1
|
Abdalla MMI. Insulin resistance as the molecular link between diabetes and Alzheimer's disease. World J Diabetes 2024; 15:1430-1447. [PMID: 39099819 PMCID: PMC11292327 DOI: 10.4239/wjd.v15.i7.1430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 07/08/2024] Open
Abstract
Diabetes mellitus (DM) and Alzheimer's disease (AD) are two major health concerns that have seen a rising prevalence worldwide. Recent studies have indicated a possible link between DM and an increased risk of developing AD. Insulin, while primarily known for its role in regulating blood sugar, also plays a vital role in protecting brain functions. Insulin resistance (IR), especially prevalent in type 2 diabetes, is believed to play a significant role in AD's development. When insulin signalling becomes dysfunctional, it can negatively affect various brain functions, making individuals more susceptible to AD's defining features, such as the buildup of beta-amyloid plaques and tau protein tangles. Emerging research suggests that addressing insulin-related issues might help reduce or even reverse the brain changes linked to AD. This review aims to explore the rela-tionship between DM and AD, with a focus on the role of IR. It also explores the molecular mechanisms by which IR might lead to brain changes and assesses current treatments that target IR. Understanding IR's role in the connection between DM and AD offers new possibilities for treatments and highlights the importance of continued research in this interdisciplinary field.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
2
|
Milewski K, Orzeł-Gajowik K, Zielińska M. Mitochondrial Changes in Rat Brain Endothelial Cells Associated with Hepatic Encephalopathy: Relation to the Blood-Brain Barrier Dysfunction. Neurochem Res 2024; 49:1489-1504. [PMID: 35917006 PMCID: PMC11106209 DOI: 10.1007/s11064-022-03698-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/17/2022] [Accepted: 07/14/2022] [Indexed: 12/06/2022]
Abstract
The mechanisms underlying cerebral vascular dysfunction and edema during hepatic encephalopathy (HE) are unclear. Blood-brain barrier (BBB) impairment, resulting from increased vascular permeability, has been reported in acute and chronic HE. Mitochondrial dysfunction is a well-documented result of HE mainly affecting astrocytes, but much less so in the BBB-forming endothelial cells. Here we review literature reports and own experimental data obtained in HE models emphasizing alterations in mitochondrial dynamics and function as a possible contributor to the status of brain endothelial cell mitochondria in HE. Own studies on the expression of the mitochondrial fusion-fission controlling genes rendered HE animal model-dependent effects: increase of mitochondrial fusion controlling genes opa1, mfn1 in cerebral vessels in ammonium acetate-induced hyperammonemia, but a decrease of the two former genes and increase of fis1 in vessels in thioacetamide-induced HE. In endothelial cell line (RBE4) after 24 h ammonia and/or TNFα treatment, conditions mimicking crucial aspects of HE in vivo, we observed altered expression of mitochondrial fission/fusion genes: a decrease of opa1, mfn1, and, increase of the fission related fis1 gene. The effect in vitro was paralleled by the generation of reactive oxygen species, decreased total antioxidant capacity, decreased mitochondrial membrane potential, as well as increased permeability of RBE4 cell monolayer to fluorescein isothiocyanate dextran. Electron microscopy documented enlarged mitochondria in the brain endothelial cells of rats in both in vivo models. Collectively, the here observed alterations of cerebral endothelial mitochondria are indicative of their fission, and decreased potential of endothelial mitochondria are likely to contribute to BBB dysfunction in HE.
Collapse
Affiliation(s)
- Krzysztof Milewski
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego St. 5, 02-106, Warsaw, Poland.
| | - Karolina Orzeł-Gajowik
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego St. 5, 02-106, Warsaw, Poland
| | - Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego St. 5, 02-106, Warsaw, Poland.
| |
Collapse
|
3
|
Carter KJ, Ward AT, Al-Subu A, Wilson AD, Zevin EL, Serlin RC, Eldridge M, Wieben O, Schrage WG. An oral glucose tolerance test does not affect cerebral blood flow: role of NOS. Am J Physiol Regul Integr Comp Physiol 2023; 325:R759-R768. [PMID: 37842740 PMCID: PMC11178292 DOI: 10.1152/ajpregu.00169.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/11/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023]
Abstract
Animal data indicate that insulin triggers a robust nitric oxide synthase (NOS)-mediated dilation in cerebral arteries similar to the peripheral tissue vasodilation observed in healthy adults. Insulin's role in regulating cerebral blood flow (CBF) in humans remains unclear but may be important for understanding the links between insulin resistance, diminished CBF, and poor brain health outcomes. We tested the hypothesis that an oral glucose challenge (oral glucose tolerance test, OGTT), which increases systemic insulin and glucose, would acutely increase CBF in healthy adults due to NOS-mediated vasodilation, and that changes in CBF would be greater in anterior regions where NOS expression or activity may be greater. In a randomized, single-blind approach, 18 young healthy adults (24 ± 5 yr) underwent magnetic resonance imaging (MRI) with a placebo before and after an OGTT (75 g glucose), and 11 of these adults also completed an NG-monomethyl-l-arginine (l-NMMA) visit. Four-dimensional (4-D) flow MRI quantified macrovascular CBF and arterial spin labeling (ASL) quantified microvascular perfusion. Subjects completed baseline imaging with a placebo (or l-NMMA), then consumed an OGTT followed by MRI scans and blood sampling every 10-15 min for 90 min. Contrary to our hypothesis, total CBF (P = 0.17) and global perfusion (P > 0.05) did not change at any time point up to 60 min after the OGTT, and no regional changes were detected. l-NMMA did not mediate any effect of OGTT on CBF. These data suggest that insulin-glucose challenge does not acutely alter CBF in healthy adults.
Collapse
Affiliation(s)
- Katrina J Carter
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| | - Aaron T Ward
- Deparment of Center for Health Disparities Research, University of Wisconsin, Madison, Wisconsin, United States
| | - Awni Al-Subu
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Allen D Wilson
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Erika L Zevin
- Department of Pediatrics, University School of Medicine, Indianapolis, Indiana, United States
| | - Ronald C Serlin
- Department of Educational Psychology, University of Wisconsin, Madison, Wisconsin, United States
| | - Marlowe Eldridge
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Oliver Wieben
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, United States
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, United States
| | - William G Schrage
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| |
Collapse
|
4
|
Daniel JM, Lindsey SH, Mostany R, Schrader LA, Zsombok A. Cardiometabolic health, menopausal estrogen therapy and the brain: How effects of estrogens diverge in healthy and unhealthy preclinical models of aging. Front Neuroendocrinol 2023; 70:101068. [PMID: 37061205 PMCID: PMC10725785 DOI: 10.1016/j.yfrne.2023.101068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
Research in preclinical models indicates that estrogens are neuroprotective and positively impact cognitive aging. However, clinical data are equivocal as to the benefits of menopausal estrogen therapy to the brain and cognition. Pre-existing cardiometabolic disease may modulate mechanisms by which estrogens act, potentially reducing or reversing protections they provide against cognitive decline. In the current review we propose mechanisms by which cardiometabolic disease may alter estrogen effects, including both alterations in actions directly on brain memory systems and actions on cardiometabolic systems, which in turn impact brain memory systems. Consideration of mechanisms by which estrogen administration can exert differential effects dependent upon health phenotype is consistent with the move towards precision or personalized medicine, which aims to determine which treatment interventions will work for which individuals. Understanding effects of estrogens in both healthy and unhealthy models of aging is critical to optimizing the translational link between preclinical and clinical research.
Collapse
Affiliation(s)
- Jill M Daniel
- Department of Psychology and Brain Institute, Tulane University, New Orleans, LA, United States.
| | - Sarah H Lindsey
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Ricardo Mostany
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Laura A Schrader
- Department of Cell & Molecular Biology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Andrea Zsombok
- Department of Physiology and Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
5
|
Carter KJ, Ward AT, Kellawan JM, Harrell JW, Peltonen GL, Roberts GS, Al-Subu A, Hagen SA, Serlin RC, Eldridge MW, Wieben O, Schrage WG. Reduced basal macrovascular and microvascular cerebral blood flow in young adults with metabolic syndrome: potential mechanisms. J Appl Physiol (1985) 2023; 135:94-108. [PMID: 37199780 PMCID: PMC10292973 DOI: 10.1152/japplphysiol.00688.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/26/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023] Open
Abstract
Ninety-million Americans suffer metabolic syndrome (MetSyn), increasing the risk of diabetes and poor brain outcomes, including neuropathology linked to lower cerebral blood flow (CBF), predominantly in anterior regions. We tested the hypothesis that total and regional CBF is lower in MetSyn more so in the anterior brain and explored three potential mechanisms. Thirty-four controls (25 ± 5 yr) and 19 MetSyn (30 ± 9 yr), with no history of cardiovascular disease/medications, underwent four-dimensional flow magnetic resonance imaging (MRI) to quantify macrovascular CBF, whereas arterial spin labeling quantified brain perfusion in a subset (n = 38/53). Contributions of cyclooxygenase (COX; n = 14), nitric oxide synthase (NOS, n = 17), or endothelin receptor A signaling (n = 13) were tested with indomethacin, NG-monomethyl-L-arginine (L-NMMA), and Ambrisentan, respectively. Total CBF was 20 ± 16% lower in MetSyn (725 ± 116 vs. 582 ± 119 mL/min, P < 0.001). Anterior and posterior brain regions were 17 ± 18% and 30 ± 24% lower in MetSyn; reductions were not different between regions (P = 0.112). Global perfusion was 16 ± 14% lower in MetSyn (44 ± 7 vs. 36 ± 5 mL/100 g/min, P = 0.002) and regionally in frontal, occipital, parietal, and temporal lobes (range 15-22%). The decrease in CBF with L-NMMA (P = 0.004) was not different between groups (P = 0.244, n = 14, 3), and Ambrisentan had no effect on either group (P = 0.165, n = 9, 4). Interestingly, indomethacin reduced CBF more in Controls in the anterior brain (P = 0.041), but CBF decrease in posterior was not different between groups (P = 0.151, n = 8, 6). These data indicate that adults with MetSyn exhibit substantially reduced brain perfusion without regional differences. Moreover, this reduction is not due to loss of NOS or gain of ET-1 signaling but rather a loss of COX vasodilation.NEW & NOTEWORTHY We tested the impact of insulin resistance (IR) on resting cerebral blood flow (CBF) in adults with metabolic syndrome (MetSyn). Using MRI and research pharmaceuticals to study the role of NOS, ET-1, or COX signaling, we found that adults with MetSyn exhibit substantially lower CBF that is not explained by changes in NOS or ET-1 signaling. Interestingly, adults with MetSyn show a loss of COX-mediated vasodilation in the anterior but not posterior circulation.
Collapse
Affiliation(s)
- Katrina J Carter
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| | - Aaron T Ward
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| | - J Mikhail Kellawan
- Department of Health and Exercise Science, University of Oklahoma, Norman, Oklahoma, United States
| | - John W Harrell
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, Ohio, United States
| | - Garrett L Peltonen
- School of Nursing and Kinesiology, Western New Mexico University, Silver City, New Mexico, United States
| | - Grant S Roberts
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, United States
| | - Awni Al-Subu
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Scott A Hagen
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Ronald C Serlin
- Department of Educational Psychology, University of Wisconsin, Madison, Wisconsin, United States
| | - Marlowe W Eldridge
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, United States
| | - Oliver Wieben
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, United States
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, United States
| | - William G Schrage
- Department of Kinesiology, University of Wisconsin, Madison, Wisconsin, United States
| |
Collapse
|
6
|
Qiao YS, Tang X, Chai YH, Gong HJ, Xu H, Patel I, Li L, Lu T, Zhao WY, Li ZY, Cardoso MA, Zhou JB. Cerebral Blood Flow Alterations and Obesity: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2022; 90:15-31. [DOI: 10.3233/jad-220601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Reduction in cerebral blood flow (CBF) plays an essential role in the cognitive impairment and dementia in obesity. However, current conclusions regarding CBF changes in patients with obesity are inconsistent. Objective: A systematic review and meta-analysis was performed to evaluate the relationship between obesity and CBF alterations. Methods: We systematically screened published cross-sectional and longitudinal studies focusing on the differences in CBF between obese and normal-weight individuals. Eighteen studies including 24,866 participants, of which seven articles reported longitudinal results, were evaluated in the present study. Results: The results of the meta-analysis showed that in cross-sectional studies, body mass index (BMI) was negatively associated with CBF (β= –0.31, 95% confidence interval [CI]: –0.44, –0.19). Moreover, this systematic review demonstrated that obese individuals showed global and regional reductions in the CBF and increased CBF in diverse functional areas of the frontal lobe, including the prefrontal cortex, left frontal superior orbital, right frontal mid-orbital cortex, and left premotor superior frontal gyrus. Conclusion: Our findings suggest that BMI, rather than waist circumference and waist-to-hip ratio, is inversely associated with CBF in cross-sectional studies. The CBF of obese individuals showed global and regional reductions, including the frontal lobe, temporal and parietal lobes, cerebellum, hippocampus, and thalamus.
Collapse
Affiliation(s)
- Yu-Shun Qiao
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | | | - Yin-He Chai
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hong-Jian Gong
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hui Xu
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ikramulhaq Patel
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Li Li
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Tong Lu
- Department of Clinical Nutrition, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wan-Ying Zhao
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ze-Yu Li
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Marly Augusto Cardoso
- Department of Nutrition, School of Public Health, University of Sao Paulo, Sao Paulo, Brazil
| | - Jian-Bo Zhou
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Zevin EL, Peterson AL, Dodge A, Zhang X, Carrel AL. Low HDL-C is a non-fasting marker of insulin resistance in children. J Pediatr Endocrinol Metab 2022; 35:890-894. [PMID: 35649511 PMCID: PMC9249377 DOI: 10.1515/jpem-2021-0751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/13/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Childhood obesity and associated comorbidities, including insulin resistance, are increasing in the United States. Our objectives were to (1) determine the prevalence of insulin resistance in children seen in dyslipidemia clinic and (2) evaluate which aspects of the lipid profile correlate with insulin resistance. METHODS Children and adolescents seen in a specialized pediatric dyslipidemia clinic without secondary diagnoses known to alter the lipid panel were included. Simultaneous fasting lipid panel, insulin, and glucose levels were available in 572 children (50.5% male). RESULTS Mean patient age was 15.0 ± 3.6 years with the majority being over 10 years of age (92.5%). Mean BMI was 29.8 ± 8.1 kg/m2 and BMI standard deviation score was 1.80 ± 0.9. Mean HOMA-IR was 6.2 ± 5.7 with a range of 0.4-49.3, and interquartile range of 2.7-7.6. Triglyceride level had a positive correlation with HOMA-IR (p<0.001). HDL-C negatively correlated with HOMA-IR even controlling for triglyceride level by multivariate analysis (p=0.001) and HDL-C <30 mg/dL predicted IR with 41.5% PPV. CONCLUSIONS In children and adolescents with dyslipidemia, insulin resistance is common and significantly correlates with reduced HDL-C levels. Non-fasting samples are easier to obtain in children and low HDL-C, which is minimally affected on non-fasting samples, could be an easily obtained indicator of IR. Increasing detection of insulin resistance in children with dyslipidemia may provide greater opportunities for lifestyle interventions and possible pharmacotherapy to modify cardiovascular risk.
Collapse
Affiliation(s)
- Erika L Zevin
- Division of Pediatric Endocrinology and Diabetes, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Amy L Peterson
- Division of Pediatric Cardiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Ann Dodge
- Division of Pediatric Cardiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Xiao Zhang
- Division of Pediatric Cardiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Aaron L Carrel
- Division of Pediatric Endocrinology and Diabetes, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
8
|
Olsthoorn L, Vreeken D, Kiliaan AJ. Gut Microbiome, Inflammation, and Cerebrovascular Function: Link Between Obesity and Cognition. Front Neurosci 2021; 15:761456. [PMID: 34938153 PMCID: PMC8685335 DOI: 10.3389/fnins.2021.761456] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity affects 13% of the adult population worldwide and this number is only expected to increase. Obesity is known to have a negative impact on cardiovascular and metabolic health, but it also impacts brain structure and function; it is associated with both gray and white matter integrity loss, as well as decreased cognitive function, including the domains of executive function, memory, inhibition, and language. Especially midlife obesity is associated with both cognitive impairment and an increased risk of developing dementia at later age. However, underlying mechanisms are not yet fully revealed. Here, we review recent literature (published between 2010 and March 2021) and discuss the effects of obesity on brain structure and cognition, with a main focus on the contributions of the gut microbiome, white adipose tissue (WAT), inflammation, and cerebrovascular function. Obesity-associated changes in gut microbiota composition may cause increased gut permeability and inflammation, therewith affecting cognitive function. Moreover, excess of WAT in obesity produces pro-inflammatory adipokines, leading to a low grade systemic peripheral inflammation, which is associated with decreased cognition. The blood-brain barrier also shows increased permeability, allowing among others, peripheral pro-inflammatory markers to access the brain, leading to neuroinflammation, especially in the hypothalamus, hippocampus and amygdala. Altogether, the interaction between the gut microbiota, WAT inflammation, and cerebrovascular integrity plays a significant role in the link between obesity and cognition. Future research should focus more on the interplay between gut microbiota, WAT, inflammation and cerebrovascular function to obtain a better understanding about the complex link between obesity and cognitive function in order to develop preventatives and personalized treatments.
Collapse
Affiliation(s)
- Lisette Olsthoorn
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| | - Debby Vreeken
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands.,Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| |
Collapse
|
9
|
Dardi P, Perazza LR, Couto GK, Campos GP, Capettini LDSA, Rossoni LV. Vena cava presents endothelial dysfunction prior to thoracic aorta in heart failure: the pivotal role of nNOS uncoupling/oxidative stress. Clin Sci (Lond) 2021; 135:2625-2641. [PMID: 34783347 DOI: 10.1042/cs20210810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022]
Abstract
Arterial endothelial dysfunction has been extensively studied in heart failure (HF). However, little is known about the adjustments shown by the venous system in this condition. Considering that inferior vena cava (VC) tone could influence cardiac performance and HF prognosis, the aim of the present study was to assess the VC and thoracic aorta (TA) endothelial function of HF-post-myocardial infarction (MI) rats, comparing both endothelial responses and signaling pathways developed. Vascular reactivity of TA and VC from HF post-MI and sham operated (SO) rats was assessed with a wire myograph, 4 weeks after coronary artery occlusion surgery. Nitric oxide (NO), H2O2 production and oxidative stress were evaluated in situ with fluorescent probes, while protein expression and dimer/monomer ratio was assessed by Western blot. VC from HF rats presented endothelial dysfunction, while TA exhibited higher acetylcholine (ACh)-induced vasodilation when compared with vessels from SO rats. TA exhibited increased ACh-induced NO production due to a higher coupling of endothelial and neuronal NO synthases isoforms (eNOS, nNOS), and enhanced expression of antioxidant enzymes. These adjustments, however, were absent in VC of HF post-MI rats, which exhibited uncoupled nNOS, oxidative stress and higher H2O2 bioavailability. Altogether, the present study suggests a differential regulation of endothelial function between VC and TA of HF post-MI rats, most likely due to nNOS uncoupling and compromised antioxidant defense.
Collapse
Affiliation(s)
- Patrizia Dardi
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Laís Rossi Perazza
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gisele Kruger Couto
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gianne Paul Campos
- Department of Pharmacology, Institute of Biological Science, University of Minas Gerais, Minas Gerais, Brazil
| | | | - Luciana Venturini Rossoni
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
10
|
Wang Y, Sun L, He G, Gang X, Zhao X, Wang G, Ning G. Cerebral perfusion alterations in type 2 diabetes mellitus - a systematic review. Front Neuroendocrinol 2021; 62:100916. [PMID: 33957174 DOI: 10.1016/j.yfrne.2021.100916] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/04/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is related to abnormal brain structure and function, increasing the risk of cognitive impairment and dementia. We systematically reviewed the published literature focusing on cerebral perfusion in patients with T2DM. Although no significant difference was found in global cerebral blood flow (CBF) between the T2DM group and the healthy control group, the regional cerebral perfusion in T2DM was significantly reduced in multiple locations, including the occipital lobe, domains involved in the default mode network and the cerebellum. The decline in regional CBF was associated with a wide range of cognitive disorders in T2DM, including learning, memory, attention, and executive processing, as well as visual function. In addition, diabetes-related biochemical indicators, such as glycated hemoglobin and insulin resistance, were negatively correlated with regional CBF. In general, these functional perfusion imaging studies indicate that decreased CBF in T2DM may be a potential cause of cognitive impairment.
Collapse
Affiliation(s)
- Yaqiong Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China
| | - Guangyu He
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xue Zhao
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Guixia Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Guang Ning
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, China; National Clinical Research Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Milewski K, Czarnecka AM, Albrecht J, Zielińska M. Decreased Expression and Uncoupling of Endothelial Nitric Oxide Synthase in the Cerebral Cortex of Rats with Thioacetamide-Induced Acute Liver Failure. Int J Mol Sci 2021; 22:6662. [PMID: 34206365 PMCID: PMC8268495 DOI: 10.3390/ijms22136662] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/04/2023] Open
Abstract
Acute liver failure (ALF) is associated with deregulated nitric oxide (NO) signaling in the brain, which is one of the key molecular abnormalities leading to the neuropsychiatric disorder called hepatic encephalopathy (HE). This study focuses on the effect of ALF on the relatively unexplored endothelial NOS isoform (eNOS). The cerebral prefrontal cortices of rats with thioacetamide (TAA)-induced ALF showed decreased eNOS expression, which resulted in an overall reduction of NOS activity. ALF also decreased the content of the NOS cofactor, tetrahydro-L-biopterin (BH4), and evoked eNOS uncoupling (reduction of the eNOS dimer/monomer ratio). The addition of the NO precursor L-arginine in the absence of BH4 potentiated ROS accumulation, whereas nonspecific NOS inhibitor L-NAME or EDTA attenuated ROS increase. The ALF-induced decrease of eNOS content and its uncoupling concurred with, and was likely causally related to, both increased brain content of reactive oxidative species (ROS) and decreased cerebral cortical blood flow (CBF) in the same model.
Collapse
Affiliation(s)
| | | | | | - Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str, 02-106 Warsaw, Poland; (K.M.); (A.M.C.); (J.A.)
| |
Collapse
|
12
|
Kiyohara T, Matsuo R, Hata J, Nakamura K, Wakisaka Y, Kamouchi M, Kitazono T, Ago T. β-Cell Function and Clinical Outcome in Nondiabetic Patients With Acute Ischemic Stroke. Stroke 2021; 52:2621-2628. [PMID: 33985365 DOI: 10.1161/strokeaha.120.031392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Takuya Kiyohara
- Department of Medicine and Clinical Science (T. Kiyohara, R.M., J.H., K.N., Y.W., T. Kitazono, T.A.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Cerebrovascular Disease and Neurology, Hakujyuji Hospital, Fukuoka, Japan (T. Kiyohara)
| | - Ryu Matsuo
- Department of Medicine and Clinical Science (T. Kiyohara, R.M., J.H., K.N., Y.W., T. Kitazono, T.A.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Health Care Administration and Management (R.M., M.K.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies (R.M., M.K., T. Kitazono), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Hata
- Department of Medicine and Clinical Science (T. Kiyohara, R.M., J.H., K.N., Y.W., T. Kitazono, T.A.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Epidemiology and Public Health (J.H.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kuniyuki Nakamura
- Department of Medicine and Clinical Science (T. Kiyohara, R.M., J.H., K.N., Y.W., T. Kitazono, T.A.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinobu Wakisaka
- Department of Medicine and Clinical Science (T. Kiyohara, R.M., J.H., K.N., Y.W., T. Kitazono, T.A.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Kamouchi
- Department of Health Care Administration and Management (R.M., M.K.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies (R.M., M.K., T. Kitazono), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science (T. Kiyohara, R.M., J.H., K.N., Y.W., T. Kitazono, T.A.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies (R.M., M.K., T. Kitazono), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science (T. Kiyohara, R.M., J.H., K.N., Y.W., T. Kitazono, T.A.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
13
|
Can dipeptidyl peptidase-4 inhibitors treat cognitive disorders? Pharmacol Ther 2020; 212:107559. [PMID: 32380197 DOI: 10.1016/j.pharmthera.2020.107559] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022]
Abstract
The linkage of neurodegenerative diseases with insulin resistance (IR) and type 2 diabetes mellitus (T2DM), including oxidative stress, mitochondrial dysfunction, excessive inflammatory responses and abnormal protein processing, and the correlation between cerebrovascular diseases and hyperglycemia has opened a new window for novel therapeutics for these cognitive disorders. Various antidiabetic agents have been studied for their potential treatment of cognitive disorders, among which the dipeptidyl peptidase-4 (DPP-4) inhibitors have been investigated more recently. So far, DPP-4 inhibitors have demonstrated neuroprotection and cognitive improvements in animal models, and cognitive benefits in diabetic patients with or without cognitive impairments. This review aims to summarize the potential mechanisms, advantages and limitations, and currently available evidence for developing DPP-4 inhibitors as a treatment of cognitive disorders.
Collapse
|
14
|
Olver TD, Laughlin MH, Padilla J. Exercise and Vascular Insulin Sensitivity in the Skeletal Muscle and Brain. Exerc Sport Sci Rev 2019; 47:66-74. [PMID: 30883470 DOI: 10.1249/jes.0000000000000182] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We present the hypothesis that exercise-induced hyperemia, perhaps through vascular shear stress, represents an important factor responsible for the effects of physical activity (PA) on vascular insulin sensitivity. Specifically, we postulate PA involving the greatest amount of skeletal muscle mass and the greatest central neural recruitment maximizes perfusion and consequently enhances vascular insulin sensitivity in the skeletal muscle and brain.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - M Harold Laughlin
- Department of Biomedical Sciences.,Dalton Cardiovascular Research Center
| | - Jaume Padilla
- Dalton Cardiovascular Research Center.,Department of Nutrition and Exercise Physiology, and.,Department of Child Health, University of Missouri, Columbia, MO
| |
Collapse
|
15
|
Natarajan M, Habib SL, Reddick RL, Delma CR, Manickam K, Prihoda TJ, Werner SL, Mohan S. Endothelial cell-specific overexpression of endothelial nitric oxide synthase in Ins2Akita mice exacerbates diabetic nephropathy. J Diabetes Complications 2019; 33:23-32. [PMID: 30424931 PMCID: PMC6344355 DOI: 10.1016/j.jdiacomp.2018.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/13/2018] [Accepted: 10/07/2018] [Indexed: 10/28/2022]
Abstract
Previous studies demonstrated that global deficiency of eNOS in diabetic mice exacerbated renal lesions and that overexpression of eNOS may protect against tissue injury. Our study revealed for the first time overexpression of eNOS leads to disease progression rather than protection. Transgenic mice selectively expressing eNOS in endothelial cells (eNOSTg) were cross bred with Ins2Akita type-1 (AK) diabetic mice to generate eNOS overexpressing eNOSTg/AK mice. Wild type, eNOSTg, AK and eNOSTg/AK mice were assessed for kidney function and blood glucose levels. Remarkably, overexpressing eNOSTg mice showed evidence of unpredicted glomerular injury with segmental mesangiolysis and occasional microaneurysms. Notably, in eNOSTg/AK mice overexpression of eNOS led to increased glomerular/endothelial injury that was associated with increased superoxide levels and renal dysfunction. Results indicate for the first time that overexpressing eNOS in endothelial cells cannot ameliorate diabetic lesions, but paradoxically leads to progression of nephropathy likely due to eNOS uncoupling and superoxide upsurge. This novel finding has a significant impact on current therapeutic strategies to improve endothelial function and prevent progression of diabetic renal disease. Further, the eNOSTg/AK model developed in this study has significant translational potentials for elucidating the underlying mechanism implicated in the deflected function of eNOS in diabetic nephropathy.
Collapse
Affiliation(s)
- Mohan Natarajan
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Samy L Habib
- Geriatric Research Education and Clinical Center, South Texas Veterans Healthcare System and Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Robert L Reddick
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Caroline R Delma
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Krishnan Manickam
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Thomas J Prihoda
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sherry L Werner
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sumathy Mohan
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
16
|
Generation and characterization of functional phosphoserine-incorporated neuronal nitric oxide synthase holoenzyme. J Biol Inorg Chem 2018; 24:1-9. [PMID: 30315355 DOI: 10.1007/s00775-018-1621-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/05/2018] [Indexed: 12/19/2022]
Abstract
Phosphorylation is an important pathway for the regulation of nitric oxide synthase (NOS) at the posttranslational level. However, the molecular underpinnings of NOS regulation by phosphorylations remain unclear to date, mainly because of the problems in making a good amount of active phospho-NOS proteins. Herein, we have established a system in which recombinant rat nNOS holoprotein can be produced with site-specific incorporation of phosphoserine (pSer) at residue 1412, using a specialized bacterial host strain for pSer incorporation. The pSer1412 nNOS protein demonstrates UV-Vis, far-UV CD and fluorescence spectral properties that are identical to those of nNOS overexpressed in other bacterial strains. The protein is also functional, possessing normal NO production and NADPH oxidation activities in the presence of abundant substrate L-Arg. Conversely, the rate of FMN-heme interdomain electron transfer (IET) in pSer1412 nNOS is considerably lower than that of wild-type (wt) nNOS, while the phosphomimetic S1142E mutant possesses similar electron transfer kinetics to that of wt. The successful incorporation and high yield of pSer1412 into rat nNOS and the significant change in the IET kinetics upon the phosphorylation demonstrate a highly useful method for incorporating native phosphorylation sites as a substantial improvement to commonly used phosphomimetics.
Collapse
|
17
|
Brooks S, Branyan KW, DeVallance E, Skinner R, Lemaster K, Sheets JW, Pitzer CR, Asano S, Bryner RW, Olfert IM, Frisbee JC, Chantler PD. Psychological stress-induced cerebrovascular dysfunction: the role of metabolic syndrome and exercise. Exp Physiol 2018; 103:761-776. [PMID: 29436736 PMCID: PMC5927836 DOI: 10.1113/ep086892] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/09/2018] [Indexed: 01/04/2023]
Abstract
NEW FINDINGS What is the central question of this study? How does chronic stress impact cerebrovascular function and does metabolic syndrome accelerate the cerebrovascular adaptations to stress? What role does exercise training have in preventing cerebrovascular changes to stress and metabolic syndrome? What is the main finding and its importance? Stressful conditions lead to pathological adaptations of the cerebrovasculature via an oxidative nitric oxide pathway, and the presence of metabolic syndrome produces a greater susceptibility to stress-induced cerebrovascular dysfunction. The results also provide insight into the mechanisms that may contribute to the influence of stress and the role of exercise in preventing the negative actions of stress on cerebrovascular function and structure. ABSTRACT Chronic unresolvable stress leads to the development of depression and cardiovascular disease. There is a high prevalence of depression with the metabolic syndrome (MetS), but to what extent the MetS concurrent with psychological stress affects cerebrovascular function is unknown. We investigated the differential effect of MetS on cerebrovascular structure/function in rats (16-17 weeks old) following 8 weeks of unpredictable chronic mild stress (UCMS) and whether exercise training could limit any cerebrovascular dysfunction. In healthy lean Zucker rats (LZR), UCMS decreased (28%, P < 0.05) ex vivo middle cerebral artery (MCA) endothelium-dependent dilatation (EDD), but changes in MCA remodelling and stiffness were not evident, though cerebral microvessel density (MVD) decreased (30%, P < 0.05). The presence of UCMS and MetS (obese Zucker rats; OZR) decreased MCA EDD (35%, P < 0.05) and dilatation to sodium nitroprusside (20%, P < 0.05), while MCA stiffness increased and cerebral MVD decreased (31%, P < 0.05), which were linked to reduced nitric oxide and increased oxidative levels. Aerobic exercise prevented UCMS impairments in MCA function and MVD in LZR, and partly restored MCA function, stiffness and MVD in OZR. Our data suggest that the benefits of exercise with UCMS were due to a reduction in oxidative stress and increased production of nitric oxide in the cerebral vessels. In conclusion, UCMS significantly impaired MCA structure and function, but the effects of UCMS were more substantial in OZR vs. LZR. Importantly, aerobic exercise when combined with UCMS prevented the MCA dysfunction through subtle shifts in nitric oxide and oxidative stress in the cerebral microvasculature.
Collapse
Affiliation(s)
- Steven Brooks
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Kayla W Branyan
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Evan DeVallance
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Roy Skinner
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Kent Lemaster
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - J Whitney Sheets
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Christopher R Pitzer
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Shinichi Asano
- Department of Health and Human Performance, Fairmont State University, WV, USA
| | - Randall W Bryner
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - I Mark Olfert
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
- Center for Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Jefferson C Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Paul D Chantler
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
- Center for Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV, USA
| |
Collapse
|
18
|
Neth BJ, Craft S. Insulin Resistance and Alzheimer's Disease: Bioenergetic Linkages. Front Aging Neurosci 2017; 9:345. [PMID: 29163128 PMCID: PMC5671587 DOI: 10.3389/fnagi.2017.00345] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a well-established feature of Alzheimer's disease (AD), evidenced by brain glucose hypometabolism that can be observed potentially decades prior to the development of AD symptoms. Furthermore, there is mounting support for an association between metabolic disease and the development of AD and related dementias. Individuals with insulin resistance, type 2 diabetes mellitus (T2D), hyperlipidemia, obesity, or other metabolic disease may have increased risk for the development of AD and similar conditions, such as vascular dementia. This association may in part be due to the systemic mitochondrial dysfunction that is common to these pathologies. Accumulating evidence suggests that mitochondrial dysfunction is a significant feature of AD and may play a fundamental role in its pathogenesis. In fact, aging itself presents a unique challenge due to inherent mitochondrial dysfunction and prevalence of chronic metabolic disease. Despite the progress made in understanding the pathogenesis of AD and in the development of potential therapies, at present we remain without a disease-modifying treatment. In this review, we will discuss insulin resistance as a contributing factor to the pathogenesis of AD, as well as the metabolic and bioenergetic disruptions linking insulin resistance and AD. We will also focus on potential neuroimaging tools for the study of the metabolic dysfunction commonly seen in AD with hopes of developing therapeutic and preventative targets.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
19
|
Abstract
Insulin resistance often refers to a pathological condition in which cells fail to respond to the normal actions of insulin. Increasing literature has noted a critical role of insulin resistance in the pathogenesis of ischemic stroke. Insulin resistance plays an important role in the pathogenesis of ischemic stroke via enhancing advanced changes of atherosclerosis. A variety of literature indicates that insulin resistance enhances platelet adhesion, activation and aggregation which are conducive to the occurrence of ischemic stroke. Insulin resistance also induces hemodynamic disturbances and contributes to the onset of ischemic stroke. In addition, insulin resistance may augment the role of the modifiable risk factors in ischemic stroke and induce the occurrence of ischemic stroke. Preclinical and clinical studies have supported that improving insulin resistance may be an effective measure to prevent or delay ischemic stroke.
Collapse
Affiliation(s)
- Xiao-Ling Deng
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan Renmin Hospital, Shiyan, 442000, Hubei Province, People's Republic of China
| | - Zhou Liu
- Department of Neurology, The Affiliated Hospital of Guangdong Medical University, and Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong Province, People's Republic of China
| | - Chuanling Wang
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yanfeng Li
- Department of Neurology, Peking Union Medical College Hospital, No.1 Shuaifuyuan Wangfujing, Dongcheng District, Beijing, 100730, People's Republic of China.
| | - Zhiyou Cai
- Department of Neurology, Chongqing General Hospital, No. 312 Zhongshan First Road, Yuzhong District, Chongqing, 400013, People's Republic of China.
| |
Collapse
|
20
|
Hoscheidt SM, Kellawan JM, Berman SE, Rivera-Rivera LA, Krause RA, Oh JM, Beeri MS, Rowley HA, Wieben O, Carlsson CM, Asthana S, Johnson SC, Schrage WG, Bendlin BB. Insulin resistance is associated with lower arterial blood flow and reduced cortical perfusion in cognitively asymptomatic middle-aged adults. J Cereb Blood Flow Metab 2017; 37:2249-2261. [PMID: 27488909 PMCID: PMC5464714 DOI: 10.1177/0271678x16663214] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Insulin resistance (IR) is associated with poor cerebrovascular health and increased risk for dementia. Little is known about the unique effect of IR on both micro- and macrovascular flow particularly in midlife when interventions against dementia may be most effective. We examined the effect of IR as indexed by the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) on cerebral blood flow in macro- and microvessels utilizing magnetic resonance imaging (MRI) among cognitively asymptomatic middle-aged individuals. We hypothesized that higher HOMA-IR would be associated with reduced flow in macrovessels and lower cortical perfusion. One hundred and twenty cognitively asymptomatic middle-aged adults (57 ± 5 yrs) underwent fasting blood draw, phase contrast-vastly undersampled isotropic projection reconstruction (PC VIPR) MRI, and arterial spin labeling (ASL) perfusion. Higher HOMA-IR was associated with lower arterial blood flow, particularly within the internal carotid arteries (ICAs), and lower cerebral perfusion in several brain regions including frontal and temporal lobe regions. Higher blood flow in bilateral ICAs predicted greater cortical perfusion in individuals with lower HOMA-IR, a relationship not observed among those with higher HOMA-IR. Findings provide novel evidence for an uncoupling of macrovascular blood flow and microvascular perfusion among individuals with higher IR in midlife.
Collapse
Affiliation(s)
- Siobhan M Hoscheidt
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Sara E Berman
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Leonardo A Rivera-Rivera
- 3 Department of Medical Physics, University of Wisconsin, Madison, WI, USA.,4 Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Rachel A Krause
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jennifer M Oh
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Michal S Beeri
- 5 The Joseph Sagol Neuroscience Center, Sheba Medical Center, Israel.,6 The Icahn School of Medicine, Mount Sinai, NY, USA
| | - Howard A Rowley
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,4 Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Oliver Wieben
- 3 Department of Medical Physics, University of Wisconsin, Madison, WI, USA.,4 Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Cynthia M Carlsson
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,7 Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA.,8 Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sanjay Asthana
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,7 Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA.,8 Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,7 Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA.,8 Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - William G Schrage
- 2 Department of Kinesiology, University of Wisconsin, Madison, WI, USA
| | - Barbara B Bendlin
- 1 Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,8 Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
21
|
Sorop O, Olver TD, van de Wouw J, Heinonen I, van Duin RW, Duncker DJ, Merkus D. The microcirculation: a key player in obesity-associated cardiovascular disease. Cardiovasc Res 2017; 113:1035-1045. [DOI: 10.1093/cvr/cvx093] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/04/2017] [Indexed: 12/11/2022] Open
|
22
|
Olver TD, McDonald MW, Klakotskaia D, Richardson RA, Jasperse JL, Melling CWJ, Schachtman TR, Yang HT, Emter CA, Laughlin MH. A chronic physical activity treatment in obese rats normalizes the contributions of ET-1 and NO to insulin-mediated posterior cerebral artery vasodilation. J Appl Physiol (1985) 2017; 122:1040-1050. [PMID: 28183819 DOI: 10.1152/japplphysiol.00811.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/11/2017] [Accepted: 02/02/2017] [Indexed: 12/31/2022] Open
Abstract
This study tested the hypotheses that obesity-induced decrements in insulin-stimulated cerebrovascular vasodilation would be normalized with acute endothelin-1a receptor antagonism and that treatment with a physical activity intervention restores vasoreactivity to insulin through augmented nitric oxide synthase (NOS)-dependent dilation. Otsuka Long-Evans Tokushima Fatty rats were divided into the following groups: 20 wk old food controlled (CON-20); 20 wk old free food access (model of obesity, OB-20); 40 wk old food controlled (CON-40); 40 wk old free food access (OB-40); and 40 wk old free food access+RUN (RUN-40; wheel-running access from 20 to 40 wk). Rats underwent Barnes maze testing and a euglycemic hyperinsulinemic clamp (EHC). In the 40-wk cohort, cerebellum and hippocampus blood flow (BF) were examined (microsphere infusion). Vasomotor responses (pressurized myography) to insulin were assessed in untreated, endothelin-1a receptor antagonism, and NOS inhibition conditions in posterior cerebral arteries. Insulin-stimulated vasodilation was attenuated in the OB vs. CON and RUN groups (P ≤ 0.04). Dilation to insulin was normalized with endothelin-1a receptor antagonism in the OB groups (between groups, P ≥ 0.56), and insulin-stimulated NOS-mediated dilation was greater in the RUN-40 vs. OB-40 group (P < 0.01). At 40 wk of age, cerebellum BF decreased during EHC in the OB-40 group (P = 0.02) but not CON or RUN groups (P ≥ 0.36). Barnes maze testing revealed increased entry errors and latencies in the RUN-40 vs. CON and OB groups (P < 0.01). These findings indicate that obesity-induced impairments in vasoreactivity to insulin involve increased endothelin-1 and decreased nitric oxide signaling. Chronic spontaneous physical activity, initiated after disease onset, reversed impaired vasodilation to insulin and decreased Barnes maze performance, possibly because of increased exploratory behavior.NEW & NOTEWORTHY The new and noteworthy findings are that 1) in rodents, obesity-related deficits in insulin-mediated vasodilation are associated with increased influence of insulin-stimulated ET-1 and depressed influence of insulin-stimulated NOS and 2) a physical activity intervention, initiated after the onset of disease, restores insulin-mediated vasodilation, likely by normalizing insulin-stimulated ET-1 and NOS balance. These data demonstrate that the treatment effects of chronic exercise on insulin-mediated vasodilation extend beyond active skeletal muscle vasculature and include the cerebrovasculature.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri;
| | - Matthew W McDonald
- Department of Kinesiology, University of Western Ontario, London, Ontario, Canada
| | - Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri; and
| | - Rachel A Richardson
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri; and
| | | | - C W James Melling
- Department of Kinesiology, University of Western Ontario, London, Ontario, Canada
| | - Todd R Schachtman
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri; and
| | - Hsiao T Yang
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
23
|
Toth P, Tarantini S, Csiszar A, Ungvari Z. Functional vascular contributions to cognitive impairment and dementia: mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am J Physiol Heart Circ Physiol 2017; 312:H1-H20. [PMID: 27793855 PMCID: PMC5283909 DOI: 10.1152/ajpheart.00581.2016] [Citation(s) in RCA: 337] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/10/2016] [Accepted: 10/26/2016] [Indexed: 12/16/2022]
Abstract
Increasing evidence from epidemiological, clinical and experimental studies indicate that age-related cerebromicrovascular dysfunction and microcirculatory damage play critical roles in the pathogenesis of many types of dementia in the elderly, including Alzheimer's disease. Understanding and targeting the age-related pathophysiological mechanisms that underlie vascular contributions to cognitive impairment and dementia (VCID) are expected to have a major role in preserving brain health in older individuals. Maintenance of cerebral perfusion, protecting the microcirculation from high pressure-induced damage and moment-to-moment adjustment of regional oxygen and nutrient supply to changes in demand are prerequisites for the prevention of cerebral ischemia and neuronal dysfunction. This overview discusses age-related alterations in three main regulatory paradigms involved in the regulation of cerebral blood flow (CBF): cerebral autoregulation/myogenic constriction, endothelium-dependent vasomotor function, and neurovascular coupling responses responsible for functional hyperemia. The pathophysiological consequences of cerebral microvascular dysregulation in aging are explored, including blood-brain barrier disruption, neuroinflammation, exacerbation of neurodegeneration, development of cerebral microhemorrhages, microvascular rarefaction, and ischemic neuronal dysfunction and damage. Due to the widespread attention that VCID has captured in recent years, the evidence for the causal role of cerebral microvascular dysregulation in cognitive decline is critically examined.
Collapse
Affiliation(s)
- Peter Toth
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Pecs, Hungary; and
| | - Stefano Tarantini
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Anna Csiszar
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma;
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
24
|
Mayhan WG, Arrick DM. Tetrahydrobiopterin rescues impaired responses of cerebral resistance arterioles during type 1 diabetes. Diab Vasc Dis Res 2017; 14:33-39. [PMID: 27941054 DOI: 10.1177/1479164116675490] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Our goal was to test the hypothesis that administration of tetrahydrobiopterin (BH4) would improve impaired endothelial nitric oxide synthase-dependent dilation of cerebral arterioles during type 1 diabetes. In addition, we examined the influence of BH4 on levels of superoxide in brain tissue. In vivo diameter of cerebral arterioles in nondiabetic and diabetic rats was measured in response to endothelial nitric oxide synthase-dependent agonists (acetylcholine and adenosine 5'-diphosphate) and an endothelial nitric oxide synthase-independent agonist (nitroglycerine) before and during application of BH4 (1.0 µM). We also measured levels of superoxide from cortex tissue in nondiabetic and diabetic rats under basal states and during BH4 Acetylcholine and adenosine 5'-diphosphate dilated cerebral arterioles in nondiabetic rats, but this vasodilation was significantly impaired in diabetic rats. In contrast, nitroglycerine produced similar vasodilation in nondiabetic and diabetic rats. Application of BH4 did not enhance vasodilation in nondiabetic rats but improved impaired cerebral vasodilation in diabetic rats. Basal superoxide levels were increased in cortex tissue from diabetic rats, and BH4 reduced these levels to that found in nondiabetic rats. Thus, BH4 is an important mediator of endothelial nitric oxide synthase-dependent responses of cerebral arterioles in diabetes and may have therapeutic potential for the treatment of cerebral vascular disease.
Collapse
Affiliation(s)
- William G Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Denise M Arrick
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
25
|
Hardigan T, Yasir A, Abdelsaid M, Coucha M, El-Shaffey S, Li W, Johnson MH, Ergul A. Linagliptin treatment improves cerebrovascular function and remodeling and restores reduced cerebral perfusion in Type 2 diabetes. Am J Physiol Regul Integr Comp Physiol 2016; 311:R466-77. [PMID: 27357799 DOI: 10.1152/ajpregu.00057.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022]
Abstract
The antihyperglycemic agent linagliptin, a dipeptidyl peptidase-4 (DPP-IV) inhibitor, has been shown to reduce inflammation and improve endothelial cell function. In this study, we hypothesized that DPP-IV inhibition with linagliptin would improve impaired cerebral perfusion in diabetic rats, as well as improve insulin-induced cerebrovascular relaxation and reverse pathological cerebrovascular remodeling. We further postulated that these changes would lead to a subsequent improvement of cognitive function. Male Type-2 diabetic and nondiabetic Goto-Kakizaki rats were treated with linagliptin for 4 wk, and blood glucose and DPP-IV plasma levels were assessed. Cerebral perfusion was assessed after treatment using laser-Doppler imaging, and dose response to insulin (10(-13) M-10(-6) M) in middle cerebral arteries was tested on a pressurized arteriograph. The impact of DPP-IV inhibition on diabetic cerebrovascular remodeling was assessed over a physiologically relevant pressure range, and changes in short-term hippocampus-dependent learning were observed using a novel object recognition test. Linagliptin lowered DPP-IV activity but did not change blood glucose or insulin levels in diabetes. Insulin-mediated vascular relaxation and cerebral perfusion were improved in the diabetic rats with linagliptin treatment. Indices of diabetic vascular remodeling, such as increased cross-sectional area, media thickness, and wall-to-lumen ratio, were also ameliorated; however, improvements in short-term hippocampal-dependent learning were not observed. The present study provides evidence that linagliptin treatment improves cerebrovascular dysfunction and remodeling in a Type 2 model of diabetes independent of glycemic control. This has important implications in diabetic patients who are predisposed to the development of cerebrovascular complications, such as stroke and cognitive impairment.
Collapse
Affiliation(s)
- Trevor Hardigan
- Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Abdul Yasir
- Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Mohammed Abdelsaid
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Maha Coucha
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Sally El-Shaffey
- Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Weiguo Li
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Maribeth H Johnson
- Department of Biostatistics, The Graduate School at Augusta University, Augusta, Georgia
| | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| |
Collapse
|
26
|
Costa ED, Rezende BA, Cortes SF, Lemos VS. Neuronal Nitric Oxide Synthase in Vascular Physiology and Diseases. Front Physiol 2016; 7:206. [PMID: 27313545 PMCID: PMC4889596 DOI: 10.3389/fphys.2016.00206] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/20/2016] [Indexed: 01/22/2023] Open
Abstract
The family of nitric oxide synthases (NOS) has significant importance in various physiological mechanisms and is also involved in many pathological processes. Three NOS isoforms have been identified: neuronal NOS (nNOS or NOS 1), endothelial NOS (eNOS or NOS 3), and an inducible NOS (iNOS or NOS 2). Both nNOS and eNOS are constitutively expressed. Classically, eNOS is considered the main isoform involved in the control of the vascular function. However, more recent studies have shown that nNOS is present in the vascular endothelium and importantly contributes to the maintenance of the homeostasis of the cardiovascular system. In physiological conditions, besides nitric oxide (NO), nNOS also produces hydrogen peroxide (H2O2) and superoxide ([Formula: see text]) considered as key mediators in non-neuronal cells signaling. This mini-review highlights recent scientific releases on the role of nNOS in vascular homeostasis and cardiovascular disorders such as hypertension and atherosclerosis.
Collapse
Affiliation(s)
- Eduardo D Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Bruno A Rezende
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas GeraisBelo Horizonte, Brazil; Department of Health Sciences, Post-graduate Institute, Medical Sciences CollegeBelo Horizonte, Brazil
| | - Steyner F Cortes
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Virginia S Lemos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| |
Collapse
|
27
|
Katakam PVG, Dutta S, Sure VN, Grovenburg SM, Gordon AO, Peterson NR, Rutkai I, Busija DW. Depolarization of mitochondria in neurons promotes activation of nitric oxide synthase and generation of nitric oxide. Am J Physiol Heart Circ Physiol 2016; 310:H1097-106. [PMID: 26945078 DOI: 10.1152/ajpheart.00759.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/07/2016] [Indexed: 11/22/2022]
Abstract
The diverse signaling events following mitochondrial depolarization in neurons are not clear. We examined for the first time the effects of mitochondrial depolarization on mitochondrial function, intracellular calcium, neuronal nitric oxide synthase (nNOS) activation, and nitric oxide (NO) production in cultured neurons and perivascular nerves. Cultured rat primary cortical neurons were studied on 7-10 days in vitro, and endothelium-denuded cerebral arteries of adult Sprague-Dawley rats were studied ex vivo. Diazoxide and BMS-191095 (BMS), activators of mitochondrial KATP channels, depolarized mitochondria in cultured neurons and increased cytosolic calcium levels. However, the mitochondrial oxygen consumption rate was unaffected by mitochondrial depolarization. In addition, diazoxide and BMS not only increased the nNOS phosphorylation at positive regulatory serine 1417 but also decreased nNOS phosphorylation at negative regulatory serine 847. Furthermore, diazoxide and BMS increased NO production in cultured neurons measured with both fluorescence microscopy and electron spin resonance spectroscopy, which was sensitive to inhibition by the selective nNOS inhibitor 7-nitroindazole (7-NI). Diazoxide also protected cultured neurons against oxygen-glucose deprivation, which was blocked by NOS inhibition and rescued by NO donors. Finally, BMS induced vasodilation of endothelium denuded, freshly isolated cerebral arteries that was diminished by 7-NI and tetrodotoxin. Thus pharmacological depolarization of mitochondria promotes activation of nNOS leading to generation of NO in cultured neurons and endothelium-denuded arteries. Mitochondrial-induced NO production leads to increased cellular resistance to lethal stress by cultured neurons and to vasodilation of denuded cerebral arteries.
Collapse
Affiliation(s)
- Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| | - Somhrita Dutta
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| | - Samuel M Grovenburg
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| | - Angellica O Gordon
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| | - Nicholas R Peterson
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| | - Ibolya Rutkai
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| | - David W Busija
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
28
|
Li C, Jiang Z, Lu W, Arrick D, McCarter K, Sun H. Effect of obesity on early blood–brain barrier disruption following transient focal cerebral ischemia. Obes Sci Pract 2016. [DOI: 10.1002/osp4.30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- C. Li
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences Center‐Shreveport Shreveport LA USA
| | - Z. Jiang
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences Center‐Shreveport Shreveport LA USA
| | - W. Lu
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences Center‐Shreveport Shreveport LA USA
| | - D. Arrick
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences Center‐Shreveport Shreveport LA USA
| | - K. McCarter
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences Center‐Shreveport Shreveport LA USA
| | - H. Sun
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences Center‐Shreveport Shreveport LA USA
| |
Collapse
|
29
|
Merdzo I, Rutkai I, Tokes T, Sure VNLR, Katakam PVG, Busija DW. The mitochondrial function of the cerebral vasculature in insulin-resistant Zucker obese rats. Am J Physiol Heart Circ Physiol 2016; 310:H830-8. [PMID: 26873973 DOI: 10.1152/ajpheart.00964.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/03/2016] [Indexed: 12/24/2022]
Abstract
Little is known about mitochondrial functioning in the cerebral vasculature during insulin resistance (IR). We examined mitochondrial respiration in isolated cerebral arteries of male Zucker obese (ZO) rats and phenotypically normal Zucker lean (ZL) rats using the Seahorse XFe24 analyzer. We investigated mitochondrial morphology in cerebral blood vessels as well as mitochondrial and nonmitochondrial protein expression levels in cerebral arteries and microvessels. We also measured reactive oxygen species (ROS) levels in cerebral microvessels. Under basal conditions, the mitochondrial respiration components (nonmitochondrial respiration, basal respiration, ATP production, proton leak, and spare respiratory capacity) showed similar levels among the ZL and ZO groups with the exception of maximal respiration, which was higher in the ZO group. We examined the role of nitric oxide by measuring mitochondrial respiration following inhibition of nitric oxide synthase with N(ω)-nitro-l-arginine methyl ester (l-NAME) and mitochondrial activation after administration of diazoxide (DZ). Both ZL and ZO groups showed similar responses to these stimuli with minor variations.l-NAME significantly increased the proton leak, and DZ decreased nonmitochondrial respiration in the ZL group. Other components were not affected. Mitochondrial morphology and distribution within vascular smooth muscle and endothelium as well as mitochondrial protein levels were similar in the arteries and microvessels of both groups. Endothelial nitric oxide synthase (eNOS) and ROS levels were increased in cerebral microvessels of the ZO. Our study suggests that mitochondrial function is not significantly altered in the cerebral vasculature of young ZO rats, but increased ROS production might be due to increased eNOS in the cerebral microcirculation during IR.
Collapse
Affiliation(s)
- Ivan Merdzo
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ibolya Rutkai
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Tunde Tokes
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Venkata N L R Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - David W Busija
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
30
|
Saad MI, Abdelkhalek TM, Saleh MM, Kamel MA, Youssef M, Tawfik SH, Dominguez H. Insights into the molecular mechanisms of diabetes-induced endothelial dysfunction: focus on oxidative stress and endothelial progenitor cells. Endocrine 2015; 50:537-67. [PMID: 26271514 DOI: 10.1007/s12020-015-0709-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/25/2015] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus is a heterogeneous, multifactorial, chronic disease characterized by hyperglycemia owing to insulin insufficiency and insulin resistance (IR). Recent epidemiological studies showed that the diabetes epidemic affects 382 million people worldwide in 2013, and this figure is expected to be 600 million people by 2035. Diabetes is associated with microvascular and macrovascular complications resulting in accelerated endothelial dysfunction (ED), atherosclerosis, and cardiovascular disease (CVD). Unfortunately, the complex pathophysiology of diabetic cardiovascular damage is not fully understood. Therefore, there is a clear need to better understand the molecular pathophysiology of ED in diabetes, and consequently, better treatment options and novel efficacious therapies could be identified. In the light of recent extensive research, we re-investigate the association between diabetes-associated metabolic disturbances (IR, subclinical inflammation, dyslipidemia, hyperglycemia, dysregulated production of adipokines, defective incretin and gut hormones production/action, and oxidative stress) and ED, focusing on oxidative stress and endothelial progenitor cells (EPCs). In addition, we re-emphasize that oxidative stress is the final common pathway that transduces signals from other conditions-either directly or indirectly-leading to ED and CVD.
Collapse
Affiliation(s)
- Mohamed I Saad
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt.
- Hudson Institute of Medical Research, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia.
| | - Taha M Abdelkhalek
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Moustafa M Saleh
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Mina Youssef
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Shady H Tawfik
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | - Helena Dominguez
- Department of Biomedical Sciences, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
31
|
Katakam PVG, Gordon AO, Sure VNLR, Rutkai I, Busija DW. Diversity of mitochondria-dependent dilator mechanisms in vascular smooth muscle of cerebral arteries from normal and insulin-resistant rats. Am J Physiol Heart Circ Physiol 2015; 307:H493-503. [PMID: 24929852 DOI: 10.1152/ajpheart.00091.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitochondrial depolarization following ATP-sensitive potassium (mitoKATP) channel activation has been shown to induce cerebral vasodilation by generation of mitochondrial reactive oxygen species (ROS), which sequentially promotes frequency of calcium sparks and activation of large conductance calcium-activated potassium channels (BKCa) in vascular smooth muscle (VSM). We previously demonstrated that cerebrovascular insulin resistance accompanies aging and obesity. It is unclear whether mitochondrial depolarization without the ROS generation enhances calcium sparks and vasodilation in phenotypically normal [Sprague Dawley (SD); Zucker lean (ZL)] and insulin-resistant [Zucker obese (ZO)] rats. We compared the mechanisms underlying the vasodilation to ROS-dependent (diazoxide) and ROS-independent [BMS-191095 (BMS)] mitoKATP channel activators in normal and ZO rats. Arterial diameter studies from SD, ZL, and ZO rats showed that BMS as well as diazoxide induced vasodilation in endothelium-denuded cerebral arteries. In normal rats, BMS-induced vasodilation was mediated by mitochondrial depolarization and calcium sparks generation in VSM and was reduced by inhibition of BKCa channels. However, unlike diazoxide-induced vasodilation, scavenging of ROS had no effect on BMS-induced vasodilation. Electron spin resonance spectroscopy confirmed that diazoxide but not BMS promoted vascular ROS generation. BMS- as well as diazoxide-induced vasodilation, mitochondrial depolarization, and calcium spark generation were diminished in cerebral arteries from ZO rats. Thus pharmacological depolarization of VSM mitochondria by BMS promotes ROS-independent vasodilation via generation of calcium sparks and activation of BKCa channels. Diminished generation of calcium sparks and reduced vasodilation in ZO arteries in response to BMS and diazoxide provide new insights into mechanisms of cerebrovascular dysfunction in insulin resistance.
Collapse
|
32
|
Dorrance AM, Matin N, Pires PW. The effects of obesity on the cerebral vasculature. Curr Vasc Pharmacol 2015; 12:462-72. [PMID: 24846235 DOI: 10.2174/1570161112666140423222411] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 04/08/2013] [Accepted: 05/01/2013] [Indexed: 12/18/2022]
Abstract
The incidence of obesity in the population is increasing at an alarming rate, with this comes an increased risk of insulin resistance (IR). Obesity and IR increase an individual's risk of having a stroke and they have been linked to several forms of dementia. Stroke and dementia are associated with, or exacerbated by, reduced cerebral blood flow, which has recently been described in obese patients. In this review we will discuss the effects of obesity on cerebral artery function and structure. Regarding their function, we will focus on the endothelium and nitric oxide (NO) dependent dilation. NO dependent dilation is impaired in cerebral arteries from obese rats, and the majority of evidence suggests this is a result of increased oxidative stress. We will also describe the limited studies showing that inward cerebral artery remodeling occurs in models of obesity, and that the remodeling is associated with an increase in the damage caused by cerebral ischemia. We will also discuss some of the more paradoxical findings associated with stroke and obesity, including the evidence that obesity is a positive factor for stroke survival. Finally we will discuss the evidence that links these changes in vascular structure and function to cognitive decline and dementia.
Collapse
Affiliation(s)
| | | | - Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, East Lansing, MI 48824, USA.
| |
Collapse
|
33
|
Lucke-Wold BP, Turner RC, Logsdon AF, Simpkins JW, Alkon DL, Smith KE, Chen YW, Tan Z, Huber JD, Rosen CL. Common mechanisms of Alzheimer's disease and ischemic stroke: the role of protein kinase C in the progression of age-related neurodegeneration. J Alzheimers Dis 2015; 43:711-24. [PMID: 25114088 PMCID: PMC4446718 DOI: 10.3233/jad-141422] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ischemic stroke and Alzheimer's disease (AD), despite being distinct disease entities, share numerous pathophysiological mechanisms such as those mediated by inflammation, immune exhaustion, and neurovascular unit compromise. An important shared mechanistic link is acute and chronic changes in protein kinase C (PKC) activity. PKC isoforms have widespread functions important for memory, blood-brain barrier maintenance, and injury repair that change as the body ages. Disease states accelerate PKC functional modifications. Mutated forms of PKC can contribute to neurodegeneration and cognitive decline. In some cases the PKC isoforms are still functional but are not successfully translocated to appropriate locations within the cell. The deficits in proper PKC translocation worsen stroke outcome and amyloid-β toxicity. Cross talk between the innate immune system and PKC pathways contribute to the vascular status within the aging brain. Unfortunately, comorbidities such as diabetes, obesity, and hypertension disrupt normal communication between the two systems. The focus of this review is to highlight what is known about PKC function, how isoforms of PKC change with age, and what additional alterations are consequences of stroke and AD. The goal is to highlight future therapeutic targets that can be applied to both the treatment and prevention of neurologic disease. Although the pathology of ischemic stroke and AD are different, the similarity in PKC responses warrants further investigation, especially as PKC-dependent events may serve as an important connection linking age-related brain injury.
Collapse
Affiliation(s)
- Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Aric F. Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - James W. Simpkins
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Daniel L. Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, USA
| | - Kelly E. Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Yi-Wen Chen
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jason D. Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Correspondence to: Charles L. Rosen, MD, PhD, Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV 26506-9183, USA. Tel.: +1 304 293 5041; Fax: +1 304 293 4819;
| |
Collapse
|
34
|
Frisbee JC, Goodwill AG, Frisbee SJ, Butcher JT, Brock RW, Olfert IM, DeVallance ER, Chantler PD. Distinct temporal phases of microvascular rarefaction in skeletal muscle of obese Zucker rats. Am J Physiol Heart Circ Physiol 2014; 307:H1714-28. [PMID: 25305181 DOI: 10.1152/ajpheart.00605.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Evolution of metabolic syndrome is associated with a progressive reduction in skeletal muscle microvessel density, known as rarefaction. Although contributing to impairments to mass transport and exchange, the temporal development of rarefaction and the contributing mechanisms that lead to microvessel loss are both unclear and critical areas for investigation. Although previous work suggests that rarefaction severity in obese Zucker rats (OZR) is predicted by the chronic loss of vascular nitric oxide (NO) bioavailability, we have determined that this hides a biphasic development of rarefaction, with both early and late components. Although the total extent of rarefaction was well predicted by the loss in NO bioavailability, the early pulse of rarefaction developed before a loss of NO bioavailability and was associated with altered venular function (increased leukocyte adhesion/rolling), and early elevation in oxidant stress, TNF-α levels, and the vascular production of thromboxane A2 (TxA2). Chronic inhibition of TNF-α blunted the severity of rarefaction and also reduced vascular oxidant stress and TxA2 production. Chronic blockade of the actions of TxA2 also blunted rarefaction, but did not impact oxidant stress or inflammation, suggesting that TxA2 is a downstream outcome of elevated reactive oxygen species and inflammation. If chronic blockade of TxA2 is terminated, microvascular rarefaction in OZR skeletal muscle resumes, but at a reduced rate despite low NO bioavailability. These results suggest that therapeutic interventions against inflammation and TxA2 under conditions where metabolic syndrome severity is moderate or mild may prevent the development of a condition of accelerated microvessel loss with metabolic syndrome.
Collapse
Affiliation(s)
- Jefferson C Frisbee
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - Adam G Goodwill
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - Stephanie J Frisbee
- Department of Health Policy, Management and Leadership, West Virginia University Health Sciences Center, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - Joshua T Butcher
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - Robert W Brock
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - I Mark Olfert
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, West Virginia; and Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - Evan R DeVallance
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, West Virginia; and Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | - Paul D Chantler
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, West Virginia; and Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center, Morgantown, West Virginia
| |
Collapse
|
35
|
Wu KL, Chao YM, Tsay SJ, Chen CH, Chan SH, Dovinova I, Chan JY. Role of Nitric Oxide Synthase Uncoupling at Rostral Ventrolateral Medulla in Redox-Sensitive Hypertension Associated With Metabolic Syndrome. Hypertension 2014; 64:815-24. [DOI: 10.1161/hypertensionaha.114.03777] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metabolic syndrome (MetS), which is rapidly becoming prevalent worldwide, is long known to be associated with hypertension and recently with oxidative stress. Of note is that oxidative stress in the rostral ventrolateral medulla (RVLM), where sympathetic premotor neurons reside, contributes to sympathoexcitation and hypertension. This study sought to identify the source of tissue oxidative stress in RVLM and their roles in neural mechanism of hypertension associated with MetS. Adult normotensive rats subjected to a high-fructose diet for 8 weeks developed metabolic traits of MetS, alongside increases in sympathetic vasomotor activity and blood pressure. In RVLM of these MetS rats, the tissue level of reactive oxygen species was increased, nitric oxide (NO) was decreased, and mitochondrial electron transport capacity was reduced. Whereas the protein expression of neuronal NO synthase (nNOS) or protein inhibitor of nNOS was increased, the ratio of nNOS dimer/monomer was significantly decreased. Oral intake of pioglitazone or intracisternal infusion of tempol or coenzyme Q
10
significantly abrogated all those molecular events in high-fructose diet–fed rats and ameliorated sympathoexcitation and hypertension. Gene silencing of protein inhibitor of nNOS mRNA in RVLM using lentivirus carrying small hairpin RNA inhibited protein inhibitor of nNOS expression, increased the ratio of nNOS dimer/monomer, restored NO content, and alleviated oxidative stress in RVLM of high-fructose diet–fed rats, alongside significantly reduced sympathoexcitation and hypertension. These results suggest that redox-sensitive and protein inhibitor of nNOS–mediated nNOS uncoupling is engaged in a vicious cycle that sustains the production of reactive oxygen species in RVLM, resulting in sympathoexcitation and hypertension associated with MetS.
Collapse
Affiliation(s)
- Kay L.H. Wu
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (K.L.H.W., Y.M.C., S.H.H.C., J.Y.H.C.); Institute of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan (S.J.T.); Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan (C.H.C.); and Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia (I.D.)
| | - Yung-Mei Chao
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (K.L.H.W., Y.M.C., S.H.H.C., J.Y.H.C.); Institute of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan (S.J.T.); Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan (C.H.C.); and Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia (I.D.)
| | - Shiow-Jen Tsay
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (K.L.H.W., Y.M.C., S.H.H.C., J.Y.H.C.); Institute of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan (S.J.T.); Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan (C.H.C.); and Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia (I.D.)
| | - Chen Hsiu Chen
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (K.L.H.W., Y.M.C., S.H.H.C., J.Y.H.C.); Institute of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan (S.J.T.); Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan (C.H.C.); and Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia (I.D.)
| | - Samuel H.H. Chan
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (K.L.H.W., Y.M.C., S.H.H.C., J.Y.H.C.); Institute of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan (S.J.T.); Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan (C.H.C.); and Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia (I.D.)
| | - Ima Dovinova
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (K.L.H.W., Y.M.C., S.H.H.C., J.Y.H.C.); Institute of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan (S.J.T.); Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan (C.H.C.); and Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia (I.D.)
| | - Julie Y.H. Chan
- From the Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan (K.L.H.W., Y.M.C., S.H.H.C., J.Y.H.C.); Institute of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan (S.J.T.); Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan (C.H.C.); and Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovakia (I.D.)
| |
Collapse
|
36
|
Bhatt MP, Lim YC, Ha KS. C-peptide replacement therapy as an emerging strategy for preventing diabetic vasculopathy. Cardiovasc Res 2014; 104:234-44. [PMID: 25239825 DOI: 10.1093/cvr/cvu211] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Lack of C-peptide, along with insulin, is the main feature of Type 1 diabetes mellitus (DM) and is also observed in progressive β-cell loss in later stage of Type 2 DM. Therapeutic approaches to hyperglycaemic control have been ineffective in preventing diabetic vasculopathy, and alternative therapeutic strategies are necessary to target both hyperglycaemia and diabetic complications. End-stage organ failure in DM seems to develop primarily due to vascular dysfunction and damage, leading to two types of organ-specific diseases, such as micro- and macrovascular complications. Numerous studies in diabetic patients and animals demonstrate that C-peptide treatment alone or in combination with insulin has physiological functions and might be beneficial in preventing diabetic complications. Current evidence suggests that C-peptide replacement therapy might prevent and ameliorate diabetic vasculopathy and organ-specific complications through conservation of vascular function, as well as prevention of endothelial cell death, microvascular permeability, vascular inflammation, and neointima formation. In this review, we describe recent advances on the beneficial role of C-peptide replacement therapy for preventing diabetic complications, such as retinopathy, nephropathy, neuropathy, impaired wound healing, and inflammation, and further discuss potential beneficial effects of combined C-peptide and insulin supplement therapy to control hyperglycaemia and to prevent organ-specific complications.
Collapse
Affiliation(s)
- Mahendra Prasad Bhatt
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Kangwondaehak-gil 1, Chuncheon, Kangwon-do 200-701, Republic of Korea
| | - Young-Cheol Lim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Kangwondaehak-gil 1, Chuncheon, Kangwon-do 200-701, Republic of Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Kangwondaehak-gil 1, Chuncheon, Kangwon-do 200-701, Republic of Korea
| |
Collapse
|
37
|
Yan LJ. Pathogenesis of chronic hyperglycemia: from reductive stress to oxidative stress. J Diabetes Res 2014; 2014:137919. [PMID: 25019091 PMCID: PMC4082845 DOI: 10.1155/2014/137919] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/27/2014] [Indexed: 02/08/2023] Open
Abstract
Chronic overnutrition creates chronic hyperglycemia that can gradually induce insulin resistance and insulin secretion impairment. These disorders, if not intervened, will eventually be followed by appearance of frank diabetes. The mechanisms of this chronic pathogenic process are complex but have been suggested to involve production of reactive oxygen species (ROS) and oxidative stress. In this review, I highlight evidence that reductive stress imposed by overflux of NADH through the mitochondrial electron transport chain is the source of oxidative stress, which is based on establishments that more NADH recycling by mitochondrial complex I leads to more electron leakage and thus more ROS production. The elevated levels of both NADH and ROS can inhibit and inactivate glyceraldehyde 3-phosphate dehydrogenase (GAPDH), respectively, resulting in blockage of the glycolytic pathway and accumulation of glycerol 3-phospate and its prior metabolites along the pathway. This accumulation then initiates all those alternative glucose metabolic pathways such as the polyol pathway and the advanced glycation pathways that otherwise are minor and insignificant under euglycemic conditions. Importantly, all these alternative pathways lead to ROS production, thus aggravating cellular oxidative stress. Therefore, reductive stress followed by oxidative stress comprises a major mechanism of hyperglycemia-induced metabolic syndrome.
Collapse
Affiliation(s)
- Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, RES-314E, Fort Worth, TX 76107, USA
- *Liang-Jun Yan:
| |
Collapse
|
38
|
Prevention of hyperglycemic signal pathways in metabolic syndrome carotid artery of rats. Transl Stroke Res 2013; 3:466-72. [PMID: 24323833 DOI: 10.1007/s12975-012-0205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 07/10/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
Abstract
Obesity is the major risk factor for metabolic syndrome and atherosclerotic cardiocerebrovascular diseases and induces insulin resistance characterized by a dysfunction of insulin to activate insulin receptor /insulin receptor substrate 1(IRS-1)/phosphoinositide 3-kinase (PI3K)/Akt pathway. Zucker fatty rats (8 weeks) were treated with vehicle (0.5 % methyl cellulose in physiological saline, p.o.), amlodipine (3 mg/kg/day, p.o.), atorvastatin (10 mg/kg/day, p.o.), or the combination of amlodipine plus atorvastatin (3 + 10 mg/kg/day, p.o.) for 28 days, and anti-insulin-like growth factor 1 (IGF-1)/IRS-1/PI3K/Akt pathways were evaluated. Our present immunohistochemical study first demonstrated that a combination of amlodipine plus atorvastatin treatment prevented an arteriosclerotic process compared to the single treatment with amlodipine or atorvastatin with strong recoveries of pTyr IRS-1, pPI3K, and pAkt expressions and with remarkable restraints of IGF-1 and pSer IRS-1. As a result, combination therapy with amlodipine plus atorvastatin showed a strong synergistic effect to prevent atherosclerotic processes. The present study newly suggests a synergistic benefit of combination therapy with amlodipine plus atorvastatin for strong prevention of atherosclerotic processes, which could reduce the clinical risk of cerebrovascular events for obesity patients.
Collapse
|
39
|
Bagi Z, Feher A, Dou H, Broskova Z. Selective up-regulation of arginase-1 in coronary arteries of diabetic patients. Front Immunol 2013; 4:293. [PMID: 24133491 PMCID: PMC3783852 DOI: 10.3389/fimmu.2013.00293] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 09/06/2013] [Indexed: 01/06/2023] Open
Abstract
Coronary artery disease (CAD) remains the leading cause of death in the Western societies. Diabetes mellitus (DM) is one of the highly prevalent diseases, which remarkably accelerates the development of CAD. Experimental evidence indicates that decreased bioavailability of coronary endothelial nitric oxide (NO) contributes to the development of CAD in DM. There are recent studies showing that a selective impairment of NO synthesis occurs in coronary arteries of DM patients, which is mainly due to the limited availability of endothelial NO synthase (eNOS) precursor, l-arginine. Importantly, these studies demonstrated that DM, independent of the presence of CAD, leads to selective up-regulation of arginase-1. Arginase-1 seems to play an important role in limiting l-arginine availability in the close proximity of eNOS in vessels of DM patients. This brief review examines recent clinical studies demonstrating the pathological role of vascular arginase-1 in human diabetes. Whether arginase-1, which is crucial in the synthesis of various fundamental polyamines in the body, will represent a potent therapeutic target for prevention of DM-associated CAD is still debated.
Collapse
Affiliation(s)
- Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University , Augusta, GA , USA
| | | | | | | |
Collapse
|
40
|
Turner RC, Lucke-Wold B, Lucke-Wold N, Elliott AS, Logsdon AF, Rosen CL, Huber JD. Neuroprotection for ischemic stroke: moving past shortcomings and identifying promising directions. Int J Mol Sci 2013; 14:1890-917. [PMID: 23344061 PMCID: PMC3565354 DOI: 10.3390/ijms14011890] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/04/2013] [Accepted: 01/10/2013] [Indexed: 02/05/2023] Open
Abstract
The translation of neuroprotective agents for ischemic stroke from bench-to-bedside has largely failed to produce improved treatments since the development of tissue plasminogen activator (tPA). One possible reason for lack of translation is the failure to acknowledge the greatest risk factor for stroke, age, and other common comorbidities such as hypertension, obesity, and diabetes that are associated with stroke. In this review, we highlight both mechanisms of studying these factors and results of those that have been addressed. We also discuss the potential role of other lifestyle factors associated with an increased stroke risk such as sleep fragmentation and/or deprivation. Furthermore, many proposed therapeutic agents have targeted molecular mechanisms occurring soon after the onset of ischemia despite data indicating delayed patient presentation following ischemic stroke. Modulating inflammation has been identified as a promising therapeutic avenue consistent with preliminary success of ongoing clinical trials for anti-inflammatory compounds such as minocycline. We review the role of inflammation in stroke and in particular, the role of inflammatory cell recruitment and macrophage phenotype in the inflammatory process. Emerging evidence indicates an increasing role of neuro-immune crosstalk, which has led to increased interest in identification of peripheral biomarkers indicative of neural injury. It is our hope that identification and investigation of factors influencing stroke pathophysiology may lead to improved therapeutics.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
| | - Brandon Lucke-Wold
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
| | - Noelle Lucke-Wold
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
- Department of Health Restoration, West Virginia University School of Nursing, Morgantown, WV 26506, USA
| | - Alisa S. Elliott
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
| | - Aric F. Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA
| | - Charles L. Rosen
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-304-293-5041; Fax: +1-304-293-4819
| | - Jason D. Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA
| |
Collapse
|
41
|
Sánchez A, Contreras C, Martínez MP, Climent B, Benedito S, García-Sacristán A, Hernández M, Prieto D. Role of neural NO synthase (nNOS) uncoupling in the dysfunctional nitrergic vasorelaxation of penile arteries from insulin-resistant obese Zucker rats. PLoS One 2012; 7:e36027. [PMID: 22540017 PMCID: PMC3335073 DOI: 10.1371/journal.pone.0036027] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 03/28/2012] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Erectile dysfunction (ED) is considered as an early sign of vascular disease due to its high prevalence in patients with cardiovascular risk factors. Endothelial and neural dysfunction involving nitric oxide (NO) are usually implicated in the pathophysiology of the diabetic ED, but the underlying mechanisms are unclear. The present study assessed the role of oxidative stress in the dysfunctional neural vasodilator responses of penile arteries in the obese Zucker rat (OZR), an experimental model of metabolic syndrome/prediabetes. METHODS AND RESULTS Electrical field stimulation (EFS) under non-adrenergic non-cholinergic (NANC) conditions evoked relaxations that were significantly reduced in penile arteries of OZR compared with those of lean Zucker rats (LZR). Blockade of NO synthase (NOS) inhibited neural relaxations in both LZR and OZR, while saturating concentrations of the NOS substrate L-arginine reversed the inhibition and restored relaxations in OZR to levels in arteries from LZR. nNOS expression was unchanged in arteries from OZR compared to LZR and nNOS selective inhibition decreased the EFS relaxations in LZR but not in OZR, while endothelium removal did not alter these responses in either strain. Superoxide anion production and nitro-tyrosine immunostaining were elevated in the erectile tissue from OZR. Treatment with the NADPH oxidase inhibitor apocynin or acute incubation with the NOS cofactor tetrahydrobiopterin (BH4) restored neural relaxations in OZR to levels in control arteries, while inhibition of the enzyme of BH4 synthesis GTP-cyclohydrolase (GCH) reduced neural relaxations in arteries from LZR but not OZR. The NO donor SNAP induced decreases in intracellular calcium that were impaired in arteries from OZR compared to controls. CONCLUSIONS The present study demonstrates nitrergic dysfunction and impaired neural NO signalling due to oxidative stress and nNOS uncoupling in penile arteries under conditions of insulin resistance. This dysfunction likely contributes to the metabolic syndrome-associated ED, along with the endothelial dysfunction also involving altered NO signalling.
Collapse
Affiliation(s)
- Ana Sánchez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Contreras
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - María Pilar Martínez
- Departamento de Anatomía y Anatomía Patológica Comparadas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Belén Climent
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Sara Benedito
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Albino García-Sacristán
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Medardo Hernández
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|