULTS Our study found that dnTrx mice with lower levels of active Trx exacerbated myogenic tone, inward arterial remodeling, arterial stiffening, phenylephrine-induced contraction, and endothelial dysfunction of MA. Additionally, FeTMPyP, a peroxynitrite decomposition catalyst, acutely decreased myogenic tone and contraction and normalized endothelial function in MA from dnTrx-Tg mice on HFS via increasing nitric oxide (NO)-mediated relaxation. CONCLUSIONS Our results indicate that deficiency of active Trx exacerbates MA contractile and relaxing properties during diet-induced obesity demonstrating that loss of redox balance in obesity is a key mechanism of vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Shannon Dunn
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Robert H Hilgers
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kumuda C Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
15
|
Li Z, Wu N, Wang J, Zhang Q. Roles of Endovascular Calyx Related Enzymes in Endothelial Dysfunction and Diabetic Vascular Complications. Front Pharmacol 2020; 11:590614. [PMID: 33328998 PMCID: PMC7734331 DOI: 10.3389/fphar.2020.590614] [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: 08/02/2020] [Accepted: 10/16/2020] [Indexed: 12/25/2022] Open
Abstract
In recent years, the number of diabetic patients has rapidly increased. Diabetic vascular complications seriously affect people’s quality of life. Studies found that endothelial dysfunction precedes the vascular complications of diabetes. Endothelial dysfunction is related to glycocalyx degradation on the surface of blood vessels. Heparanase (HPSE), matrix metalloproteinase (MMP), hyaluronidase (HYAL), hyaluronic acid synthase (HAS), and neuraminidase (NEU) are related to glycocalyx degradation. Therefore, we reviewed the relationship between endothelial dysfunction and the vascular complications of diabetes from the perspective of enzymes.
Collapse
Affiliation(s)
- Zhi Li
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ning Wu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Wang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Quanbin Zhang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Castorena-Gonzalez JA, Srinivasan RS, King PD, Simon AM, Davis MJ. Simplified method to quantify valve back-leak uncovers severe mesenteric lymphatic valve dysfunction in mice deficient in connexins 43 and 37. J Physiol 2020; 598:2297-2310. [PMID: 32267537 PMCID: PMC8170716 DOI: 10.1113/jp279472] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Lymphatic valve defects are one of the major causes of lymph transport dysfunction; however, there are no accessible methods for quantitatively assessing valve function. This report describes a novel technique for quantifying lymphatic valve back-leak. Postnatal endothelial-specific deletion of connexin 43 (Cx43) in connexin 37 null (Cx37-/- ) mice results in rapid regression of valve leaflets and severe valve dysfunction. This method can also be used for assessing the function of venous and lymphatic valves from various species, including humans. ABSTRACT The lymphatic system relies on robust, spontaneous contractions of collecting lymphatic vessels and one-way secondary lymphatic valves to efficiently move lymph forward. Secondary valves prevent reflux and allow for the generation of propulsive pressure during each contraction cycle. Lymphatic valve defects are one of the major causes of lymph transport dysfunction. Genetic mutations in multiple genes have been associated with the development of primary lymphoedema in humans; and many of the same mutations in mice result in valve defects that subsequently lead to chylous ascites or chylothorax. At present the only experimental technique for the quantitative assessment of lymphatic valve function utilizes the servo-null micropressure system, which is highly accurate and precise, but relatively inaccessible and difficult to use. We developed a novel, simplified alternative method for quantifying valve function and determining the degree of pressure back-leak through an intact valve in pressurized, single-valve segments of isolated lymphatic vessels. With this diameter-based method, the competence of each lymphatic valve is challenged over a physiological range of pressures (e.g. 0.5-10cmH2 O) and pressure back-leak is extrapolated from calibrated, pressure-driven changes in diameter upstream from the valve. Using mesenteric lymphatic vessels from C57BL/6J, Ub-CreERT2 ;Rasa1fx/fx , Foxc2Cre/+ , Lyve1-Cre;Cx43fx/fx , and Prox1-CreERT2 ;Cx43fx/fx ;Cx37-/- mice, we tested our method on lymphatic valves displaying a wide range of dysfunction, from fully competent to completely incompetent. Our results were validated by simultaneous direct measurement of pressure back-leak using a servo-null micropressure system. Our diameter-based technique can be used to quantify valve function in isolated lymphatic valves from a variety of species. This method also revealed that haplodeficiency in Foxc2 (Foxc2Cre/+ ) is not sufficient to cause significant valve dysfunction; however, postnatal endothelial-specific deletion of Cx43 in Cx37-/- mice results in rapid regression of valve leaflets and severe valve dysfunction.
Collapse
Affiliation(s)
- Jorge A Castorena-Gonzalez
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Michael J Davis
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
17
|
Jelinic M, Kahlberg N, Leo CH, Ng HH, Rosli S, Deo M, Li M, Finlayson S, Walsh J, Parry LJ, Ritchie RH, Qin CX. Annexin-A1 deficiency exacerbates pathological remodelling of the mesenteric vasculature in insulin-resistant, but not insulin-deficient, mice. Br J Pharmacol 2020; 177:1677-1691. [PMID: 31724161 DOI: 10.1111/bph.14927] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/04/2019] [Accepted: 10/27/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Arterial stiffness, a characteristic feature of diabetes, increases the risk of cardiovascular complications. Potential mechanisms that promote arterial stiffness in diabetes include oxidative stress, glycation and inflammation. The anti-inflammatory protein annexin-A1 has cardioprotective properties, particularly in the context of ischaemia. However, the role of endogenous annexin-A1 in the vasculature in both normal physiology and pathophysiology remains largely unknown. Hence, this study investigated the role of endogenous annexin-A1 in diabetes-induced remodelling of mouse mesenteric vasculature. EXPERIMENTAL APPROACH Insulin-resistance was induced in male mice (AnxA1+/+ and AnxA1-/- ) with the combination of streptozotocin (55mg/kg i.p. x 3 days) with high fat diet (42% energy from fat) or citrate vehicle with normal chow diet (20-weeks). Insulin-deficiency was induced in a separate cohort of mice using a higher total streptozocin dose (55mg/kg i.p. x 5 days) on chow diet (16-weeks). At study endpoint, mesenteric artery passive mechanics were assessed by pressure myography. KEY RESULTS Insulin-resistance induced significant outward remodelling but had no impact on passive stiffness. Interestingly, vascular stiffness was significantly increased in AnxA1-/- mice when subjected to insulin-resistance. In contrast, insulin-deficiency induced outward remodelling and increased volume compliance in mesenteric arteries, regardless of genotype. In addition, the annexin-A1 / formyl peptide receptor axis is upregulated in both insulin-resistant and insulin-deficient mice. CONCLUSION AND IMPLICATIONS Our study provided the first evidence that endogenous AnxA1 may play an important vasoprotective role in the context of insulin-resistance. AnxA1-based therapies may provide additional benefits over traditional anti-inflammatory strategies for reducing vascular injury in diabetes.
Collapse
Affiliation(s)
- Maria Jelinic
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Nicola Kahlberg
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia
| | - Chen Huei Leo
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia.,Science, Math and Technology, Singapore University of Technology and Design, Singapore
| | - Hooi Hooi Ng
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia.,Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Sarah Rosli
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Minh Deo
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mandy Li
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Siobhan Finlayson
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jesse Walsh
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Laura J Parry
- School of BioSciences, University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Cheng Xue Qin
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
18
|
McCallinhart PE, Biwer LA, Clark OE, Isakson BE, Lilly B, Trask AJ. Myoendothelial Junctions of Mature Coronary Vessels Express Notch Signaling Proteins. Front Physiol 2020; 11:29. [PMID: 32116749 PMCID: PMC7010921 DOI: 10.3389/fphys.2020.00029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
RATIONALE Myoendothelial junctions (MEJs) within the fenestrae of the internal elastic lamina (IEL) are critical sites that allow for endothelial cell (EC) - vascular smooth muscle cell (VSMC) contact and communication. Vascular Notch signaling is a critical determinant of normal vasculogenesis and remodeling, and it regulates cell phenotype via contact between ECs and VSMCs. To date, no studies have linked Notch signaling to the MEJ despite it requiring cell-cell contact. Furthermore, very little is known about Notch in the adult coronary circulation or the localization of Notch signaling and activity within the mature intact blood vessel. OBJECTIVE We tested the hypothesis that vascular Notch signaling between ECs and VSMCs occurs at MEJs. METHODS AND RESULTS Notch receptor and ligand immunofluorescence was performed in human coronary EC and VSMC co-cultures across transwell inserts (in vitro MEJs) and in the intact mouse coronary circulation. Human coronary VSMC Notch activity induced by human coronary ECs at the in vitro MEJ was assessed using a CBF-luciferase construct. We observed Jagged1, Notch1, Notch2, and Notch3 expression within the in vitro and in vivo MEJs. We also demonstrated a 3-fold induction (p < 0.001) of human coronary VSMC Notch signaling by ECs at the in vitro MEJ, which was completely blocked by the Notch inhibitor, DAPT (p < 0.01). CONCLUSION We demonstrate for the first time in mature blood vessels that Notch receptors and ligands are expressed within and are active at coronary MEJs, demonstrating a previously unrecognized mode of Notch signaling regulation between the endothelium and smooth muscle.
Collapse
Affiliation(s)
- Patricia E. McCallinhart
- Center for Cardiovascular Research, The Heart Center, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Lauren A. Biwer
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Olivia E. Clark
- Center for Cardiovascular Research, The Heart Center, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Brenda Lilly
- Center for Cardiovascular Research, The Heart Center, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Aaron J. Trask
- Center for Cardiovascular Research, The Heart Center, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
19
|
Metabolomics Elucidates Dose-Dependent Molecular Beneficial Effects of Hesperidin Supplementation in Rats Fed an Obesogenic Diet. Antioxidants (Basel) 2020; 9:antiox9010079. [PMID: 31963315 PMCID: PMC7023145 DOI: 10.3390/antiox9010079] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome (MetS) is a global epidemic concern. Polyphenols are proposed as good candidates for its prevention, although their mechanisms are not fully understood. The gut microbiota seems to play a key role in polyphenol beneficial effects. Here, we assessed the effects of the citrus polyphenol hesperidin combining an untargeted metabolomics approach, which has an inherent potential to elucidate the host-microbiome interplay, with extensive anthropometric and biochemical characterizations and integrating metabolomics results with our previous 16S rRNA bacterial sequencing data. The rats were fed either a standard or an obesogenic cafeteria diet (CAF) for 17 weeks. After nine weeks, rats were supplemented with vehicle; low- (H1), or high- (H2) hesperidin doses. CAF animals developed MetS features. Hesperidin supplementation in CAF rats decreased the total cholesterol, LDL-C, and free fatty acids. The highest hesperidin dose also ameliorated blood pressure, insulin sensitivity, and decreased markers of arterial stiffness and inflammation. Metabolomics revealed an improvement of the lipidomic profile, decreases in circulating amino acids, and lower excretions of inflammation- and oxidative stress-related metabolites. Bacteroidaceae increases in the CAF-H2 group paralleled higher excretions of microbial-derived metabolites. Overall, our results provide detailed insights into the molecular effects of hesperidin on MetS and suggest that it is a promising prebiotic for the treatment of MetS and related conditions.
Collapse
|
20
|
Padilla J, Woodford ML, Lastra-Gonzalez G, Martinez-Diaz V, Fujie S, Yang Y, Lising AMC, Ramirez-Perez FI, Aroor AR, Morales-Quinones M, Ghiarone T, Whaley-Connell A, Martinez-Lemus LA, Hill MA, Manrique-Acevedo C. Sexual Dimorphism in Obesity-Associated Endothelial ENaC Activity and Stiffening in Mice. Endocrinology 2019; 160:2918-2928. [PMID: 31617909 PMCID: PMC6853665 DOI: 10.1210/en.2019-00483] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 10/10/2019] [Indexed: 02/08/2023]
Abstract
Obesity and insulin resistance stiffen the vasculature, with females appearing to be more adversely affected. As augmented arterial stiffness is an independent predictor of cardiovascular disease (CVD), the increased predisposition of women with obesity and insulin resistance to arterial stiffening may explain their heightened risk for CVD. However, the cellular mechanisms by which females are more vulnerable to arterial stiffening associated with obesity and insulin resistance remain largely unknown. In this study, we provide evidence that female mice are more susceptible to Western diet-induced endothelial cell stiffening compared with age-matched males. Mechanistically, we show that the increased stiffening of the vascular intima in Western diet-fed female mice is accompanied by enhanced epithelial sodium channel (ENaC) activity in endothelial cells (EnNaC). Our data further indicate that: (i) estrogen signaling through estrogen receptor α (ERα) increases EnNaC activity to a larger extent in females compared with males, (ii) estrogen-induced activation of EnNaC is mediated by the serum/glucocorticoid inducible kinase 1 (SGK-1), and (iii) estrogen signaling stiffens endothelial cells when nitric oxide is lacking and this stiffening effect can be reduced with amiloride, an ENaC inhibitor. In aggregate, we demonstrate a sexual dimorphism in obesity-associated endothelial stiffening, whereby females are more vulnerable than males. In females, endothelial stiffening with obesity may be attributed to estrogen signaling through the ERα-SGK-1-EnNaC axis, thus establishing a putative therapeutic target for female obesity-related vascular stiffening.
Collapse
Affiliation(s)
- Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Makenzie L Woodford
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Guido Lastra-Gonzalez
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
| | - Vanesa Martinez-Diaz
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
| | - Shumpei Fujie
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Faculty of Sport and Health Sciences, University of Tsukuba, Ibaraki, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Alexandre M C Lising
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biological Engineering, University of Missouri, Columbia, Missouri
| | - Annayya R Aroor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
| | | | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Adam Whaley-Connell
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
- Division of Nephrology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biological Engineering, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
- Correspondence: Camila Manrique-Acevedo, MD, Department of Medicine, University of Missouri, D109 Diabetes Center UHC, One Hospital Drive, Columbia, Missouri 65212. E-mail:
| |
Collapse
|
21
|
Aroor AR, Habibi J, Nistala R, Ramirez-Perez FI, Martinez-Lemus LA, Jaffe IZ, Sowers JR, Jia G, Whaley-Connell A. Diet-Induced Obesity Promotes Kidney Endothelial Stiffening and Fibrosis Dependent on the Endothelial Mineralocorticoid Receptor. Hypertension 2019; 73:849-858. [PMID: 30827147 PMCID: PMC6448566 DOI: 10.1161/hypertensionaha.118.12198] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/04/2019] [Indexed: 12/29/2022]
Abstract
Obesity is characterized by enhanced MR (mineralocorticoid receptor) activation, vascular stiffness, and associated cardiovascular and kidney disease. Consumption of a Western-style diet (WD), high in saturated fat and refined carbohydrates, by female mice, leads to obesity and vascular stiffening. Use of ECMR (endothelial cell-specific MR) knockout mice supports that ECMR activation is critical for development of vascular and cardiac fibrosis and stiffening. However, the role of ECMR activation in kidney inflammation and fibrosis remains unknown. We hypothesized that cell-specific deletion of ECMR would prevent WD-induced central aortic stiffness and protect the kidney from endothelial dysfunction and vascular stiffening. Four-week-old female ECMR KO and wild-type mice were fed either mouse chow or WD for 16 weeks. WD feeding increased body weight and fat mass, proteinuria, as well as vascular stiffness indices (pulse wave velocity and kidney artery stiffening) and impaired endothelial-dependent vasodilatation without blood pressure changes. The WD-induced kidney arterial stiffening was associated with attenuated eNOS (endothelial NO synthase) activation, increased oxidative stress, proinflammatory immune responses, alterations in extracellular matrix degradation pathways, and fibrosis. ECMR deletion prevented these abnormalities by improving eNOS activation and reducing macrophage proinflammatory M1 polarization, expression of TG2 (transglutaminase 2), and MMP (matrix metalloproteinase)-9. Our data support the concept that ECMR activation contributes to endothelial dysfunction, increased kidney artery fibrosis/stiffening, and impaired NOS (NO synthase) activation, processes associated with macrophage infiltration and polarization, inflammation, and oxidative stress, collectively resulting in tubulointerstitial fibrosis in females consuming a WD.
Collapse
Affiliation(s)
- Annayya R Aroor
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Javad Habibi
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Ravi Nistala
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Nephrology and Hypertension (R.N., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri-Columbia School of Medicine (F.I.R.-P., L.A.M.-L., J.R.S.)
| | - Luis A Martinez-Lemus
- Department of Medical Pharmacology and Physiology (L.A.M.-L., J.R.S.)
- Dalton Cardiovascular Research Center, University of Missouri-Columbia School of Medicine (F.I.R.-P., L.A.M.-L., J.R.S.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.)
| | - James R Sowers
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Department of Medical Pharmacology and Physiology (L.A.M.-L., J.R.S.)
- Dalton Cardiovascular Research Center, University of Missouri-Columbia School of Medicine (F.I.R.-P., L.A.M.-L., J.R.S.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| | - Guanghong Jia
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
| | - Adam Whaley-Connell
- From the Diabetes and Cardiovascular Center (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Department of Medicine (A.R.A., J.H., R.N., J.R.S., G.J., A.W.-C.)
- Division of Endocrinology and Metabolism (A.R.A., J.H., J.R.S., G.J., A.W.-C.)
- Division of Nephrology and Hypertension (R.N., A.W.-C.)
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO (A.R.A., J.H., R.N., L.A.M.-L., J.R.S., A.W.-C.)
| |
Collapse
|
22
|
Glanz VY, Myasoedova VA, Grechko AV, Orekhov AN. Sialidase activity in human pathologies. Eur J Pharmacol 2018; 842:345-350. [PMID: 30439363 DOI: 10.1016/j.ejphar.2018.11.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/03/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
Sialic acid residues are frequently located at the terminal positions of glycoconjugate chains of cellular glycocalyx. Sialidases, or neuraminidases, catalyse removal of these residues thereby modulating various normal and pathological cellular activities. Recent studies have revealed the involvement of sialidases in a wide range of human disorders, including neurodegenerative disorders, cancers, infectious diseases and cardiovascular diseases. The accumulating data make sialidases an interesting potential therapeutic target. Modulating the activity of these enzymes may have beneficial effects in several pathologies. Four types of mammalian sialidases have been described: NEU1, NEU2, NEU3 and NEU4. They are encoded by different genes and characterized by different subcellular localization. In this review, we will summarize the current knowledge on the roles of different sialidases in pathological conditions.
Collapse
Affiliation(s)
- Victor Yu Glanz
- Department of Genetics, Cytology and Bioengineering, Faculty of Biology and Medicine, Voronezh State University, Voronezh, Russia
| | - Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Andrey V Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 109240 Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia; Centre of Collective Use, Institute of Gene Biology, Russian Academy of Sciences, Moscow 121552, Russia.
| |
Collapse
|
23
|
Kramer B, França LM, Zhang Y, Paes AMDA, Gerdes AM, Carrillo-Sepulveda MA. Western diet triggers Toll-like receptor 4 signaling-induced endothelial dysfunction in female Wistar rats. Am J Physiol Heart Circ Physiol 2018; 315:H1735-H1747. [PMID: 30265151 DOI: 10.1152/ajpheart.00218.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Overconsumption of a diet rich in fat and carbohydrates, called the Western diet, is a major contributor to the global epidemic of cardiovascular disease. Despite previously documented cardiovascular protection exhibited in female rats, this safeguard may be lost under certain metabolic stressors. We hypothesized that female Wistar rats challenged by a Western diet composed of 21% fat and 50% carbohydrate (34.1% sucrose) for 17 wk would develop endothelial dysfunction via endothelial Toll-like receptor 4 (TLR4) signaling. Western diet-fed female rats exhibited dysregulation of metabolism, revealing increased body weight and abdominal fat, decreased expression of adiponectin in white adipose tissue, glucose intolerance, and impaired insulin sensitivity. Western diet exposure increased hepatic triglycerides and cholesterol alongside hepatic steatosis, categorizing nonalcoholic fatty liver disease. Moreover, a Western diet negatively affected vascular function, revealing hypertension, impaired endothelium-dependent vasorelaxation, aortic remodeling, and increased reactive oxygen species (ROS) production. Aortic protein expression of TLR4 and its downstream proteins were markedly increased in the Western diet-fed group in association with elevated serum levels of free fatty acids. In vitro experiments were conducted to test whether free fatty acids contribute to vascular ROS overproduction via the TLR4 signaling pathway. Cultured endothelial cells were stimulated with palmitate in the presence of TAK-242, a TLR4 signaling inhibitor. Palmitate-induced overgeneration of ROS in endothelial cells was abolished in the presence of TAK-242. Our data show that a Western diet induced endothelial dysfunction in female rats and suggest that endothelial TLR4 signaling may play a key role in abolishing female cardiovascular protection. NEW & NOTEWORTHY A Western diet induced elevated levels of free fatty acids, produced nonalcoholic fatty liver disease, and provoked endothelial dysfunction in female rats in association with Toll-like receptor 4 signaling-mediated vascular reactive oxygen species production. Limited consumption of a Western diet in premenopausal women may decrease their risk of cardiovascular complications.
Collapse
Affiliation(s)
- Benjamin Kramer
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York
| | - Lucas Martins França
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão , Sao Luis , Brazil
| | - Youhua Zhang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York
| | - Antonio Marcus de Andrade Paes
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão , Sao Luis , Brazil
| | - A Martin Gerdes
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York
| | - Maria Alicia Carrillo-Sepulveda
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York
| |
Collapse
|
24
|
Elrashidy RA, Zhang J, Liu G. Long-term consumption of Western diet contributes to endothelial dysfunction and aortic remodeling in rats: Implication of Rho-kinase signaling. Clin Exp Hypertens 2018; 41:174-180. [DOI: 10.1080/10641963.2018.1462375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Rania A. Elrashidy
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Jing Zhang
- Department of Hyperbaric Oxygen, Capital Medical University Beijing Chao-Yang Hospital, Beijing, China
| | - Guiming Liu
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
25
|
Bloksgaard M, Thorsted B, Brewer JR, De Mey JGR. Assessing Collagen and Elastin Pressure-dependent Microarchitectures in Live, Human Resistance Arteries by Label-free Fluorescence Microscopy. J Vis Exp 2018. [PMID: 29683445 DOI: 10.3791/57451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pathogenic contribution of resistance artery remodeling is documented in essential hypertension, diabetes and the metabolic syndrome. Investigations and development of microstructurally motivated mathematical models for understanding the mechanical properties of human resistance arteries in health and disease have the potential to aid understanding how disease and medical treatments affect the human microcirculation. To develop these mathematical models, it is essential to decipher the relationship between the mechanical and microarchitectural properties of the microvascular wall. In this work, we describe an ex vivo method for passive mechanical testing and simultaneous label-free three-dimensional imaging of the microarchitecture of elastin and collagen in the arterial wall of isolated human resistance arteries. The imaging protocol can be applied to resistance arteries of any species of interest. Image analyses are described for quantifying i) pressure-induced changes in internal elastic lamina branching angles and adventitial collagen straightness using Fiji and ii) collagen and elastin volume densities determined using Ilastik software. Preferably all mechanical and imaging measurements are performed on live, perfused arteries, however, an alternative approach using standard video-microscopy pressure myography in combination with post-fixation imaging of re-pressurized vessels is discussed. This alternative method provides users with different options for analysis approaches. The inclusion of the mechanical and imaging data in mathematical models of the arterial wall mechanics is discussed, and future development and additions to the protocol are proposed.
Collapse
Affiliation(s)
- Maria Bloksgaard
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark;
| | - Bjarne Thorsted
- Department of Biochemistry and Molecular Biology, University of Southern Denmark
| | - Jonathan R Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark
| | - Jo G R De Mey
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark; Department of Cardiac, Thoracic and Vascular Surgery, Odense University Hospital
| |
Collapse
|
26
|
Aroor AR, Jia G, Habibi J, Sun Z, Ramirez-Perez FI, Brady B, Chen D, Martinez-Lemus LA, Manrique C, Nistala R, Whaley-Connell AT, Demarco VG, Meininger GA, Sowers JR. Uric acid promotes vascular stiffness, maladaptive inflammatory responses and proteinuria in western diet fed mice. Metabolism 2017; 74:32-40. [PMID: 28764846 PMCID: PMC5577816 DOI: 10.1016/j.metabol.2017.06.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/22/2017] [Accepted: 06/15/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Aortic vascular stiffness has been implicated in the development of cardiovascular disease (CVD) and chronic kidney disease (CKD) in obese individuals. However, the mechanism promoting these adverse effects are unclear. In this context, promotion of obesity through consumption of a western diet (WD) high in fat and fructose leads to excess circulating uric acid. There is accumulating data implicating elevated uric acid in the promotion of CVD and CKD. Accordingly, we hypothesized that xanthine oxidase(XO) inhibition with allopurinol would prevent a rise in vascular stiffness and proteinuria in a translationally relevant model of WD-induced obesity. MATERIALS/METHODS Four-week-old C57BL6/J male mice were fed a WD with excess fat (46%) and fructose (17.5%) with or without allopurinol (125mg/L in drinking water) for 16weeks. Aortic endothelial and extracellular matrix/vascular smooth muscle stiffness was evaluated by atomic force microscopy. Aortic XO activity, 3-nitrotyrosine (3-NT) and aortic endothelial sodium channel (EnNaC) expression were evaluated along with aortic expression of inflammatory markers. In the kidney, expression of toll like receptor 4 (TLR4) and fibronectin were assessed along with evaluation of proteinuria. RESULTS XO inhibition significantly attenuated WD-induced increases in plasma uric acid, vascular XO activity and oxidative stress, in concert with reductions in proteinuria. Further, XO inhibition prevented WD-induced increases in aortic EnNaC expression and associated endothelial and subendothelial stiffness. XO inhibition also reduced vascular pro-inflammatory and maladaptive immune responses induced by consumption of a WD. XO inhibition also decreased WD-induced increases in renal TLR4 and fibronectin that associated proteinuria. CONCLUSIONS Consumption of a WD leads to elevations in plasma uric acid, increased vascular XO activity, oxidative stress, vascular stiffness, and proteinuria all of which are attenuated with allopurinol administration.
Collapse
Affiliation(s)
- Annayya R Aroor
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| | - Guanghong Jia
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Javad Habibi
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Zhe Sun
- Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Barron Brady
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Dongqing Chen
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Luis A Martinez-Lemus
- Department of Medical Pharmacology and Physiology, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Camila Manrique
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Ravi Nistala
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Division of Nephrology and Hypertension, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Adam T Whaley-Connell
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Division of Nephrology and Hypertension, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Vincent G Demarco
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Department of Medical Pharmacology and Physiology, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Gerald A Meininger
- Department of Medical Pharmacology and Physiology, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, Department of Medicine, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Department of Medical Pharmacology and Physiology, University of Missouri Columbia, School of Medicine, Columbia, MO 65212, USA; Research Service Harry S Truman Memorial Veterans Hospital, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
27
|
Martinez-Lemus LA, Aroor AR, Ramirez-Perez FI, Jia G, Habibi J, DeMarco VG, Barron B, Whaley-Connell A, Nistala R, Sowers JR. Amiloride Improves Endothelial Function and Reduces Vascular Stiffness in Female Mice Fed a Western Diet. Front Physiol 2017; 8:456. [PMID: 28713285 PMCID: PMC5492307 DOI: 10.3389/fphys.2017.00456] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/15/2017] [Indexed: 01/25/2023] Open
Abstract
Obese premenopausal women lose their sex related cardiovascular disease protection and develop greater arterial stiffening than age matched men. In female mice, we have shown that consumption of a Western diet (WD), high in fat and refined sugars, is associated with endothelial dysfunction and vascular stiffening, which occur via activation of mineralocorticoid receptors and associated increases in epithelial Na+ channel (ENaC) activity on endothelial cells (EnNaC). Herein our aim was to determine the effect that reducing EnNaC activity with a very-low-dose of amiloride would have on decreasing endothelial and arterial stiffness in young female mice consuming a WD. To this end, we fed female mice either a WD or control diet and treated them with or without a very-low-dose of the ENaC-inhibitor amiloride (1 mg/kg/day) in the drinking water for 20 weeks beginning at 4 weeks of age. Mice consuming a WD were heavier and had greater percent body fat, proteinuria, and aortic stiffness as assessed by pulse-wave velocity than those fed control diet. Treatment with amiloride did not affect body weight, body composition, blood pressure, urinary sodium excretion, or insulin sensitivity, but significantly reduced the development of endothelial and aortic stiffness, aortic fibrosis, aortic oxidative stress, and mesenteric resistance artery EnNaC abundance and proteinuria in WD-fed mice. Amiloride also improved endothelial-dependent vasodilatory responses in the resistance arteries of WD-fed mice. These results indicate that a very-low-dose of amiloride, not affecting blood pressure, is sufficient to improve endothelial function and reduce aortic stiffness in female mice fed a WD, and suggest that EnNaC-inhibition may be sufficient to ameliorate the pathological vascular stiffening effects of WD-induced obesity in females.
Collapse
Affiliation(s)
- Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of MissouriColumbia, MO, United States.,Department of Biological Engineering, University of MissouriColumbia, MO, United States.,Department of Medical Pharmacology and Physiology, University of MissouriColumbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' HospitalColumbia, MO, United States
| | - Annayya R Aroor
- Research Service, Harry S. Truman Memorial Veterans' HospitalColumbia, MO, United States.,Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of MissouriColumbia, MO, United States.,Department of Biological Engineering, University of MissouriColumbia, MO, United States
| | - Guanghong Jia
- Research Service, Harry S. Truman Memorial Veterans' HospitalColumbia, MO, United States.,Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States
| | - Javad Habibi
- Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States
| | - Vincent G DeMarco
- Department of Medical Pharmacology and Physiology, University of MissouriColumbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' HospitalColumbia, MO, United States.,Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States
| | - Brady Barron
- Research Service, Harry S. Truman Memorial Veterans' HospitalColumbia, MO, United States.,Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States
| | - Adam Whaley-Connell
- Research Service, Harry S. Truman Memorial Veterans' HospitalColumbia, MO, United States.,Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States.,Division of Nephrology and Hypertension, University of MissouriColumbia, MO, United States
| | - Ravi Nistala
- Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States.,Division of Nephrology and Hypertension, University of MissouriColumbia, MO, United States
| | - James R Sowers
- Dalton Cardiovascular Research Center, University of MissouriColumbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' HospitalColumbia, MO, United States.,Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, United States
| |
Collapse
|
28
|
Manrique-Acevedo C, Ramirez-Perez FI, Padilla J, Vieira-Potter VJ, Aroor AR, Barron BJ, Chen D, Haertling D, Declue C, Sowers JR, Martinez-Lemus LA. Absence of Endothelial ERα Results in Arterial Remodeling and Decreased Stiffness in Western Diet-Fed Male Mice. Endocrinology 2017; 158:1875-1885. [PMID: 28430983 PMCID: PMC5460939 DOI: 10.1210/en.2016-1831] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/13/2017] [Indexed: 01/16/2023]
Abstract
The role of estrogen receptor-α (ERα) signaling in the vasculature of females has been described under different experimental conditions and our group recently reported that lack of endothelial cell (EC) ERα in female mice fed a Western diet (WD) results in amelioration of vascular stiffness. Conversely, the role of ERα in the male vasculature in this setting has not been explored. In conditions of overnutrition and insulin resistance, augmented arterial stiffness, endothelial dysfunction, and arterial remodeling contribute to the development of cardiovascular disease. Here, we used a rodent model of decreased ERα expression in ECs [endothelial cell estrogen receptor-α knockout (EC-ERαKO)] to test the hypothesis that, similar to our findings in females, loss of ERα signaling in the endothelium of insulin-resistant males would result in decreased arterial stiffness. EC-ERαKO male mice and same-sex littermates were fed a WD (high in fructose and fat) for 20 weeks and then assessed for vascular function and stiffness. EC-ERαKO mice were heavier than littermates but exhibited decreased vascular stiffness without differences in endothelial-dependent vasodilatory responses. Mesenteric arteries from EC-ERαKO mice had significantly increased diameters, wall cross-sectional areas, and mean wall thicknesses, indicative of outward hypertrophic remodeling. This remodeling paralleled an increased vessel wall content of collagen and elastin, inhibition of matrix metalloproteinase activation and a decrease of the incremental modulus of elasticity. In addition, internal elastic lamina fenestrae were more abundant in the EC-ERαKO mice. In conclusion, loss of endothelial ERα reduces vascular stiffness in male mice fed a WD with an associated outward hypertrophic remodeling of resistance arteries.
Collapse
Affiliation(s)
- Camila Manrique-Acevedo
- Department of Medicine, Division of Endocrinology, University of Missouri, Columbia, Missouri 65212
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
- Department of Biological Engineering, University of Missouri, Columbia, Missouri 65211
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri 65211
- Department of Child Health, University of Missouri, Columbia, Missouri 65212
| | - Victoria J Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri 65211
| | - Annayya R Aroor
- Department of Medicine, Division of Endocrinology, University of Missouri, Columbia, Missouri 65212
| | - Brady J Barron
- Department of Medicine, Division of Endocrinology, University of Missouri, Columbia, Missouri 65212
| | - Dongqing Chen
- Department of Medicine, Division of Endocrinology, University of Missouri, Columbia, Missouri 65212
| | - Dominic Haertling
- School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - Cory Declue
- School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - James R Sowers
- Department of Medicine, Division of Endocrinology, University of Missouri, Columbia, Missouri 65212
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65212
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65201
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
- Department of Biological Engineering, University of Missouri, Columbia, Missouri 65211
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65212
| |
Collapse
|
29
|
Chen K, Zheng X, Feng M, Li D, Zhang H. Gut Microbiota-Dependent Metabolite Trimethylamine N-Oxide Contributes to Cardiac Dysfunction in Western Diet-Induced Obese Mice. Front Physiol 2017; 8:139. [PMID: 28377725 PMCID: PMC5359299 DOI: 10.3389/fphys.2017.00139] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/23/2017] [Indexed: 12/24/2022] Open
Abstract
Excessive consumption of diets high in sugars and saturated fat, frequently known as western diet (WD), may lead to obesity and metabolic syndrome. Recent evidence shows that WD-induced obesity impairs cardiac function, but the underlying mechanisms are not fully understood. Trimethylamine N-oxide (TMAO), a gut microbiota-dependent metabolite of specific dietary nutrients, has emerged as a key contributor to cardiovascular disease pathogenesis. We tested the hypothesis that elevated circulating TMAO levels contribute to cardiac dysfunction in WD-induced obesity. CD1 mice were fed a normal diet (ND) or a WD, without or with 1.0% 3,3-Dimethyl-1-butanol (DMB, an inhibitor of trimethylamine formation) in drinking water for 8 weeks. Compared with mice fed a ND, mice fed a WD showed a significant increase in body weight and dyslipidemia, and had markedly higher plasma TMAO levels at the end of the feeding protocol. Echocardiography revealed that cardiac systolic and diastolic function was impaired in mice fed a WD. DMB treatment had no effects on body weight and dyslipidemia, but significantly reduced plasma TMAO levels and prevented cardiac dysfunction in mice fed a WD. In addition, mice fed a WD had elevated expression of pro-inflammatory cytokines tumor necrosis factor-α and interleukin IL-1β, decreased expression of anti-inflammatory cytokine IL-10, and increased interstitial fibrosis in the hearts, all of which were prevented by DMB treatment. Notably, DMB treatment also reduced plasma TMAO levels in mice fed a ND but did not alter other parameters. These results suggest that consumption of a WD increases circulating TMAO levels, which lead to cardiac inflammation and fibrosis, contributing to cardiac dysfunction. Interventions that reduce circulating TMAO levels may be a novel therapeutic strategy for prevention and treatment of WD-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Kui Chen
- Department of Anesthesiology, Jining NO.1 People's Hospital Jining, China
| | - Xiaoqian Zheng
- Outpatient Department, Jining NO.1 People's Hospital Jining, China
| | - Mingchen Feng
- Department of Critical Care Medicine, Jining NO.1 People's Hospital Jining, China
| | - Dongliang Li
- Department of Anesthesiology, Qilu Hospital, Shandong University Jinan, China
| | - Hongqi Zhang
- Department of Anesthesiology, Jining NO.1 People's Hospital Jining, China
| |
Collapse
|