1
|
Addisu D, Mitiku Y, Yazie Ferede W, Mekuriaw BY, Erega BB, Bazezew LY, Belachew TW, Tadesse SG, Goshu YA, Mengistie BA, Mekie M, Misker AD, Mihretie GN. Determinants of new-onset postpartum preeclampsia among mothers who delivered in hospitals in the South Gondar Zone, Northwest Ethiopia: a multicenter case-control study. BMC Pregnancy Childbirth 2025; 25:161. [PMID: 39953424 PMCID: PMC11829449 DOI: 10.1186/s12884-025-07274-z] [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: 12/25/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND New-onset postpartum preeclampsia has emerging as a significant public health concern in Ethiopia, with a notable increase in incidence in the study area. While substantial research exists on antepartum preeclampsia, data on the determinants of new-onset postpartum preeclampsia are limited. Therefore, this study aimed to investigate determinants of new-onset postpartum preeclampsia among mothers who gave birth at hospitals in the South Gondar Zone, Northwest Ethiopia. METHODS An unmatched case-control study was conducted with 355 postpartum mothers (89 cases and 266 controls). A multistage sampling method was used to select the study participants. Data were collected using structured questionnaires and medical chart reviews, and analyzed using SPSS Version 27. Binary logistic regression (bivariable and multivariable analysis) was used to identify factors associated with new-onset postpartum preeclampsia. Statistical significance was set at p < 0.05, and strength of associations were measured using adjusted odds ratios (AOR) with 95% confidence intervals (CIs). RESULT New-onset postpartum preeclampsia was significantly associated with several factors, including advanced maternal age, fewer antenatal care (ANC) visits, contraceptive use, physical inactivity, a history of multiple gestations, and gestational diabetes mellitus. Specifically, maternal age ≥ 40 years (AOR = 11.63, 95% CI: 4.24-31.86), fewer than four ANC visits (AOR = 8.45, 95% CI: 3.96-18.05), contraceptive use (AOR = 4.04, 95% CI: 1.26-13.37), irregular physical activity (AOR = 4.05, 95% CI: 1.32-12.44), physical inactivity (AOR = 8.25, 95% CI: 4.62-19.29), a history of multiple gestations (AOR = 2.66, 95% CI: 1.26-5.60), and gestational diabetes mellitus (AOR = 17.79, 95% CI: 7.72-40.95) were identified as key determinants. CONCLUSIONS Advanced maternal age, fewer ANC visits, contraceptive use, physical inactivity, multiple gestations, and gestational diabetes were strongly associated with new-onset postpartum preeclampsia. Increased ANC visits and postpartum monitoring are essential for early detection and management of postpartum preeclampsia. Additionally, promoting physical activity should be incorporated into maternal health strategies to reduce the incidence of new onset postpartum preeclampsia.
Collapse
Affiliation(s)
- Dagne Addisu
- Department of Midwifery, Debre Tabor University, Debre Tabor, Ethiopia.
| | - Yekaba Mitiku
- Department of Midwifery, Debre Tabor Comprehensive Specialized Hospital, Debre Tabor, Ethiopia
| | | | | | | | - Lakachew Yismaw Bazezew
- Department of Pediatrics and Neonatal Nursing, Debre Tabor University, Debre Tabor, Ethiopia
| | | | | | | | | | - Maru Mekie
- Department of Midwifery, Debre Tabor University, Debre Tabor, Ethiopia
| | | | | |
Collapse
|
2
|
Rocca MS, Pannella M, Bayraktar E, Marino S, Bortolozzi M, Di Nisio A, Foresta C, Ferlin A. Extragonadal function of follicle-stimulating hormone: Evidence for a role in endothelial physiology and dysfunction. Mol Cell Endocrinol 2024; 594:112378. [PMID: 39332467 DOI: 10.1016/j.mce.2024.112378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
AIMS Follicle-stimulating hormone (FSH) plays a fundamental role in reproduction stimulating ovarian folliculogenesis, Sertoli cells function and spermatogenesis. However, the recent identification of FSH receptor (FSHR) also in extra-gonadal tissues has suggested that FSH activity may not be limited only to fertility regulation, with conflicting results on the possible role of FSH in endothelial cells. The aim of this study was to investigate FSH role on endothelial function in Human Umbilical Vein Endothelial Cells (HUVECs). RESULTS Endothelial Nitric oxide synthase (eNOS) expression, eNOS phosphorylation and Nitric Oxide (NO) production resulted increased after the stimulation of HUVEC with recombinant human FSH (rhFSH) at 3.6x103 ng/ml, with increasing Calcium release from intracellular stores. Furthermore, IP3 production increased after rhFSH stimulation despite PTX treatment and NFAT1 was observed prevalently in nucleus. We observed a statistical difference between untreated cells and cells stimulated with 0.36x103 ng/ml and between cells stimulated with 0.36x103 ng/ml and cells stimulated with 1.8x103 ng/ml at 4 and 8 h by Wound healing assay, respectively. Furthermore, a higher cellular permeability was observed in stimulated cells, with atypical VE-cadherin distribution, as well as filamentous actin. CONCLUSIONS Our findings suggest that FSH at high concentrations elicits a signalling that could compromise the endothelial membrane. Indeed, VE-cadherin anomalies may severely affect the endothelial barrier, resulting in an increased membrane permeability. Although NO is an important vasodilatation factor, probably an excessive production could impact on endothelial functionality, partially explaining the increased risk of cardiovascular diseases in menopausal women and men with hypogonadism.
Collapse
Affiliation(s)
- Maria Santa Rocca
- University Hospital of Padua, Unit of Andrology and Reproductive Medicine, Padua, Italy
| | | | - Erva Bayraktar
- University of Padua, Department of Physics and Astronomy "G. Galilei", Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129, Padua, Italy
| | - Saralea Marino
- University of Padua, Department of Physics and Astronomy "G. Galilei", Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129, Padua, Italy
| | - Mario Bortolozzi
- University of Padua, Department of Physics and Astronomy "G. Galilei", Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Via Orus 2, 35129, Padua, Italy
| | - Andrea Di Nisio
- University of Padua, Department of Medicine, Padua, Italy; Department of Wellbeing, Nutrition and Sport, Pegaso Telematic University, Centro Direzionale Isola F2, Naples, Italy
| | - Carlo Foresta
- University of Padua, Department of Medicine, Padua, Italy
| | - Alberto Ferlin
- University Hospital of Padua, Unit of Andrology and Reproductive Medicine, Padua, Italy; University of Padua, Department of Medicine, Padua, Italy.
| |
Collapse
|
3
|
Catella J, Turpin E, Connes P, Nader E, Carin R, Martin M, Rezigue H, Nougier C, Dargaud Y, Josset‐Lamaugarny A, Dugrain J, Marano M, Leuci A, Boisson C, Renoux C, Joly P, Poutrel S, Hot A, Guillot N, Fromy B. Impaired microvascular function in patients with sickle cell anemia and leg ulcers improved with healing. Br J Haematol 2024; 205:2459-2469. [PMID: 39318045 PMCID: PMC11637740 DOI: 10.1111/bjh.19785] [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: 07/13/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024]
Abstract
Leg Ulcer (LU) pathophysiology is still not well understood in sickle cell anaemia (SCA). We hypothesised that SCA patients with LU would be characterised by lower microvascular reactivity. The aim of the present study was to compare the microcirculatory function (transcutaneous oxygen pressure (TcPO2) on the foot and laser Doppler flowmetry on the arm) and several blood biological parameters between nine SCA patients with active LU (LU+) and 56 SCA patients with no positive history of LU (LU-). We also tested the effects of plasma from LU+ and LU- patients on endothelial cell activation. We observed a reduction of the TcPO2 in LU+ compared to LU- patients. In addition, LU+ patients exhibited lower cutaneous microvascular vasodilatory capacity in response to acetylcholine, current and local heating compared to LU- patients. Inflammation and endothelial cell activation in response to plasma did not differ between the two groups. Among the nine patients from the LU+ group, eight were followed and six achieved healing in 4.4 ± 2.5 months. Among thus achieving healing, microvascular vasodilatory capacity in response to acetylcholine, current and local heating and TcPO2 improved after healing. In conclusion, microcirculatory function is impaired in patients with LU, and improves with healing.
Collapse
Affiliation(s)
- Judith Catella
- Service de Médecine Interne et VasculaireHôpital Edouard Herriot, Hospices Civils de LyonLyonFrance
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Etienne Turpin
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Philippe Connes
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Elie Nader
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Romain Carin
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Marie Martin
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Hamdi Rezigue
- Service d'hématologie‐hémostaseHospices civils de LyonBronFrance
- EA 4609‐Hémostase et cancer, UFR LaennecUniversité Claude BernardLyon 1France
| | - Christophe Nougier
- Service d'hématologie‐hémostaseHospices civils de LyonBronFrance
- EA 4609‐Hémostase et cancer, UFR LaennecUniversité Claude BernardLyon 1France
| | - Yesim Dargaud
- EA 4609‐Hémostase et cancer, UFR LaennecUniversité Claude BernardLyon 1France
- Unité d'hémostase CliniqueHôpital Cardiologique Louis PradelLyonFrance
| | - Audrey Josset‐Lamaugarny
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305)CNRS/Université Claude Bernard Lyon 1LyonFrance
| | - Justine Dugrain
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305)CNRS/Université Claude Bernard Lyon 1LyonFrance
| | - Muriel Marano
- EA 4609‐Hémostase et cancer, UFR LaennecUniversité Claude BernardLyon 1France
| | - Alexandre Leuci
- EA 4609‐Hémostase et cancer, UFR LaennecUniversité Claude BernardLyon 1France
| | - Camille Boisson
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi‐siteHospices Civils de LyonLyonFrance
| | - Celine Renoux
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi‐siteHospices Civils de LyonLyonFrance
| | - Philippe Joly
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
- Service de Biochimie et Biologie Moléculaire, Laboratoire de Biologie Médicale Multi‐siteHospices Civils de LyonLyonFrance
| | - Solène Poutrel
- Service de Médecine Interne et VasculaireHôpital Edouard Herriot, Hospices Civils de LyonLyonFrance
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Arnaud Hot
- Service de Médecine Interne et VasculaireHôpital Edouard Herriot, Hospices Civils de LyonLyonFrance
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Nicolas Guillot
- Laboratoire d'Excellence du Globule Rouge (Labex GR‐Ex), SorbonneParisFrance
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe «Biologie Vasculaire et du Globule Rouge»Université Claude Bernard Lyon 1, Université de LyonLyonFrance
| | - Berengère Fromy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305)CNRS/Université Claude Bernard Lyon 1LyonFrance
| |
Collapse
|
4
|
Baranauskas MN, Blechschmid TH, Long EB, Coggan AR, Carter SJ. Dietary NO 3- does not enhance endothelial dependent cutaneous vascular conductance in older women. Microvasc Res 2024; 155:104706. [PMID: 38871050 DOI: 10.1016/j.mvr.2024.104706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/24/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024]
Abstract
Prior work has yet to determine whether the reduction of dietary nitrate (NO3-) to NO, via the enterosalivary pathway, may modify cutaneous vascular conductance (CVC) responses to local heating in older women. Changes occurring with the transition to menopause related to hormonal flux, increased adiposity, and/or decreased physical activity may further compound the negative influence of aging on nitric oxide (NO)-dependent CVC. Herein, we characterized changes in NO-dependent CVC following acute ingestion of 140 mL of NO3--rich beetroot juice in 24 older women (age: 65 ± 5 y, BMI: 31.2 ± 3.7 kg/m2). Red blood cell (RBC) flux was measured continuously via laser-Doppler flowmetry on the dorsal aspect of the forearm during local skin heating to 39 °C/44 °C before and 3 h after NO3- ingestion. NO-dependent changes in CVC were calculated as RBC flux/mean arterial blood pressure at 39 °C and normalized as a proportion of maximal CVC at 44 °C (%CVCmax). Changes (Δ) in fractional exhaled NO (FeNO) following NO3- ingestion were used an index of NO bioavailability. Despite increased FeNO (+81 ± 70 %, P < 0.001), %CVCmax at 39 °C was reduced (-16 ± 10 %, P < 0.001) following NO3- ingestion. A greater reduction in %CVCmax was weakly to moderately associated with higher body fat% (r = 0.45 [0.05-0.72], P = 0.029), central adiposity% (r = 0.50 [0.13-0.75], P = 0.012), neutrophil% (r = 0.42 [0.02-0.70], P = 0.041), and higher neutrophil to lymphocyte ratio (r = 0.49 [0.11-0.75], P = 0.016). These findings demonstrate a single dose of dietary NO3- does not promote CVC responses to local heating in sedentary older women with overweight and obesity. Correlation with multiple biomarkers suggest systemic inflammation may be involved.
Collapse
Affiliation(s)
- Marissa N Baranauskas
- Department of Human Physiology & Nutrition, University of Colorado, Colorado Springs, CO, United States of America.
| | - Tyler H Blechschmid
- Department of Kinesiology, School of Public Health, Bloomington, IN, United States of America
| | - Emily B Long
- Department of Kinesiology, School of Public Health, Bloomington, IN, United States of America
| | - Andrew R Coggan
- Department of Kinesiology, Indiana University Indianapolis, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Stephen J Carter
- Department of Kinesiology, School of Public Health, Bloomington, IN, United States of America
| |
Collapse
|
5
|
Parmaksiz D, Kim Y. Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories. Neuroscientist 2024:10738584241268754. [PMID: 39113465 DOI: 10.1177/10738584241268754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Complex mechanisms govern the transport and action of oxytocin (Oxt), a neuropeptide and hormone that mediates diverse physiologic processes. While Oxt exerts site-specific and rapid effects in the brain via axonal and somatodendritic release, volume transmission via CSF and the neurovascular interface can act as an additional mechanism to distribute Oxt signals across distant brain regions on a slower timescale. This review focuses on modes of Oxt transport and action in the CNS, with particular emphasis on the roles of perivascular spaces, the blood-brain barrier (BBB), and circumventricular organs in coordinating the triadic interaction among circulating blood, CSF, and parenchyma. Perivascular spaces, critical conduits for CSF flow, play a pivotal role in Oxt diffusion and distribution within the CNS and reciprocally undergo Oxt-mediated structural and functional reconstruction. While the BBB modulates the movement of Oxt between systemic and cerebral circulation in a majority of brain regions, circumventricular organs without a functional BBB can allow for diffusion, monitoring, and feedback regulation of bloodborne peripheral signals such as Oxt. Recognition of these additional transport mechanisms provides enhanced insight into the systemic propagation and regulation of Oxt activity.
Collapse
Affiliation(s)
- Deniz Parmaksiz
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
6
|
Martin LS, Josset-Lamaugarny A, El Jammal T, Ducreux S, Chevalier FP, Fromy B. Aging is associated with impaired triggering of TRPV3-mediated cutaneous vasodilation: a crucial process for local heat exposure. GeroScience 2024; 46:3567-3580. [PMID: 37855862 PMCID: PMC11226586 DOI: 10.1007/s11357-023-00981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Sensing temperature is vitally important to adapt our body to environmental changes. Local warm detection is required to initiate regulation of cutaneous blood flow, which is part of the peripheral thermoregulatory mechanisms, and thus avoid damage to surrounding tissues. The mechanisms mediating cutaneous vasodilation during local heat stress are impaired with aging. However, the impact of aging on the ability of the skin to detect subtle thermal changes is unknown. Among heat-activated cation channels, transient receptor potential vanilloid 3 (TRPV3) is a thermo-sensor predominantly expressed on keratinocytes and involved in local vascular thermoregulatory mechanisms of the skin in young mice. In the present study, using a murine in vivo model of local heat exposure of the skin, we showed that heat-induced vasodilation was reduced in old mice associated with reduced expression of TRPV3 channels. We also found a decrease in expression and activity of TRPV3 channel, as well as reduced TRPV3-dependent adenosine tri-phosphate release in human primary keratinocytes from old donors. This study shows that aging alters the epidermal TRPV3 channels, which might delay the detection of changes in skin temperature, thereby limiting the mechanisms triggered for local vascular thermoregulation in the old skin.
Collapse
Affiliation(s)
- Lisa S Martin
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69007, Lyon, France
- Claude Bernard University Lyon 1, 69100, Villeurbanne, France
| | - Audrey Josset-Lamaugarny
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69007, Lyon, France
- Claude Bernard University Lyon 1, 69100, Villeurbanne, France
| | - Thomas El Jammal
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69007, Lyon, France
- Claude Bernard University Lyon 1, 69100, Villeurbanne, France
- Department of Internal Medicine, University Hospital Lyon Croix-Rousse, Claude Bernard University Lyon 1, Lyon, France
| | - Sylvie Ducreux
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, Claude Bernard University Lyon 1, 69500, Bron, France
| | - Fabien P Chevalier
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69007, Lyon, France
- Claude Bernard University Lyon 1, 69100, Villeurbanne, France
| | - Bérengère Fromy
- CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69007, Lyon, France.
- Claude Bernard University Lyon 1, 69100, Villeurbanne, France.
| |
Collapse
|
7
|
Collins HE, Alexander BT, Care AS, Davenport MH, Davidge ST, Eghbali M, Giussani DA, Hoes MF, Julian CG, LaVoie HA, Olfert IM, Ozanne SE, Bytautiene Prewit E, Warrington JP, Zhang L, Goulopoulou S. Guidelines for assessing maternal cardiovascular physiology during pregnancy and postpartum. Am J Physiol Heart Circ Physiol 2024; 327:H191-H220. [PMID: 38758127 PMCID: PMC11380979 DOI: 10.1152/ajpheart.00055.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Maternal mortality rates are at an all-time high across the world and are set to increase in subsequent years. Cardiovascular disease is the leading cause of death during pregnancy and postpartum, especially in the United States. Therefore, understanding the physiological changes in the cardiovascular system during normal pregnancy is necessary to understand disease-related pathology. Significant systemic and cardiovascular physiological changes occur during pregnancy that are essential for supporting the maternal-fetal dyad. The physiological impact of pregnancy on the cardiovascular system has been examined in both experimental animal models and in humans. However, there is a continued need in this field of study to provide increased rigor and reproducibility. Therefore, these guidelines aim to provide information regarding best practices and recommendations to accurately and rigorously measure cardiovascular physiology during normal and cardiovascular disease-complicated pregnancies in human and animal models.
Collapse
Grants
- HL169157 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HD088590 NICHD NIH HHS
- HD083132 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- The Biotechnology and Biological Sciences Research Council
- P20GM103499 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- British Heart Foundation (BHF)
- R21 HD111908 NICHD NIH HHS
- Distinguished University Professor
- The Lister Insititute
- ES032920 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- Canadian Insitute's of Health Research Foundation Grant
- HL149608 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Royal Society (The Royal Society)
- U.S. Department of Defense (DOD)
- HL138181 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- MC_00014/4 UKRI | Medical Research Council (MRC)
- RG/17/8/32924 British Heart Foundation
- Jewish Heritage Fund for Excellence
- HD111908 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- APP2002129 NHMRC Ideas Grant
- HL159865 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL131182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NS103017 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL138181 NHLBI NIH HHS
- 20CSA35320107 American Heart Association (AHA)
- RG/17/12/33167 British Heart Foundation (BHF)
- National Heart Foundation Future Leader Fellowship
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL155295 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HD088590-06 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- WVU SOM Synergy Grant
- R01 HL146562 NHLBI NIH HHS
- R01 HL159865 NHLBI NIH HHS
- HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- ES034646-01 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL150472 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 2021T017 Dutch Heart Foundation Dekker Grant
- MC_UU_00014/4 Medical Research Council
- R01 HL163003 NHLBI NIH HHS
- Christenson professor In Active Healthy Living
- National Heart Foundation
- Dutch Heart Foundation Dekker
- WVU SOM Synergy
- Jewish Heritage
- Department of Health | National Health and Medical Research Council (NHMRC)
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Helen E Collins
- University of Louisville, Louisville, Kentucky, United States
| | - Barbara T Alexander
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alison S Care
- University of Adelaide, Adelaide, South Australia, Australia
| | | | | | - Mansoureh Eghbali
- University of California Los Angeles, Los Angeles, California, United States
| | | | | | - Colleen G Julian
- University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Holly A LaVoie
- University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - I Mark Olfert
- West Virginia University School of Medicine, Morgantown, West Virginia, United States
| | | | | | - Junie P Warrington
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Lubo Zhang
- Loma Linda University School of Medicine, Loma Linda, California, United States
| | | |
Collapse
|
8
|
Mahoney SA, Clayton ZS. Reply to Jaster. Am J Physiol Heart Circ Physiol 2024; 327:H000. [PMID: 38958689 DOI: 10.1152/ajpheart.00354.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024]
Affiliation(s)
- Sophia A Mahoney
- University of Colorado Boulder, Boulder, Colorado, United States
| | - Zachary S Clayton
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
9
|
Darvish S, Mahoney SA, Venkatasubramanian R, Rossman MJ, Clayton ZS, Murray KO. Socioeconomic status as a potential mediator of arterial aging in marginalized ethnic and racial groups: current understandings and future directions. J Appl Physiol (1985) 2024; 137:194-222. [PMID: 38813611 PMCID: PMC11389897 DOI: 10.1152/japplphysiol.00188.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 05/31/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the United States. However, disparities in CVD-related morbidity and mortality exist as marginalized racial and ethnic groups are generally at higher risk for CVDs (Black Americans, Indigenous People, South and Southeast Asians, Native Hawaiians, and Pacific Islanders) and/or development of traditional CVD risk factors (groups above plus Hispanics/Latinos) relative to non-Hispanic Whites (NHW). In this comprehensive review, we outline emerging evidence suggesting these groups experience accelerated arterial dysfunction, including vascular endothelial dysfunction and large elastic artery stiffening, a nontraditional CVD risk factor that may predict risk of CVDs in these groups with advancing age. Adverse exposures to social determinants of health (SDOH), specifically lower socioeconomic status (SES), are exacerbated in most of these groups (except South Asians-higher SES) and may be a potential mediator of accelerated arterial aging. SES negatively influences the ability of marginalized racial and ethnic groups to meet aerobic exercise guidelines, the first-line strategy to improve arterial function, due to increased barriers, such as time and financial constraints, lack of motivation, facility access, and health education, to performing conventional aerobic exercise. Thus, identifying alternative interventions to conventional aerobic exercise that 1) overcome these common barriers and 2) target the biological mechanisms of aging to improve arterial function may be an effective, alternative method to aerobic exercise to ameliorate accelerated arterial aging and reduce CVD risk. Importantly, dedicated efforts are needed to assess these strategies in randomized-controlled clinical trials in these marginalized racial and ethnic groups.
Collapse
Affiliation(s)
- Sanna Darvish
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | | | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Kevin O Murray
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
10
|
Blagov AV, Summerhill VI, Sukhorukov VN, Zhigmitova EB, Postnov AY, Orekhov AN. Potential use of antioxidants for the treatment of chronic inflammatory diseases. Front Pharmacol 2024; 15:1378335. [PMID: 38818374 PMCID: PMC11137403 DOI: 10.3389/fphar.2024.1378335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
The excessive production of various reactive oxidant species over endogenous antioxidant defense mechanisms leads to the development of a state of oxidative stress, with serious biological consequences. The consequences of oxidative stress depend on the balance between the generation of reactive oxidant species and the antioxidant defense and include oxidative damage of biomolecules, disruption of signal transduction, mutation, and cell apoptosis. Accumulating evidence suggests that oxidative stress is involved in the physiopathology of various debilitating illnesses associated with chronic inflammation, including cardiovascular diseases, diabetes, cancer, or neurodegenerative processes, that need continuous pharmacological treatment. Oxidative stress and chronic inflammation are tightly linked pathophysiological processes, one of which can be simply promoted by another. Although, many antioxidant trials have been unsuccessful (some of the trials showed either no effect or even harmful effects) in human patients as a preventive or curative measure, targeting oxidative stress remains an interesting therapeutic approach for the development of new agents to design novel anti-inflammatory drugs with a reliable safety profile. In this regard, several natural antioxidant compounds were explored as potential therapeutic options for the treatment of chronic inflammatory diseases. Several metalloenzymes, such as superoxide dismutase, catalase, and glutathione peroxidase, are among the essential enzymes that maintain the low nanomolar physiological concentrations of superoxide (O2•-) and hydrogen peroxide (H2O2), the major redox signaling molecules, and thus play important roles in the alteration of the redox homeostasis. These enzymes have become a striking source of motivation to design catalytic drugs to enhance the action of these enzymes under pathological conditions related to chronic inflammation. This review is focused on several major representatives of natural and synthetic antioxidants as potential drug candidates for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | | | - Anton Y. Postnov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - Alexander N. Orekhov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Institute for Atherosclerosis Research, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| |
Collapse
|
11
|
Zhang H, Muhetarijiang M, Chen RJ, Hu X, Han J, Zheng L, Chen T. Mitochondrial Dysfunction: A Roadmap for Understanding and Tackling Cardiovascular Aging. Aging Dis 2024:AD.2024.0058. [PMID: 38739929 DOI: 10.14336/ad.2024.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
Cardiovascular aging is a progressive remodeling process constituting a variety of cellular and molecular alterations that are closely linked to mitochondrial dysfunction. Therefore, gaining a deeper understanding of the changes in mitochondrial function during cardiovascular aging is crucial for preventing cardiovascular diseases. Cardiac aging is accompanied by fibrosis, cardiomyocyte hypertrophy, metabolic changes, and infiltration of immune cells, collectively contributing to the overall remodeling of the heart. Similarly, during vascular aging, there is a profound remodeling of blood vessel structure. These remodeling present damage to endothelial cells, increased vascular stiffness, impaired formation of new blood vessels (angiogenesis), the development of arteriosclerosis, and chronic vascular inflammation. This review underscores the role of mitochondrial dysfunction in cardiac aging, exploring its impact on fibrosis and myocardial alterations, metabolic remodeling, immune response remodeling, as well as in vascular aging in the heart. Additionally, we emphasize the significance of mitochondria-targeted therapies in preventing cardiovascular diseases in the elderly.
Collapse
Affiliation(s)
- Han Zhang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mairedan Muhetarijiang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ryan J Chen
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaosheng Hu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Han
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liangrong Zheng
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
12
|
Mahoney SA, VanDongen NS, Greenberg NT, Venkatasubramanian R, Rossman MJ, Widlansky ME, Brunt VE, Bernaldo de Quirós Y, Seals DR, Clayton ZS. Role of the circulating milieu in age-related arterial dysfunction: a novel ex vivo approach. Am J Physiol Heart Circ Physiol 2024; 326:H1279-H1290. [PMID: 38517225 PMCID: PMC11380963 DOI: 10.1152/ajpheart.00014.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
The circulating milieu, bioactive molecules in the bloodstream, is altered with aging and interfaces constantly with the vasculature. This anatomic juxtaposition suggests that circulating factors may actively modulate arterial function. Here, we developed a novel, translational experimental model that allows for direct interrogation of the influence of the circulating milieu on age-related arterial dysfunction (aortic stiffening and endothelial dysfunction). To do so, we exposed young and old mouse arteries to serum from young and old mice and young and midlife/older (ML/O) adult humans. We found that old mouse and ML/O adult human, but not young, serum stiffened young mouse aortic rings, assessed via elastic modulus (mouse and human serum, P = 0.003 vs. young serum control), and impaired carotid artery endothelial function, assessed by endothelium-dependent dilation (EDD) (mouse serum, P < 0.001; human serum, P = 0.006 vs. young serum control). Furthermore, young mouse and human, but not old, serum reduced aortic elastic modulus (mouse serum, P = 0.009; human serum, P < 0.001 vs. old/MLO serum control) and improved EDD (mouse and human serum, P = 0.015 vs. old/MLO serum control) in old arteries. In human serum-exposed arteries, in vivo arterial function assessed in the human donors correlated with circulating milieu-modulated arterial function in young mouse arteries (aortic stiffness, r = 0.634, P = 0.005; endothelial function, r = 0.609, P = 0.004) and old mouse arteries (aortic stiffness, r = 0.664, P = 0.001; endothelial function, r = 0.637, P = 0.003). This study establishes novel experimental approaches for directly assessing the effects of the circulating milieu on arterial function and implicates changes in the circulating milieu as a mechanism of in vivo arterial aging.NEW & NOTEWORTHY Changes in the circulating milieu with advancing age may be a mechanism underlying age-related arterial dysfunction. Ex vivo exposure of young mouse arteries to the circulating milieu from old mice or midlife/older adults impairs arterial function whereas exposure of old mouse arteries to the circulating milieu from young mice or young adults improves arterial function. These findings establish that the circulating milieu directly influences arterial function with aging.
Collapse
Affiliation(s)
- Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nicholas S VanDongen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nathan T Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | | | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Michael E Widlansky
- Department of Medicine and Pharmacology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Yara Bernaldo de Quirós
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
- Institute of Animal Health and Food Safety, Universidad de Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
13
|
Coelho-Junior HJ, Calvani R, Tosato M, Russo A, Landi F, Picca A, Marzetti E. Associations between hypertension and cognitive, mood, and behavioral parameters in very old adults: results from the IlSIRENTE study. Front Public Health 2024; 11:1268983. [PMID: 38533244 PMCID: PMC10964923 DOI: 10.3389/fpubh.2023.1268983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/11/2023] [Indexed: 03/28/2024] Open
Abstract
Introduction Studies on the associations between hypertension-related parameters and cognitive function, mood, and behavioral symptoms in older adults have produced mixed findings. A possible explanation for these divergent results is that investigations have not adequately adjusted their analyses according to the use of angiotensin-converting enzyme inhibitors (ACEIs). Therefore, the present study examined the cross-sectional associations between hypertension-related parameters, ACEI use, and cognitive function, mood, and behavioral symptoms in very old adults. Methods This study was conducted by analyzing the IlSIRENTE database, a prospective cohort study that collected data on all individuals aged 80 years and older residing in the Sirente geographic area (n = 364). Blood pressure (BP) was assessed after 20 to 40 min of rest, while participants sat in an upright position. Drugs were coded according to the Anatomical Therapeutic and Chemical codes. Cognitive function, mood, and behavioral symptoms were recorded using the Minimum Data Set Home Care instrument. Blood inflammatory markers were measured. Results Hypertension-related parameters were significantly associated with many cognitive, mood, and behavioral parameters after adjustment for covariates. However, only the inverse association between hypertension and lesser problems with short-term memory remained significant. Participants with hypertension had lower blood concentrations of inflammatory markers in comparison to their normotensive peers. Conclusion Findings from the present study indicate that high BP values are associated with fewer complaints about memory problems in very old adults. Furthermore, a lower concentration of inflammatory markers was found in hypertensive participants. ACEI use might affect this scenario.
Collapse
Affiliation(s)
- Helio José Coelho-Junior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Matteo Tosato
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Andrea Russo
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| |
Collapse
|
14
|
Mahoney SA, Venkatasubramanian R, Darrah MA, Ludwig KR, VanDongen NS, Greenberg NT, Longtine AG, Hutton DA, Brunt VE, Campisi J, Melov S, Seals DR, Rossman MJ, Clayton ZS. Intermittent supplementation with fisetin improves arterial function in old mice by decreasing cellular senescence. Aging Cell 2024; 23:e14060. [PMID: 38062873 PMCID: PMC10928570 DOI: 10.1111/acel.14060] [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: 07/19/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 01/17/2024] Open
Abstract
Cellular senescence and the senescence-associated secretory phenotype (SASP) contribute to age-related arterial dysfunction, in part, by promoting oxidative stress and inflammation, which reduce the bioavailability of the vasodilatory molecule nitric oxide (NO). In the present study, we assessed the efficacy of fisetin, a natural compound, as a senolytic to reduce vascular cell senescence and SASP factors and improve arterial function in old mice. We found that fisetin decreased cellular senescence in human endothelial cell culture. In old mice, vascular cell senescence and SASP-related inflammation were lower 1 week after the final dose of oral intermittent (1 week on-2 weeks off-1 weeks on dosing) fisetin supplementation. Old fisetin-supplemented mice had higher endothelial function. Leveraging old p16-3MR mice, a transgenic model allowing genetic clearance of p16INK4A -positive senescent cells, we found that ex vivo removal of senescent cells from arteries isolated from vehicle- but not fisetin-treated mice increased endothelium-dependent dilation, demonstrating that fisetin improved endothelial function through senolysis. Enhanced endothelial function with fisetin was mediated by increased NO bioavailability and reduced cellular- and mitochondrial-related oxidative stress. Arterial stiffness was lower in fisetin-treated mice. Ex vivo genetic senolysis in aorta rings from p16-3MR mice did not further reduce mechanical wall stiffness in fisetin-treated mice, demonstrating lower arterial stiffness after fisetin was due to senolysis. Lower arterial stiffness with fisetin was accompanied by favorable arterial wall remodeling. The findings from this study identify fisetin as promising therapy for clinical translation to target excess cell senescence to treat age-related arterial dysfunction.
Collapse
Affiliation(s)
- Sophia A. Mahoney
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | | | - Mary A. Darrah
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Katelyn R. Ludwig
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Nicholas S. VanDongen
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Nathan T. Greenberg
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Abigail G. Longtine
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - David A. Hutton
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Vienna E. Brunt
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Judith Campisi
- Buck Institute for Research on AgingNovatoCaliforniaUSA
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Simon Melov
- Buck Institute for Research on AgingNovatoCaliforniaUSA
| | - Douglas R. Seals
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Matthew J. Rossman
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Zachary S. Clayton
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| |
Collapse
|
15
|
El-Awaisi J, Kavanagh DPJ, Kalia N. Monitoring coronary blood flow by laser speckle contrast imaging after myocardial ischaemia reperfusion injury in adult and aged mice. Front Cardiovasc Med 2024; 11:1358472. [PMID: 38410244 PMCID: PMC10895051 DOI: 10.3389/fcvm.2024.1358472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/19/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction Investigating coronary microvascular perfusion responses after myocardial infarction (MI) would aid in the development of flow preserving therapies. Laser speckle contrast imaging (LSCI) is a powerful tool used for real-time, non-contact, full-field imaging of blood flow in various tissues/organs. However, its use in the beating heart has been limited due to motion artifacts. Methods In this paper, we report the novel use of LSCI, combined with custom speckle analysis software (SpAn), to visualise and quantitate changes in ventricular perfusion in adult and aged mice undergoing ischaemia-reperfusion (IR) injury. The therapeutic benefit of inhibiting the actions of the pro-inflammatory cytokine interleukin-36 (IL-36) was also investigated using an IL-36 receptor antagonist (IL-36Ra). Results Imaging from uncovered and covered regions of the left ventricle demonstrated that whilst part of the LSCI flux signal was derived from beating motion, a significant contributor to the flux signal came from ventricular microcirculatory blood flow. We show that a biphasic flux profile corresponding to diastolic and systolic phases of the cardiac cycle can be detected without mathematically processing the total flux data to denoise motion artifacts. Furthermore, perfusion responses to ischaemia and postischaemia were strong, reproducible and could easily be detected without the need to subtract motion-related flux signals. LSCI also identified significantly poorer ventricular perfusion in injured aged mice following IR injury which markedly improved with IL-36Ra. Discussion We therefore propose that LSCI of the heart is possible despite motion artifacts and may facilitate future investigations into the role of the coronary microcirculation in cardiovascular diseases and development of novel therapies.
Collapse
Affiliation(s)
| | | | - Neena Kalia
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
16
|
Siervo M, Hussin AM, Calella P, Ashor A, Shannon OM, Mendes I, Stephan BC, Zheng D, Hill T, Mathers JC. Associations between Aging and Vitamin D Status with Whole-Body Nitric Oxide Production and Markers of Endothelial Function. J Nutr 2024; 154:469-478. [PMID: 38048992 DOI: 10.1016/j.tjnut.2023.12.002] [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: 10/17/2023] [Revised: 11/23/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Aging and vitamin D deficiency have been associated with reduced nitric oxide (NO) synthesis and impaired endothelial function (EF) but the evidence in humans remains weak. OBJECTIVES Two independent cross-sectional studies were designed to evaluate the association between age, sex, and plasma vitamin D concentrations with physiological and biochemical biomarkers of NO synthesis and EF in young and older healthy participants (Study 1) and in overweight and obese postmenopausal females (Study 2). METHODS In Study 1, 40 young (20-49 y) and older (50-75 y) males and females (10 participants per age and sex group) were included. Resting blood pressure and ear-to-finger peripheral pulse wave velocity (PWV) were measured. A stable-isotopic method was used to determine whole-body NO production. Plasma 25-hydroxyvitamin D (25(OH)D), nitrate, nitrite, and asymmetric dimethylarginine (ADMA) concentrations were determined. In Study 2, 80 older overweight and obese females (age 61.2 ± 6.2 y, body mass index 29.5 ± 4.4 kg/m2) were recruited. Postocclusion reactive hyperemia (PORH) and peripheral PWV were measured. Plasma concentrations of 25(OH)D, nitrate, cyclic guanosine monophosphate, 3-nitrotyrosine (3-NT), endothelin-1, vascular endothelial growth factor, and ADMA were determined. RESULTS In Study 1, whole-body NO production was significantly greater in young compared with older participants (0.61 ± 0.30 μmol·h-1·kg-1 compared with 0.39 ± 0.10 μmol·h-1·kg-1, P = 0.01) but there was no evidence of a sex difference (P = 0.81). Plasma 25(OH)D concentration was not associated with PWV (r = 0.18, P = 0.28) or whole-body NO production (r = -0.20, P = 0.22). Plasma ADMA concentration was associated positively with age (r = 0.35, P = 0.03) and negatively with whole-body NO production (r = -0.33, P = 0.04). In Study 2, age was associated with lower PORH (r = -0.28, P = 0.02) and greater ADMA concentrations (r = 0.22, P = 0.04). Plasma 25(OH)D concentration was inversely associated with 3-NT concentrations (r = -0.31, P = 0.004). CONCLUSIONS Older age was associated with lower whole-body NO production. Plasma vitamin D concentrations were not associated with NO production or markers of EF but showed a weak, significant correlation with oxidative stress in postmenopausal overweight females.
Collapse
Affiliation(s)
- Mario Siervo
- School of Population Health, Curtin University, Perth, WA, Australia; Curtin Dementia Centre of Excellence, Enable Institute, Curtin University, Perth, WA, Australia.
| | - Azizah Mat Hussin
- Human Nutrition & Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle, United Kingdom; Institute of Medical Science Technology-Universiti Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Patrizia Calella
- Department of Movement Sciences and Wellbeing, University of Naples "Parthenope," Naples, Italy
| | - Ammar Ashor
- Department of Internal Medicine, College of Medicine, University of Al-Mustansiriyah, Baghdad, Iraq
| | - Oliver M Shannon
- Human Nutrition & Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Ines Mendes
- Human Nutrition & Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Blossom Cm Stephan
- Curtin Dementia Centre of Excellence, Enable Institute, Curtin University, Perth, WA, Australia
| | - Dingchang Zheng
- Research Centre for Intelligent Healthcare, Coventry University, Coventry, United Kingdom
| | - Tom Hill
- Human Nutrition & Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle, United Kingdom
| | - John C Mathers
- Human Nutrition & Exercise Research Centre, Centre for Healthier Lives, Population Health Sciences Institute, Newcastle University, Newcastle, United Kingdom
| |
Collapse
|
17
|
Cavero-Redondo I, Saz-Lara A, Martínez-García I, Otero-Luis I, Martínez-Rodrigo A. Validation of an early vascular aging construct model for comprehensive cardiovascular risk assessment using external risk indicators for improved clinical utility: data from the EVasCu study. Cardiovasc Diabetol 2024; 23:33. [PMID: 38218806 PMCID: PMC10787504 DOI: 10.1186/s12933-023-02104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) remain a major global health concern, necessitating advanced risk assessment beyond traditional factors. Early vascular aging (EVA), characterized by accelerated vascular changes, has gained importance in cardiovascular risk assessment. METHODS The EVasCu study in Spain examined 390 healthy participants using noninvasive measurements. A construct of four variables (Pulse Pressure, Pulse Wave Velocity, Glycated Hemoglobin, Advanced Glycation End Products) was used for clustering. K-means clustering with principal component analysis revealed two clusters, healthy vascular aging (HVA) and early vascular aging (EVA). External validation variables included sociodemographic, adiposity, glycemic, inflammatory, lipid profile, vascular, and blood pressure factors. RESULTS EVA cluster participants were older and exhibited higher adiposity, poorer glycemic control, dyslipidemia, altered vascular properties, and higher blood pressure. Significant differences were observed for age, smoking status, body mass index, waist circumference, fat percentage, glucose, insulin, C-reactive protein, diabetes prevalence, lipid profiles, arterial stiffness, and blood pressure levels. These findings demonstrate the association between traditional cardiovascular risk factors and EVA. CONCLUSIONS This study validates a clustering model for EVA and highlights its association with established risk factors. EVA assessment can be integrated into clinical practice, allowing early intervention and personalized cardiovascular risk management.
Collapse
Affiliation(s)
- Iván Cavero-Redondo
- Health and Social Research Center, University of Castilla-La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Alicia Saz-Lara
- Health and Social Research Center, University of Castilla-La Mancha, Cuenca, Spain.
| | | | - Iris Otero-Luis
- Health and Social Research Center, University of Castilla-La Mancha, Cuenca, Spain
| | - Arturo Martínez-Rodrigo
- Research Group in Electronic, Biomedical, and Telecommunication Engineering, University of Castilla-La Mancha, Cuenca, Spain
| |
Collapse
|
18
|
Mahoney SA, Dey AK, Basisty N, Herman AB. Identification and functional analysis of senescent cells in the cardiovascular system using omics approaches. Am J Physiol Heart Circ Physiol 2023; 325:H1039-H1058. [PMID: 37656130 PMCID: PMC10908411 DOI: 10.1152/ajpheart.00352.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide, and senescent cells have emerged as key contributors to its pathogenesis. Senescent cells exhibit cell cycle arrest and secrete a range of proinflammatory factors, termed the senescence-associated secretory phenotype (SASP), which promotes tissue dysfunction and exacerbates CVD progression. Omics technologies, specifically transcriptomics and proteomics, offer powerful tools to uncover and define the molecular signatures of senescent cells in cardiovascular tissue. By analyzing the comprehensive molecular profiles of senescent cells, omics approaches can identify specific genetic alterations, gene expression patterns, protein abundances, and metabolite levels associated with senescence in CVD. These omics-based discoveries provide insights into the mechanisms underlying senescence-induced cardiovascular damage, facilitating the development of novel diagnostic biomarkers and therapeutic targets. Furthermore, integration of multiple omics data sets enables a systems-level understanding of senescence in CVD, paving the way for precision medicine approaches to prevent or treat cardiovascular aging and its associated complications.
Collapse
Affiliation(s)
- Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, Colorado, United States
| | - Amit K Dey
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Nathan Basisty
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Allison B Herman
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| |
Collapse
|
19
|
Shiwakoti S, Gong D, Sharma K, Kang KW, Schini-Kerth VB, Kim HJ, Ko JY, Oak MH. γ-Oryzanol ameliorates fine dust-induced premature endothelial senescence and dysfunction via attenuating oxidative stress. Food Chem Toxicol 2023; 179:113981. [PMID: 37549806 DOI: 10.1016/j.fct.2023.113981] [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: 05/11/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Various cardiovascular diseases are associated with endothelial senescence, and a recent study showed that fine dust (FD)-induced premature endothelial senescence and dysfunction is associated with increased oxidative stress. The aim of the present study was to investigate protective effect of rice bran extract (RBE) and its major component of γ-Oryzanol (γ-Ory) against FD-induced premature endothelial senescence. Porcine coronary artery endothelial cells (PCAECs) were treated with FD alone or with RBE or γ-Ory. Senescence-associated β-galactosidase (SA-β-gal) activity, expression of cell cycle regulatory proteins, and oxidative stress levels were evaluated. The results indicated that SA-β-gal activity in the FD-treated PCAECs was attenuated by RBE and γ-Ory. Additionally, γ-Ory inhibited FD-induced cell cycle arrest, restored cell proliferation, and reduced the expression of cell cycle regulatory proteins. γ-Ory also inhibited oxidative stress and prevented senescence-associated NADPH oxidase and LAS activity in FD-exposed ECs suggesting that γ-Ory could protect against FD-induced ECs senescence and dysfunction.
Collapse
Affiliation(s)
- Saugat Shiwakoti
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Dalseong Gong
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea; Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260, INSERM (French National Institute of Health and Medical Research), University of Strasbourg, 67000, Strasbourg, France
| | - Kushal Sharma
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Ki-Woon Kang
- Division of Cardiology, College of Medicine, Heart Reasearch Institute and Biomedical Research Institute, Chung-Ang University Hospital, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Valérie B Schini-Kerth
- Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260, INSERM (French National Institute of Health and Medical Research), University of Strasbourg, 67000, Strasbourg, France
| | - Hyun Jung Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea.
| | - Min-Ho Oak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea.
| |
Collapse
|
20
|
Gentilin A, Moghetti P, Cevese A, Mattioli AV, Schena F, Tarperi C. Circadian and sex differences in post-ischemic vasodilation and reactive hyperemia in young individuals and elderly with and without type 2 diabetes. Microcirculation 2023; 30:e12818. [PMID: 37246844 DOI: 10.1111/micc.12818] [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: 07/27/2022] [Revised: 01/26/2023] [Accepted: 05/17/2023] [Indexed: 05/30/2023]
Abstract
OBJECTIVE Cardiovascular events show morning preference and sex differences, and are related to aging and type 2 diabetes. We assessed circadian variations and sex differences in vascular conductance (VC) and blood flow (BF) regulations following a brief bout of forearm ischemia. METHODS Young healthy individuals (H18-30) and elderly without (H50-80) and with type 2 diabetes (T2DM50-80) of both sexes were included. Forearm VC and BF, and mean arterial pressure (MAP) at baseline and following circulatory reperfusion were measured at 6 a.m. and 9 p.m. RESULTS In the morning compared to evening, following reperfusion, the VC and BF increments were similar in H18-30 (p>.71), but lower in H50-80 (p<.001) and T2DM50-80 (p<.01). VC and BF following circulatory reperfusion were higher in men than women in H18-30 (p<.001), but similar between sexes in the older groups (p>.23). CONCLUSIONS Forearm vasodilation following reperfusion is attenuated in the morning in the elderly, impairing BF towards an ischemic area. Diabetes does not affect the circadian regulation of VC and BF, but that of MAP. There are sex differences in VC and BF at baseline and after circulatory reperfusion at a young age, being greater in men, which disappear with aging without being affected by diabetes.
Collapse
Affiliation(s)
- Alessandro Gentilin
- Department of Neuroscience, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
- Italian Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Paolo Moghetti
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, Verona, Italy
- Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Antonio Cevese
- Department of Neuroscience, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
- Italian Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Anna Vittoria Mattioli
- Italian Institute for Cardiovascular Research (INRC), Bologna, Italy
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Schena
- Department of Neuroscience, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
- Italian Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Cantor Tarperi
- Department of Neuroscience, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
21
|
Wang Y, Wang H, Zhou J, Wang J, Wu H, Wu J. Interaction between body mass index and blood pressure on the risk of vascular stiffness : A community-based cross-sectional study and implications for nursing. Int J Nurs Sci 2023; 10:325-331. [PMID: 37545779 PMCID: PMC10401353 DOI: 10.1016/j.ijnss.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/06/2023] [Accepted: 06/19/2023] [Indexed: 08/08/2023] Open
Abstract
Objective This study aimed to analyze associations between body mass index (BMI) and vascular measurements (brachial ankle pulse wave velocity [baPWV] and ankle-brachial index [ABI]), whether blood pressure (BP) was involved in the relationship, and implications for nursing. Methods A cross-sectional study was conducted, including 1,894 middle-aged and older adults who underwent routine health screening at a community medical center in the Zhangjiang community in Shanghai, China. Participants were divided into three groups based on BMI: normal weight (n = 1,202), overweight (n = 480), and obese (n = 212). Multivariate linear regression models and smooth curve fittings were used to evaluate the associations between BMI and indices of vascular stiffness. Mediation analysis examined whether blood pressure mediate the association between BMI and vascular stiffness. Results Multiple linear regression analysis showed that BMI to be significantly and negatively associated with baPWV (β = -0.06 [-0.10, -0.03]) and ABI (β = -0.004 [-0.005, -0.003]), respectively. The interaction test results of systolic blood pressure (SBP) in the relationship between BMI and baPWV were significant (P for interaction = 0.01). After adjusting for age and sex, mediation analyses showed that BMI and baPWV were correlated (β = 0.090, P < 0.001) and mediated by SBP (β = 0.533, P < 0.001) and diastolic blood pressure (DBP) (β = 0.338, P < 0.001). A negative association was found between BMI and ABI (β = -0.135, P < 0.001), which appeared to be partially mediated by SBP (β = 0.124, P < 0.001) and DBP (β = 0.053, P < 0.001). Additional subgroup analysis based on blood pressure levels did not revealed statistically significant mediating effects. Conclusions Our findings showed conflicting associations between BMI and non-invasive vascular measurements of arterial stiffness. BP may have a biological interaction in the relationship between BMI and baPWV. Managing blood pressure and weight through comprehensive clinical care is crucial for preventing stiffness or blockage of vessels in middle-aged and older adults.
Collapse
Affiliation(s)
- Yiyan Wang
- School of Nursing, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Wang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Sichuan, China
| | - Jie Zhou
- School of Nursing, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaqi Wang
- School of Nursing, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hengjing Wu
- Clinical Center for Intelligent Rehabilitation Research, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Yangzhi Rehabilitation Hospital, Tongji University, Shanghai, China
| | - Jing Wu
- School of Nursing, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
Li K, Dai M, Sacirovic M, Zemmrich C, Pagonas N, Ritter O, Grisk O, Lubomirov LT, Lauxmann MA, Bramlage P, Persson AB, Buschmann E, Buschmann I, Hillmeister P. Leukocyte telomere length and mitochondrial DNA copy number associate with endothelial function in aging-related cardiovascular disease. Front Cardiovasc Med 2023; 10:1157571. [PMID: 37342445 PMCID: PMC10277745 DOI: 10.3389/fcvm.2023.1157571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/22/2023] [Indexed: 06/22/2023] Open
Abstract
Background We investigated the association between leukocyte telomere length, mitochondrial DNA copy number, and endothelial function in patients with aging-related cardiovascular disease (CVD). Methods In total 430 patients with CVD and healthy persons were enrolled in the current study. Peripheral blood was drawn by routine venipuncture procedure. Plasma and peripheral blood mononuclear cells (PBMCs) were collected. Cell-free genomic DNA (cfDNA) and leukocytic genomic DNA (leuDNA) were extracted from plasma and PBMCs, respectively. Relative telomere length (TL) and mitochondrial DNA copy number (mtDNA-CN) were analyzed using quantitative polymerase chain reaction. Endothelial function was evaluated by measuring flow-mediated dilation (FMD). The correlation between TL of cfDNA (cf-TL), mtDNA-CN of cfDNA (cf-mtDNA), TL of leuDNA (leu-TL), mtDNA-CN of leuDNA (leu-mtDNA), age, and FMD were analyzed based on Spearman's rank correlation. The association between cf-TL, cf-mtDNA, leu-TL, leu-mtDNA, age, gender, and FMD were explored using multiple linear regression analysis. Results cf-TL positively correlated with cf-mtDNA (r = 0.1834, P = 0.0273), and leu-TL positively correlated with leu-mtDNA (r = 0.1244, P = 0.0109). In addition, both leu-TL (r = 0.1489, P = 0.0022) and leu-mtDNA (r = 0.1929, P < 0.0001) positively correlated with FMD. In a multiple linear regression analysis model, both leu-TL (β = 0.229, P = 0.002) and leu-mtDNA (β = 0.198, P = 0.008) were positively associated with FMD. In contrast, age was inversely associated with FMD (β = -0.426, P < 0.0001). Conclusion TL positively correlates mtDNA-CN in both cfDNA and leuDNA. leu-TL and leu-mtDNA can be regarded as novel biomarkers of endothelial dysfunction.
Collapse
Affiliation(s)
- Kangbo Li
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mengjun Dai
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mesud Sacirovic
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Claudia Zemmrich
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Nikolaos Pagonas
- Department for Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Oliver Ritter
- Department for Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Lubomir T. Lubomirov
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Martin A. Lauxmann
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Anja Bondke Persson
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Buschmann
- Department of Cardiology, University Clinic Graz, Graz, Austria
| | - Ivo Buschmann
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Philipp Hillmeister
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| |
Collapse
|
23
|
Kunkel ON, Rand TA, Pyle JG, Baumfalk DR, Horn AG, Opoku‐Acheampong AB, Ade CJ, Musch TI, Ramsey MW, Delp MD, Behnke BJ. Head-up tilt does not enhance prostate tumor perfusion or oxygenation in young rats. Physiol Rep 2022; 10:e15548. [PMID: 36564177 PMCID: PMC9788965 DOI: 10.14814/phy2.15548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/25/2022] Open
Abstract
Solid tumors contain hypoxic regions that contribute to anticancer therapy resistance. Thus, mitigating tumor hypoxia may enhance the efficacy of radiation therapy which is commonly utilized for patients with prostate cancer. Increasing perfusion pressure in the prostate with head-up tilt (HUT) may augment prostate tumor perfusion and decrease hypoxia. The purpose of this study was to determine if an increase in the vascular hydrostatic gradient via 70° HUT increases tumor perfusion and decreases tumor hypoxia in a preclinical orthotopic model of prostate cancer. Male Copenhagen rats (n = 17) were orthotopically injected with Dunning R-3327 (AT-1) prostate adenocarcinoma cells to induce prostate tumors. After tumors were established, prostate tumor perfusion and hypoxia were measured in rats during level (0°) and 70° HUT positions. To compare the magnitude of the hydrostatic column to that present in humans, ultrasound was used to measure the heart to prostate distance in male human subjects to estimate the prostate vascular hydrostatic pressure with the upright posture. In young rats, no differences were detected in prostate tumor perfusion or prostate tumor hypoxia with 70° HUT versus the level position. However, from the retrospective study, young rats increased prostate vascular resistance to HUT, whereas aged rats lacked this response. Tumor vessels co-opted from existing functional vasculature in young rats may be sufficient to negate increases in perfusion pressure with HUT seen in aged rats. Additionally, in humans, the estimated hydrostatic column at the level of the prostate is five times greater than that of the rat. Therefore, 70° HUT may elicit increases in prostate/prostate tumor blood flow in humans that is not seen in rats.
Collapse
Affiliation(s)
- Olivia N. Kunkel
- Department of KinesiologyKansas State UniversityManhattanKansasUSA
| | - Taylor A. Rand
- Department of KinesiologyKansas State UniversityManhattanKansasUSA
| | - Joseph G. Pyle
- Department of KinesiologyKansas State UniversityManhattanKansasUSA
| | | | - Andrew G. Horn
- Department of KinesiologyKansas State UniversityManhattanKansasUSA
| | | | - Carl J. Ade
- Department of KinesiologyKansas State UniversityManhattanKansasUSA
| | - Timothy I. Musch
- Department of KinesiologyKansas State UniversityManhattanKansasUSA,Department of Anatomy and PhysiologyKansas State UniversityManhattanKansasUSA
| | - Michael W. Ramsey
- Department of Sport, Exercise, Recreation, and KinesiologyEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Michael D. Delp
- Department of Nutrition, Food and Exercise SciencesFlorida State UniversityTallahasseeFloridaUSA
| | - Bradley J. Behnke
- Department of KinesiologyKansas State UniversityManhattanKansasUSA,Johnson Cancer Research CenterKansas State UniversityManhattanKansasUSA
| |
Collapse
|
24
|
Michels EHA, Butler JM, Reijnders TDY, Cremer OL, Scicluna BP, Uhel F, Peters-Sengers H, Schultz MJ, Knight JC, van Vught LA, van der Poll T, Bos LDJ, Glas GJ, Hoogendijk AJ, van Hooijdonk RTM, Horn J, Huson MA, Schouten LRA, Straat M, Wieske L, Wiewel MA, Witteveen E, Bonten MJM, Cremer OM, Ong DSY, Frencken JF, Klouwenberg PMCK, Koster‐Brouwer ME, van de Groep K, Verboom DM. Association between age and the host response in critically ill patients with sepsis. Crit Care 2022; 26:385. [PMID: 36514130 PMCID: PMC9747080 DOI: 10.1186/s13054-022-04266-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The association of ageing with increased sepsis mortality is well established. Nonetheless, current investigations on the influence of age on host response aberrations are largely limited to plasma cytokine levels while neglecting other pathophysiological sepsis domains like endothelial cell activation and function, and coagulation activation. The primary objective of this study was to gain insight into the association of ageing with aberrations in key host response pathways and blood transcriptomes in sepsis. METHODS We analysed the clinical outcome (n = 1952), 16 plasma biomarkers providing insight in deregulation of specific pathophysiological domains (n = 899), and blood leukocyte transcriptomes (n = 488) of sepsis patients stratified according to age decades. Blood transcriptome results were validated in an independent sepsis cohort and compared with healthy individuals. RESULTS Older age was associated with increased mortality independent of comorbidities and disease severity. Ageing was associated with lower endothelial cell activation and dysfunction, and similar inflammation and coagulation activation, despite higher disease severity scores. Blood leukocytes of patients ≥ 70 years, compared to patients < 50 years, showed decreased expression of genes involved in cytokine signaling, and innate and adaptive immunity, and increased expression of genes involved in hemostasis and endothelial cell activation. The diminished expression of gene pathways related to innate immunity and cytokine signaling in subjects ≥ 70 years was sepsis-induced, as healthy subjects ≥ 70 years showed enhanced expression of these pathways compared to healthy individuals < 50 years. CONCLUSIONS This study provides novel evidence that older age is associated with relatively mitigated sepsis-induced endothelial cell activation and dysfunction, and a blood leukocyte transcriptome signature indicating impaired innate immune and cytokine signaling. These data suggest that age should be considered in patient selection in future sepsis trials targeting the immune system and/or the endothelial cell response.
Collapse
Affiliation(s)
- Erik H. A. Michels
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Joe M. Butler
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Tom D. Y. Reijnders
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Olaf L. Cremer
- grid.7692.a0000000090126352Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brendon P. Scicluna
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands ,grid.4462.40000 0001 2176 9482Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta ,grid.4462.40000 0001 2176 9482Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Fabrice Uhel
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Hessel Peters-Sengers
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Marcus J. Schultz
- grid.7177.60000000084992262Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands ,grid.10223.320000 0004 1937 0490Mahidol-Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand ,grid.4991.50000 0004 1936 8948Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Julian C. Knight
- grid.4991.50000 0004 1936 8948Nuffield Department of Medicine, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lonneke A. van Vught
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands ,grid.7177.60000000084992262Department of Intensive Care, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- grid.7177.60000000084992262Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands ,grid.7177.60000000084992262Division of Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
El Assar M, García-Rojo E, Sevilleja-Ortiz A, Sánchez-Ferrer A, Fernández A, García-Gómez B, Romero-Otero J, Rodríguez-Mañas L, Angulo J. Functional Role of STIM-1 and Orai1 in Human Microvascular Aging. Cells 2022; 11:cells11223675. [PMID: 36429103 PMCID: PMC9688234 DOI: 10.3390/cells11223675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
The impact of aging on vascular function is heterogeneous depending on the vascular territories. Calcium regulation plays a key role in vascular function and has been implicated in aging-related hypercontractility of corpus cavernosum. We aimed to evaluate stromal interaction molecule (STIM)/Orai system involvement in aging-related vascular alterations in the human macro and microvasculature. Aortae specimens and mesenteric arteries (MA), obtained from 45 organ donors, were functionally evaluated in organ chambers and wire myographs. Subjects were divided into groups either younger or older than 65-years old. The expressions of STIM-1, Orai1, and Orai3 were determined by immunofluorescence in the aorta and MA, and by Western blot in the aorta homogenates. The inhibition of STIM/Orai with YM-58483 (20 μM) reversed adrenergic hypercontractility in MA from older subjects but did not modify aging-related hypercontractility in the aortic strips. Aging was related to an increased expression of Orai1 in human aorta, while Orai1 and STIM-1 were upregulated in MA. STIM-1 and Orai1 protein expressions were inversely correlated to endothelial function in MA. Circulating levels of Orai1 were correlated with the inflammatory factor TNF-α and with the endothelial dysfunction marker asymmetric dimethylarginine. Aging is associated with an increased expression of the STIM/Orai system in human vessels with functional relevance only in the microvascular territory, suggesting its role in aging-related microvascular dysfunction.
Collapse
Affiliation(s)
- Mariam El Assar
- Fundación para la Investigación Biomédica, Hospital Universitario de Getafe, 28905 Getafe, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Esther García-Rojo
- Servicio de Urología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Alejandro Sevilleja-Ortiz
- Fundación para la Investigación Biomédica, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Alberto Sánchez-Ferrer
- Fundación para la Investigación Biomédica, Hospital Universitario de Getafe, 28905 Getafe, Spain
| | - Argentina Fernández
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Borja García-Gómez
- Servicio de Urología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Javier Romero-Otero
- Servicio de Urología, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Leocadio Rodríguez-Mañas
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Geriatría, Hospital Universitario de Getafe, 28905 Getafe, Spain
| | - Javier Angulo
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Correspondence:
| |
Collapse
|
26
|
Khalafi M, Sakhaei MH, Kazeminasab F, Symonds ME, Rosenkranz SK. The impact of high-intensity interval training on vascular function in adults: A systematic review and meta-analysis. Front Cardiovasc Med 2022; 9:1046560. [PMID: 36465439 PMCID: PMC9713318 DOI: 10.3389/fcvm.2022.1046560] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023] Open
Abstract
Aim We performed a systematic review and meta-analysis to investigate the effects of high-intensity interval training (HIIT) compared with moderate-intensity continuous training (MICT) or with no exercise (CON) on vascular function in adults who were free of cardiometabolic diseases and those with cardiometabolic diseases. Methods A search across three electronic databases including Scopus, PubMed, and Web of Science was conducted through February 2022 to identify the randomized trials evaluating HIIT vs. MICT and/or CON on vascular function as measured using brachial artery flow-mediated dilation (FMD) in adults. Separate analyses were conducted for HIIT vs. MICT and/or CON to calculate weighted mean differences (WMD) and 95% confidence intervals (95% CIs) using random or fixed models. Results A total of 36 studies involving 1,437 participants who were either free of cardiometabolic diseases or had cardiometabolic diseases were included in the meta-analysis. HIIT effectively increased FMD when compared with MICT [1.59% (95% CI 0.87-2.31), p = 0.001] or CON [3.80% (95% CI 2.58-5.01), p = 0.001]. Subgroup analysis showed that HIIT increased FMD in participants with cardiovascular and metabolic diseases, but not in participants who were free of cardiometabolic diseases. In addition, HIIT effectively increased FMD regardless of age and body mass index. Conclusion We confirm that HIIT is effective for improving vascular function in individuals with metabolic disorders and cardiovascular diseases and has a superior effect compared to MICT, demonstrating time efficiency. Systematic review registration [https://www.crd.york.ac.uk/prospero], identifier [CRD42022320863].
Collapse
Affiliation(s)
- Mousa Khalafi
- Department of Physical Education and Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran
| | - Mohammad Hossein Sakhaei
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Guilan, Iran
| | - Fatemeh Kazeminasab
- Department of Physical Education and Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran
| | - Michael E. Symonds
- Academic Unit of Population and Lifespan Sciences, Centre for Perinatal Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Sara K. Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
27
|
Parabiosis Improves Endothelial Dysfunction in Aged Female Mice. J Surg Res 2022; 278:119-131. [DOI: 10.1016/j.jss.2022.04.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 01/26/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022]
|
28
|
Gyanwali B, Tan CS, Petr J, Escobosa LLT, Vrooman H, Chen C, Mutsaerts HJ, Hilal S. Arterial Spin-Labeling Parameters and Their Associations with Risk Factors, Cerebral Small-Vessel Disease, and Etiologic Subtypes of Cognitive Impairment and Dementia. AJNR Am J Neuroradiol 2022; 43:1418-1423. [PMID: 36562454 PMCID: PMC9575536 DOI: 10.3174/ajnr.a7630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/01/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral small-vessel disease may alter cerebral blood flow (CBF) leading to brain changes and, hence, cognitive impairment and dementia. CBF and the spatial coefficient of variation can be measured quantitatively by arterial spin-labeling. We aimed to investigate the associations of demographics, vascular risk factors, location, and severity of cerebral small-vessel disease as well as the etiologic subtypes of cognitive impairment and dementia with CBF and the spatial coefficient of variation. MATERIALS AND METHODS Three hundred ninety patients with a diagnosis of no cognitive impairment, cognitive impairment no dementia, vascular cognitive impairment no dementia, Alzheimer disease, and vascular dementia were recruited from the memory clinic. Cerebral microbleeds and lacunes were categorized into strictly lobar, strictly deep, and mixed-location and enlarged perivascular spaces into the centrum semiovale and basal ganglia. Total and region-specific white matter hyperintensity volumes were segmented using FreeSurfer. CBF (n = 333) and the spatial coefficient of variation (n = 390) were analyzed with ExploreASL from 2D-EPI pseudocontinuous arterial spin-labeling images in white matter (WM) and gray matter (GM). To analyze the effect of demographic and vascular risk factors as well as the location and severity of cerebral small-vessel disease markers on arterial spin-labeling parameters, we constructed linear regression models, whereas logistic regression models were used to determine the association between arterial spin-labeling parameters and cognitive impairment no dementia, vascular cognitive impairment no dementia, Alzheimer disease, and vascular dementia. RESULTS Increasing age, male sex, hypertension, hyperlipidemia, history of heart disease, and smoking were associated with lower CBF and a higher spatial coefficient of variation. Higher numbers of lacunes and cerebral microbleeds were associated with lower CBF and a higher spatial coefficient of variation. Location-specific analysis showed mixed-location lacunes and cerebral microbleeds were associated with lower CBF. Higher total, anterior, and posterior white matter hyperintensity volumes were associated with a higher spatial coefficient of variation. No association was observed between enlarged perivascular spaces and arterial spin-labeling parameters. A higher spatial coefficient of variation was associated with the diagnosis of vascular cognitive impairment no dementia, Alzheimer's disease, and vascular dementia. CONCLUSIONS Reduced CBF and an increased spatial coefficient of variation were associated with cerebral small-vessel disease, and more specifically lacunes, whereas cerebral microbleeds and white matter hyperintensities were associated with WM-CBF and GM spatial coefficient of variation. The spatial coefficient of variation was associated with cognitive impairment and dementia, suggesting that hypoperfusion might be the key underlying mechanism for vascular brain damage.
Collapse
Affiliation(s)
- B Gyanwali
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
| | - C S Tan
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
| | - J Petr
- Helmholtz-Zentrum Dresden-Rossendorf (J.P.), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - L L T Escobosa
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
| | - H Vrooman
- Department of Radiology and Nuclear Medicine (H.V.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - C Chen
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
- Department of Pharmacology (C.C., S.H.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - H J Mutsaerts
- Department of Radiology (H.J.M.), VU University Medical Center, Amsterdam, the Netherlands
- Department of Radiology (H.J.M.), Brain Center Rudolf Magnus, University Medical Center, Utrecht, the Netherlands
| | - S Hilal
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
- Department of Pharmacology (C.C., S.H.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
29
|
Role of MicroRNAs in Cardiac Disease with Stroke in Pregnancy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5260085. [PMID: 36132229 PMCID: PMC9484966 DOI: 10.1155/2022/5260085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022]
Abstract
Pregnancy-related cardiovascular disease with stroke remains a considerable source of higher maternal morbidity and mortality occurs in periods of pregnancy, delivery, and postpartum. It is essential to counsel the mother before pregnancy by an expert cardiologist and obstetric team to discuss any event related to preexistent cardiac or past preeclampsia for estimation of maternal and fetal risks. In pregnancy, the cardiac state includes hypertensive disorders, ischemic heart disease, valvular disease, and postpartum stroke. The incidence of stroke is increasing in pregnancy, particularly in postpartum, and its strong relationship with hypertensive disorders of pregnancy (preeclampsia). The combined cardiologist and obstetrics team requires during pregnancy mainly due to the approach to the management of a cardiac disease that subsequently prevents stroke postpartum. Therefore, a general perception of cardiac disease during pregnancy, delivery, and postpartum should be a core knowledge extent for all cardiovascular and clinicians. Many studies provided linked that deregulation of microRNAs (miRNAs) in maternal circulation and placenta tissue may development of pregnancy complications including preeclampsia considered a diagnostic marker. The desire of this review provides a detailed outline of current knowledge and dealing in this field with strength on the physiological changes during pregnancy.
Collapse
|
30
|
Pasha M, Kirschenman R, Wooldridge A, Spaans F, Cooke CLM, Davidge ST. The Effect of Tauroursodeoxycholic Acid (TUDCA) Treatment on Pregnancy Outcomes and Vascular Function in a Rat Model of Advanced Maternal Age. Antioxidants (Basel) 2022; 11:1275. [PMID: 35883766 PMCID: PMC9312116 DOI: 10.3390/antiox11071275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Advanced maternal age (≥35 years) increases the risk of vascular complications in pregnancy that can result in fetal growth restriction and preeclampsia. Endoplasmic reticulum (ER) stress has been linked to adverse pregnancy outcomes in these complicated pregnancies. However, the role of ER stress in advanced maternal age is not known. We hypothesize that increased ER stress contributes to altered vascular function and poor pregnancy outcomes, and that treatment with the ER-stress inhibitor TUDCA will improve pregnancy outcomes. First, young and aged non-pregnant/pregnant rats were used to assess ER stress markers in mesenteric arteries; mesenteric artery phospho-eIF2α and CHOP expression were increased in aged dams compared to young dams. In a second study, young and aged control and TUDCA-treated dams were studied on gestational day (GD) 20 (term = 22 days). TUDCA treatment was provided via the drinking water throughout pregnancy (GD0-GD20; calculated dose of 150 mg/kg/day TUDCA). ER stress markers were quantified in mesenteric arteries, blood pressure was measured, pregnancy outcomes were recorded, mesenteric and main uterine arteries were isolated and vascular function was assessed by wire myography. Aged dams had increased phospho-eIF2α and CHOP expression, reduced fetal weight, reduced litter size, and impaired uterine artery relaxation. In the aged dams, TUDCA treatment reduced phospho-eIF2α and CHOP expression, reduced blood pressure, improved fetal body weight, and tended to improve uterine artery function compared to control-treated aged dams. In conclusion, our data illustrate the role of ER stress, as well as TUDCA as a potential therapeutic that may benefit pregnancy outcomes in advanced maternal age.
Collapse
Affiliation(s)
- Mazhar Pasha
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; (R.K.); (A.W.); (F.S.); (C.-L.M.C.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; (R.K.); (A.W.); (F.S.); (C.-L.M.C.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Amy Wooldridge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; (R.K.); (A.W.); (F.S.); (C.-L.M.C.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; (R.K.); (A.W.); (F.S.); (C.-L.M.C.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Christy-Lynn M. Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; (R.K.); (A.W.); (F.S.); (C.-L.M.C.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Sandra T. Davidge
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2S2, Canada; (R.K.); (A.W.); (F.S.); (C.-L.M.C.)
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, AB T6G 1C9, Canada
| |
Collapse
|
31
|
Plant-Based Foods and Vascular Function: A Systematic Review of Dietary Intervention Trials in Older Subjects and Hypothesized Mechanisms of Action. Nutrients 2022; 14:nu14132615. [PMID: 35807796 PMCID: PMC9268664 DOI: 10.3390/nu14132615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Cardiovascular diseases, still the leading cause of mortality in the world, are closely related to vascular function. Older subjects are more susceptible to endothelial dysfunction and therefore it is important to define possible preventive or support strategies, such as consumption of foods with health-promoting effects. This systematic review aims to summarize the currently available evidence on acute or chronic trials testing the effect of selected plant-based foods on vascular function parameters in older subjects, and consider plausible mechanisms that may support the main findings. A total of 15 trials were included and analyzed, testing the effects of beetroot, plum, blueberry, and vegetable oils. We found some interesting results regarding markers of vascular reactivity, in particular for beetroot, while no effects were found for markers of arterial stiffness. The amelioration of vascular function seems to be more related to the restoration of a condition of nitric oxide impairment, exacerbated by diseases or hypoxic condition, rather than the enhancement of a physiological situation, as indicated by the limited effects on healthy older subjects or in control groups with young subjects. However, the overall set of selected studies is, in any case, rather limited and heterogeneous in terms of characteristics of the studies, indicating the need for additional high-quality intervention trials to better clarify the role of vegetable foods in restoring and/or improving vascular function in order to better elucidate the mechanisms through which these foods may exert their vascular health benefits in older subjects.
Collapse
|
32
|
He Y, Chen Y, Yao L, Wang J, Sha X, Wang Y. The Inflamm-Aging Model Identifies Key Risk Factors in Atherosclerosis. Front Genet 2022; 13:865827. [PMID: 35706446 PMCID: PMC9191626 DOI: 10.3389/fgene.2022.865827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Atherosclerosis, one of the main threats to human life and health, is driven by abnormal inflammation (i.e., chronic inflammation or oxidative stress) during accelerated aging. Many studies have shown that inflamm-aging exerts a significant impact on the occurrence of atherosclerosis, particularly by inducing an immune homeostasis imbalance. However, the potential mechanism by which inflamm-aging induces atherosclerosis needs to be studied more thoroughly, and there is currently a lack of powerful prediction models.Methods: First, an improved inflamm-aging prediction model was constructed by integrating aging, inflammation, and disease markers with the help of machine learning methods; then, inflamm-aging scores were calculated. In addition, the causal relationship between aging and disease was identified using Mendelian randomization. A series of risk factors were also identified by causal analysis, sensitivity analysis, and network analysis.Results: Our results revealed an accelerated inflamm-aging pattern in atherosclerosis and suggested a causal relationship between inflamm-aging and atherosclerosis. Mechanisms involving inflammation, nutritional balance, vascular homeostasis, and oxidative stress were found to be driving factors of atherosclerosis in the context of inflamm-aging.Conclusion: In summary, we developed a model integrating crucial risk factors in inflamm-aging and atherosclerosis. Our computation pipeline could be used to explore potential mechanisms of related diseases.
Collapse
Affiliation(s)
- Yudan He
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Yao Chen
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Lilin Yao
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Junyi Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Xianzheng Sha
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
| | - Yin Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, China
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Yin Wang,
| |
Collapse
|
33
|
Jiménez-Altayó F, Cabrera A, Bagán A, Giménez-Llort L, D’Ocon P, Pérez B, Pallàs M, Escolano C. An Imidazoline 2 Receptor Ligand Relaxes Mouse Aorta via Off-Target Mechanisms Resistant to Aging. Front Pharmacol 2022; 13:826837. [PMID: 35645795 PMCID: PMC9133327 DOI: 10.3389/fphar.2022.826837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Imidazoline receptors (IR) are classified into three receptor subtypes (I1R, I2R, and I3R) and previous studies showed that regulation of I2R signaling has neuroprotective potential. In order to know if I2R has a role in modulating vascular tone in health and disease, we evaluated the putative vasoactive effects of two recently synthesized I2R ligands, diethyl (1RS,3aSR,6aSR)-5-(3-chloro-4-fluorophenyl)-4,6-dioxo-1-phenyl-1,3a,4,5,6,6a-hexahydropyrrolo[3,4-c]pyrrole -1-phosphonate (B06) and diethyl [(1-(3-chloro-4-fluorobenzyl)-5,5-dimethyl-4-phenyl-4,5-dihydro-1H-imidazol-4-yl]phosphonate] (MCR5). Thoracic aortas from Oncins France 1 (3- to 4-months-old) and C57BL/6 (3- to 4- and 16- to 17-months-old mice) were mounted in tissue baths to measure isometric tension. In young mice of both strains, MCR5 induced greater relaxations than either B06 or the high-affinity I2R selective ligand 2-(2-benzofuranyl)-2-imidazoline (2-BFI), which evoked marginal responses. MCR5 relaxations were independent of I2R, as IR ligands did not significantly affect them, involved activation of smooth muscle KATP channels and inhibition of L-type voltage-gated Ca2+ channels, and were only slightly modulated by endothelium-derived nitric oxide (negatively) and prostacyclin (positively). Notably, despite the presence of endothelial dysfunction in old mice, MCR5 relaxations were preserved. In conclusion, the present study provides evidence against a functional contribution of I2R in the modulation of vascular tone in the mouse aorta. Moreover, the I2R ligand MCR5 is an endothelium-independent vasodilator that acts largely via I2R-independent pathways and is resistant to aging. We propose MCR5 as a candidate drug for the management of vascular disease in the elderly.
Collapse
Affiliation(s)
- Francesc Jiménez-Altayó
- Department of Pharmacology, Therapeutics and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- *Correspondence: Francesc Jiménez-Altayó,
| | - Anna Cabrera
- Department of Pharmacology, Therapeutics and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pilar D’Ocon
- Department of Pharmacology, School of Medicine, Universidad de Valencia, Burjassot, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, Valencia, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutics and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mercè Pallàs
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, Barcelona, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Shamoon L, Espitia-Corredor JA, Dongil P, Menéndez-Ribes M, Romero A, Valencia I, Díaz-Araya G, Sánchez-Ferrer CF, Peiró C. RESOLVIN E1 ATTENUATES DOXORUBICIN-INDUCED ENDOTHELIAL SENESCENCE BY MODULATING NLRP3 INFLAMMASOME ACTIVATION. Biochem Pharmacol 2022; 201:115078. [PMID: 35551917 DOI: 10.1016/j.bcp.2022.115078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/14/2022] [Accepted: 05/02/2022] [Indexed: 01/10/2023]
Abstract
Endothelial cell senescence contributes to chronic inflammation and endothelial dysfunction, while favoring cardiovascular disorders and frailty. Senescent cells acquire a pro-inflammatory secretory phenotype that further propagates inflammation and senescence to neighboring cells. Cell senescence can be provoked by plethora of stressors, including inflammatory molecules and chemotherapeutic drugs. Doxorubicin (Doxo) is a powerful anthracycline anticancer drug whose clinical application is constrained by a dose-limiting cardiovascular toxicity. We here investigated whether cell senescence can contribute to the vascular damage elicited by Doxo. In human umbilical vein endothelial cells (HUVEC) cultures, Doxo (10-100 nM) increased the number of SA-β-gal positive cells and the levels of γH2AX, p21 and p53, used as markers of senescence. Moreover, we identified Doxo-induced senescence to be mediated by the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome, a key player of the immune innate system capable of releasing interleukin (IL)-1β. In fact, IL-1β itself mimicked the stimulatory action of Doxo on both NLRP3 activation and cellular senescence, while the pharmacological blockade of IL-1 receptors markedly attenuated the pro-senescence effects of Doxo. In search of additional pharmacological strategies to attenuate Doxo-induced endothelial senescence, we identified resolvin E1 (RvE1), an endogenous pro-resolving mediator, as capable of reducing cell senescence induced by both Doxo and IL-1β by interfering with the increased expression of pP65, NLRP3, and pro-IL-1β proteins and with the formation of active NLRP3 inflammasome complexes. Overall, RvE1 and the blockade of the NLRP3 inflammasome-IL-1β axis may offer a novel therapeutic approach against Doxo-induced cardiovascular toxicity and subsequent sequelae.
Collapse
Affiliation(s)
- Licia Shamoon
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Jenaro A Espitia-Corredor
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain; Laboratorio de Farmacología Molecular (FARMOLAB), Department of Pharmaceutical and Toxicological Chemistry, Faculty of Chemical Sciences and Pharmacy, Universidad de Chile, Santiago, Chile
| | - Pilar Dongil
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Marta Menéndez-Ribes
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Alejandra Romero
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Inés Valencia
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Guillermo Díaz-Araya
- Laboratorio de Farmacología Molecular (FARMOLAB), Department of Pharmaceutical and Toxicological Chemistry, Faculty of Chemical Sciences and Pharmacy, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic diseases ACCDiS, Universidad de Chile, Santiago, Chile.
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain.
| | - Concepción Peiró
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain.
| |
Collapse
|
35
|
Sun Z, Jiang D, Liu P, Muccio M, Li C, Cao Y, Wisniewski TM, Lu H, Ge Y. Age-Related Tortuosity of Carotid and Vertebral Arteries: Quantitative Evaluation With MR Angiography. Front Neurol 2022; 13:858805. [PMID: 35572919 PMCID: PMC9099009 DOI: 10.3389/fneur.2022.858805] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/30/2022] [Indexed: 01/14/2023] Open
Abstract
Background and Purpose The vascular tortuosity (VT) of the internal carotid artery (ICA), and vertebral artery (VA) can impact blood flow and neuronal function. However, few studies involved quantitative investigation of VT based on magnetic resonance imaging (MRI). The main purpose of our study was to evaluate the age and gender effects on ICA and VA regarding the tortuosity and flow changes by applying automatic vessel segmentation, centerline tracking, and phase mapping on MR angiography. Methods A total of 247 subjects (86 males and 161 females) without neurological diseases participated in this study. All subjects obtained T1-weighted MRI, 3D time-of-flight MR angiography, and 2D phase-contrast (PC) MRI scans. To generate quantitative tortuosity metrics from TOF images, the vessel segmentation and centerline tracking were implemented based on Otsu thresholding and fast marching algorithms, respectively. Blood flow and velocity were measured using PC MRI. Among the 247 subjects, 144 subjects (≤ 60 years, 49 males/95 females) were categorized as the young group; 103 subjects (>60 years, 37 males/66 females) were categorized as the old group. Results Independent t-test showed that older subjects had higher tortuosity metrics, whereas lower blood flow and velocity than young subjects (p < 0.0025, Bonferroni-corrected). Cerebral blood flow calculated using the sum flux of four target arteries normalized by the brain mass also showed significantly lower values in older subjects (p < 0.001). The age was observed to be positively correlated with the VT metrics. Compared to the males, the females demonstrated higher geometric indices within VAs as well as faster age-related vascular profile changes. After adjusting age and gender as covariates, maximum blood velocity is negatively correlated with geometric measurements. No association was observed between blood flux and geometric measures. Conclusions Vascular auto-segmentation, centerline tracking, and phase mapping provide promising quantitative assessments of tortuosity and its effects on blood flow. The neck arteries demonstrate quantifiable and significant age-related morphological and hemodynamic alterations. Moreover, females showed more distinct vascular changes with age. Our work is built upon a comprehensive quantitative investigation of a large cohort of populations covering adult lifespan using MRI, the results can serve as reference ranges of each decade in the general population.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, United States
- Vilcek Institute of Biomedical Science, NYU Grossman School of Medicine, New York, NY, United States
| | - Dengrong Jiang
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peiying Liu
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marco Muccio
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, United States
| | - Chenyang Li
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, United States
- Vilcek Institute of Biomedical Science, NYU Grossman School of Medicine, New York, NY, United States
| | - Yan Cao
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, TX, United States
| | - Thomas M. Wisniewski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, United States
| | - Hanzhang Lu
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yulin Ge
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
36
|
Remila L, Guenday-Tuereli N, Houngue U, Belcastro E, Bruckert C, Vandamme T, Tuereli E, Kerth P, Auger C, Schini-Kerth V. Intake of coated EPA:DHA 6:1 nanoparticles improves age-related endothelial dysfunction by restoring the endothelial formation of NO and improving oxidative stress: Role of the local angiotensin system. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
37
|
Fifor A, Krukowski K, Honda JR. Sex, ancestry, senescence, and aging (SAnSA) are stark drivers of nontuberculous mycobacterial pulmonary disease. J Clin Tuberc Other Mycobact Dis 2022; 26:100297. [PMID: 35059508 PMCID: PMC8760511 DOI: 10.1016/j.jctube.2022.100297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nontuberculous mycobacterial (NTM) pulmonary disease (PD) disproportionately affects otherwise healthy, older, Caucasian females. The reasons behind this are likely multifactorial involving several conspiring factors. A variety of factors are thought to contribute to increased susceptibility to NTM in the older adult including exposure to various environmental conditions and contaminants across the lifespan, genetic risk factors, hormonal changes, and immunodeficiency. Independent of sex and ancestry, respiratory muscle atrophy intensifies with age and an aging immune system can show functional decline of macrophages, poor lung migration and homing of dendritic cells, promotion of aberrant pro-inflammatory responses, acceleration of inflammation related to aging, and increased immunosenescence. The purpose of this review is to synthesize the current body of knowledge regarding the roles of sex, ancestry, senescence, and aging (SAnSA) in NTM acquisition and the possible mechanisms involved in NTM PD, highlighting age-related respiratory and immune system changes. We also summarize molecular tools and biomarkers of these fields and contextualize these into the study of NTM PD. Finally, we discuss the relevance of biomarkers described for senescence and aging and senolytic therapies as potentially new adjunctive strategies to reduce the burden of NTM PD.
Collapse
|
38
|
Assar ME, Angulo J, García-Rojo E, Sevilleja-Ortiz A, García-Gómez B, Fernández A, Sánchez-Ferrer A, La Fuente JM, Romero-Otero J, Rodríguez-Mañas L. Early manifestation of aging-related vascular dysfunction in human penile vasculature-A potential explanation for the role of erectile dysfunction as a harbinger of systemic vascular disease. GeroScience 2022; 44:485-501. [PMID: 34962617 PMCID: PMC8811115 DOI: 10.1007/s11357-021-00507-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/22/2021] [Indexed: 01/05/2023] Open
Abstract
Advanced age is related to functional alterations of human vasculature, but erectile dysfunction precedes systemic manifestations of vascular disease. The current study aimed to simultaneously evaluate the influence of aging on vascular function (relaxation and contraction responses) in systemic human vascular territories: aorta (HA) and resistance mesenteric arteries (HMA) and human corpus cavernosum (HCC) and penile resistance arteries (HPRA). Associations of oxidative stress and inflammation circulating biomarkers with age and functional responses were also determined. Vascular specimens were obtained from 76 organ donors (age range 18-87). Four age-groups were established: < 40, 40-55, 56-65 and > 65 years old. Increasing age was associated with a decline in endothelium-dependent relaxation induced by BK in HMA (r = -0.597, p = 0.0001), or by ACh in HCC (r = -0.505, p = 0.0022), and HPRA (r = -0.601, p = 0.0012). Significant impairment was detected at > 65 years old in HMA but earlier in penile vasculature (> 55 years old). Age-related reduction to H2O2-vasodilatory response started before in HCC (56-65 years old) than in HA (> 65 years old). In contrast to relaxation responses, aging-related hypercontractility to adrenergic stimulation was homogeneous: contractions significantly increased in subjects > 55 years old in all tested vessels. Although not significantly age related, circulating levels of ADMA (r = -0.681, p = 0.0052) and TNF-α (r = -0.537, p = 0.0385) were negatively correlated with endothelial vasodilation in HMA but not in HCC or HPRA. Penile vasculature exhibits an early impairment of endothelium-dependent and H2O2-induced vasodilations when compared to mesenteric microcirculation and aorta. Therefore, functional susceptibility of penile vasculature to the aging process may account for anticipation of erectile dysfunction to systemic manifestations of vascular disease.
Collapse
Affiliation(s)
- Mariam El Assar
- Fundación de Investigación Biomédica, del Hospital Universitario de Getafe, Getafe, Spain
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Angulo
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón Y Cajal, Madrid, Spain
| | - Esther García-Rojo
- Department of Urology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), Madrid, Spain
| | - Alejandro Sevilleja-Ortiz
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón Y Cajal, Madrid, Spain
| | - Borja García-Gómez
- Department of Urology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), Madrid, Spain
| | - Argentina Fernández
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón Y Cajal, Madrid, Spain
| | - Alberto Sánchez-Ferrer
- Fundación de Investigación Biomédica, del Hospital Universitario de Getafe, Getafe, Spain
| | - José M La Fuente
- Serviço de Urologia, Hospital Geral Santo Antonio, Porto, Portugal
| | - Javier Romero-Otero
- Department of Urology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), Madrid, Spain
| | - Leocadio Rodríguez-Mañas
- Fundación de Investigación Biomédica, del Hospital Universitario de Getafe, Getafe, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
- Division of Geriatric Medicine, Servicio de Geriatría, Hospital Universitario de Getafe, Ctra de Toledo km 12, 500, 8905, Getafe, Spain.
| |
Collapse
|
39
|
Romero A, Dongil P, Valencia I, Vallejo S, Hipólito-Luengo ÁS, Díaz-Araya G, Bartha JL, González-Arlanzón MM, Rivilla F, de la Cuesta F, Sánchez-Ferrer CF, Peiró C. Pharmacological Blockade of NLRP3 Inflammasome/IL-1β-Positive Loop Mitigates Endothelial Cell Senescence and Dysfunction. Aging Dis 2022; 13:284-297. [PMID: 35111374 PMCID: PMC8782550 DOI: 10.14336/ad.2021.0617] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
The clinical relevance of IL-1β in chronic inflammation underlying atherosclerosis has been reinforced by recent evidence associating pharmacological inhibition of the cytokine with lower cardiovascular risk. Previously, we have demonstrated a direct involvement of IL-1β in endothelial senescence. Therefore, this can be a key mechanism contributing to the sterile inflammatory milieu associated with aging, termed inflammaging. In the present study, we have evaluated whether a positive feedback of IL-1β in the NLRP3 inflammasome via NF-κB could promote human endothelial senescence in vitro and murine endothelial dysfunction in vivo. Our results indicate that the NLRP3 inflammasome is pivotal in mediating the detrimental effects of IL-1β, showing that auto-activation is a crucial feature boosting endothelial cell senescence in vitro, which is paralleled by vascular dysfunction in vivo. Hence, the inhibitor of NLRP3 inflammasome assembly, MCC 950, was able to disrupt the aforementioned positive loop, thus alleviating inflammation, cell senescence and vascular dysfunction. Besides, we explored alternative NLRP3 inflammasome inhibitory agents such as the RAS heptapeptide Ang-(1-7) and the anti-aging protein klotho, both of which demonstrated protective effects in vitro and in vivo. Altogether, our results highlight a fundamental role for the hereby described NLRP3 inflammasome/IL-1β positive feedback loop in stress-induced inflammaging and the associated vascular dysfunction, additionally providing evidence of a potential therapeutic use of MCC 950, Ang-(1-7) and recombinant klotho to block this loop and its deleterious effects.
Collapse
Affiliation(s)
- Alejandra Romero
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Pilar Dongil
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Inés Valencia
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain.,3PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Vallejo
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Álvaro San Hipólito-Luengo
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Guillermo Díaz-Araya
- 4Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile.,5Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Bartha
- 2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain.,6Department of Obstetrics and Gynecology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - María M González-Arlanzón
- 6Department of Obstetrics and Gynecology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fernando Rivilla
- 7Division of Pediatric Surgery, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Fernando de la Cuesta
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Carlos F Sánchez-Ferrer
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Concepción Peiró
- 1Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,2Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
40
|
Sanhueza-Olivares F, Troncoso MF, Pino-de la Fuente F, Martinez-Bilbao J, Riquelme JA, Norambuena-Soto I, Villa M, Lavandero S, Castro PF, Chiong M. A potential role of autophagy-mediated vascular senescence in the pathophysiology of HFpEF. Front Endocrinol (Lausanne) 2022; 13:1057349. [PMID: 36465616 PMCID: PMC9713703 DOI: 10.3389/fendo.2022.1057349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is one of the most complex and most prevalent cardiometabolic diseases in aging population. Age, obesity, diabetes, and hypertension are the main comorbidities of HFpEF. Microvascular dysfunction and vascular remodeling play a major role in its development. Among the many mechanisms involved in this process, vascular stiffening has been described as one the most prevalent during HFpEF, leading to ventricular-vascular uncoupling and mismatches in aged HFpEF patients. Aged blood vessels display an increased number of senescent endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). This is consistent with the fact that EC and cardiomyocyte cell senescence has been reported during HFpEF. Autophagy plays a major role in VSMCs physiology, regulating phenotypic switch between contractile and synthetic phenotypes. It has also been described that autophagy can regulate arterial stiffening and EC and VSMC senescence. Many studies now support the notion that targeting autophagy would help with the treatment of many cardiovascular and metabolic diseases. In this review, we discuss the mechanisms involved in autophagy-mediated vascular senescence and whether this could be a driver in the development and progression of HFpEF.
Collapse
Affiliation(s)
- Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Mayarling F. Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Francisco Pino-de la Fuente
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Javiera Martinez-Bilbao
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Jaime A. Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ignacio Norambuena-Soto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Monica Villa
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Pablo F. Castro
- Advanced Center for Chronic Diseases, Faculty of Medicine, Pontifical University Catholic of Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- *Correspondence: Mario Chiong,
| |
Collapse
|
41
|
Vascular Regulation of Hematopoietic Stem Cell Homeostasis, Regeneration, and Aging. CURRENT STEM CELL REPORTS 2021; 7:194-203. [PMID: 34868826 PMCID: PMC8639543 DOI: 10.1007/s40778-021-00198-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2021] [Indexed: 12/26/2022]
Abstract
Purpose of Review Hematopoietic stem cells (HSCs) sit at the top of the hierarchy that meets the daily burden of blood production. HSC maintenance relies on extrinsic cues from the bone marrow (BM) microenvironment to balance stem cell self-renewal and cell fate decisions. In this brief review, we will highlight the studies and model systems that define the centralized role of BM vascular endothelium in modulating HSC activity in health and stress. Recent Findings The BM microenvironment is composed of a diverse array of intimately associated vascular and perivascular cell types. Recent dynamic imaging studies, coupled with single-cell RNA sequencing (scRNA-seq) and functional readouts, have advanced our understanding of the HSC-supportive cell types and their cooperative mechanisms that govern stem cell fate during homeostasis, regeneration, and aging. These findings have established complex and discrete vascular microenvironments within the BM that express overlapping and unique paracrine signals that modulate HSC fate. Summary Understanding the spatial and reciprocal HSC-niche interactions and the molecular mechanisms that govern HSC activity in the BM vascular microenvironment will be integral in developing therapies aimed at ameliorating hematological disease and supporting healthy hematopoietic output.
Collapse
|
42
|
Barros PR, Costa TJ, Akamine EH, Tostes RC. Vascular Aging in Rodent Models: Contrasting Mechanisms Driving the Female and Male Vascular Senescence. FRONTIERS IN AGING 2021; 2:727604. [PMID: 35821995 PMCID: PMC9261394 DOI: 10.3389/fragi.2021.727604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Increasing scientific interest has been directed to sex as a biological and decisive factor on several diseases. Several different mechanisms orchestrate vascular function, as well as vascular dysfunction in cardiovascular and metabolic diseases in males and females. Certain vascular sex differences are present throughout life, while others are more evident before the menopause, suggesting two important and correlated drivers: genetic and hormonal factors. With the increasing life expectancy and aging population, studies on aging-related diseases and aging-related physiological changes have steeply grown and, with them, the use of aging animal models. Mouse and rat models of aging, the most studied laboratory animals in aging research, exhibit sex differences in many systems and physiological functions, as well as sex differences in the aging process and aging-associated cardiovascular changes. In the present review, we introduce the most common aging and senescence-accelerated animal models and emphasize that sex is a biological variable that should be considered in aging studies. Sex differences in the cardiovascular system, with a focus on sex differences in aging-associated vascular alterations (endothelial dysfunction, remodeling and oxidative and inflammatory processes) in these animal models are reviewed and discussed.
Collapse
Affiliation(s)
- Paula R. Barros
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Tiago J. Costa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Eliana H. Akamine
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
- *Correspondence: Rita C. Tostes, ; Eliana H. Akamine,
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Rita C. Tostes, ; Eliana H. Akamine,
| |
Collapse
|
43
|
Bhatia K, Ahmad S, Kindelin A, Ducruet AF. Complement C3a receptor-mediated vascular dysfunction: a complex interplay between aging and neurodegeneration. J Clin Invest 2021; 131:144348. [PMID: 33393493 DOI: 10.1172/jci144348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Vascular dysfunction resulting in compromised blood-brain barrier (BBB) integrity is evident in aging and disease. Although the complement C3a/C3a receptor (C3a/C3aR) axis influences normal brain aging and disease progression, the mechanisms governing endothelial C3aR-mediated neurovascular inflammation and BBB permeability remain unexplored. In this issue of the JCI, Propson et al. investigated endothelial C3a/C3aR signaling in normal, aged, and neurodegenerative mouse models. Endothelial C3aR signaling modulated age-dependent increases in VCAM1, initiated peripheral lymphocyte infiltration, and enhanced microglial activity. Increased calcium release downstream of C3aR signaling disrupted the vascular endothelial cadherin (VE-cadherin) junctions, increased BBB permeability, and degraded vascular structure and function. Mice lacking C3aR (C3ar1-/-) and mice treated with a C3aR antagonist showed attenuated age-related microglial reactivity and neurodegeneration. These results confirm that complement-mediated signaling impacts vascular health and BBB function in normal aging and neurodegenerative disease, suggesting that complement inhibitors represent a therapeutic option for cerebral microvascular dysfunction.
Collapse
Affiliation(s)
- Kanchan Bhatia
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center (SJHMC), Dignity Health, Phoenix, Arizona, USA.,School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona, USA
| | - Saif Ahmad
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center (SJHMC), Dignity Health, Phoenix, Arizona, USA
| | - Adam Kindelin
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center (SJHMC), Dignity Health, Phoenix, Arizona, USA
| | - Andrew F Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center (SJHMC), Dignity Health, Phoenix, Arizona, USA
| |
Collapse
|
44
|
Ling HZ, Garcia Jara P, Nicolaides KH, Kametas NA. Effect of maternal age on cardiac adaptation in pregnancy. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2021; 58:285-292. [PMID: 33592675 DOI: 10.1002/uog.23614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To compare longitudinal maternal hemodynamic changes throughout gestation between different age groups. METHODS This was a prospective longitudinal study assessing maternal hemodynamics using a bioreactance technique at 11 + 0 to 13 + 6, 19 + 0 to 24 + 0, 30 + 0 to 34 + 0 and 35 + 0 to 37 + 0 weeks' gestation. Women were divided into four groups according to maternal age at the first visit at 11 + 0 to 13 + 6 weeks: Group 1, < 25.0 years; Group 2, 25.0-30.0 years; Group 3, 30.1-34.9 years; and Group 4, ≥ 35.0 years. A multilevel linear mixed-effects model was performed to compare the repeat measurements of hemodynamic variables, correcting for demographics, medical and obstetric history, pregnancy complications, maternal age and gestational-age window. RESULTS The study population included 254 women in Group 1, 442 in Group 2, 618 in Group 3 and 475 in Group 4. Younger women (Group 1) had the highest cardiac output (CO) and lowest peripheral vascular resistance (PVR), and older women (Group 4) had the lowest CO and highest PVR throughout pregnancy. The higher CO seen in younger women was achieved through an increase in heart rate alone and not with a concomitant rise in stroke volume. Although the youngest age group demonstrated an apparently more favorable hemodynamic profile, it had the highest incidence of a small-for-gestational-age neonate. There was no significant difference between the groups in the incidence of pre-eclampsia. CONCLUSION Age-specific differences in maternal hemodynamic adaptation do not explain the differences in the incidence of a small-for-gestational-age neonate between age groups. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- H Z Ling
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - P Garcia Jara
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - K H Nicolaides
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - N A Kametas
- Fetal Medicine Research Institute, King's College Hospital, London, UK
| |
Collapse
|
45
|
Yi Q, Zha M, Yang Q, Zhang Y, Hou L, Ye X, Chen G, Shao J, Xia W, Song P. Trends in the prevalence of hypertension according to severity and phenotype in Chinese adults over two decades (1991-2015). J Clin Hypertens (Greenwich) 2021; 23:1302-1315. [PMID: 34128308 PMCID: PMC8678778 DOI: 10.1111/jch.14306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/22/2021] [Accepted: 05/23/2021] [Indexed: 11/28/2022]
Abstract
Until recently, few studies have reported the secular trend and associated factors of hypertension severities and phenotypes in China. In this study, the authors aimed to assess the trend in the prevalence of hypertension according to severity and phenotype in Chinese adults from 1991 to 2015 and to explore potential cardiometabolic factors. From the China Health and Nutrition Survey (CHNS), the authors included 164 682 records of adults (≥18 years). The prevalence of hypertension by severity (stage 1 and stage 2 hypertension) and by phenotype (isolated systolic hypertension [ISH], isolated diastolic hypertension [IDH], and systolic-diastolic hypertension [SDH]), during 1991-2015 was explored. The potential effects of demographic, socioeconomic, geographic, and cardiometabolic factors on hypertension severities and phenotypes were assessed by multivariable logistic regression. During 1991-2015, the overall prevalence of hypertension increased dramatically from 15.7% to 23.3%. For stage 1 and stage 2 hypertension, the age-standardized prevalence increased from 10.1% to 15.6% and from 5.5% to 7.4%, respectively. For ISH and SDH, the age-standardized prevalence rates rose from 3.6% to 6.4% and from 6.9% to 10.4%, respectively. Advanced age and medium/high urbanization were positively associated factors, whereas females, higher educational attainments, and residing in Southern China were negatively associated factors. Additionally, general obesity, central obesity, diabetes, and elevated triglyceride levels were linked to ISH, IDH, and SDH. The present study documents an increasing trend in the prevalence of hypertension, including different severities and phenotypes, among Chinese adults over more than two decades. Efforts for the prevention and management of hypertension are in urgent need in China.
Collapse
Affiliation(s)
- Qian Yi
- School of Public HealthZhejiang University School of MedicineHangzhouChina
| | - Mingming Zha
- Department of Neurology, Jinling HospitalMedical School of Southeast UniversityNanjingChina
| | - Qingwen Yang
- Department of Neurology, Jinling HospitalMedical School of Southeast UniversityNanjingChina
| | - Yan Zhang
- Faculty of Life Science and MedicineKings College LondonLondonUK
| | - Leying Hou
- School of Public HealthZhejiang University School of MedicineHangzhouChina
| | - Xinxin Ye
- School of Public HealthZhejiang University School of MedicineHangzhouChina
| | - Ge Chen
- Medical Research and Biometrics CenterNational Center for Cardiovascular DiseasesBeijingChina
| | - Jing Shao
- School of NursingZhejiang University School of MedicineHangzhouChina
| | - Wei Xia
- School of NursingSun Yat‐Sen UniversityGuangzhouChina
| | - Peige Song
- School of Public HealthZhejiang University School of MedicineHangzhouChina
- Women’s HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
46
|
Clayton ZS, Hutton DA, Brunt VE, VanDongen NS, Ziemba BP, Casso AG, Greenberg NT, Mercer AN, Rossman MJ, Campisi J, Melov S, Seals DR. Apigenin restores endothelial function by ameliorating oxidative stress, reverses aortic stiffening, and mitigates vascular inflammation with aging. Am J Physiol Heart Circ Physiol 2021; 321:H185-H196. [PMID: 34114892 DOI: 10.1152/ajpheart.00118.2021] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We assessed the efficacy of oral supplementation with the flavanoid apigenin on arterial function during aging and identified critical mechanisms of action. Young (6 mo) and old (27 mo) C57BL/6N mice (model of arterial aging) consumed drinking water containing vehicle (0.2% carboxymethylcellulose; 10 young and 7 old) or apigenin (0.5 mg/mL in vehicle; 10 young and 9 old) for 6 wk. In vehicle-treated animals, isolated carotid artery endothelium-dependent dilation (EDD), bioassay of endothelial function, was impaired in old versus young (70% ± 9% vs. 92% ± 1%, P < 0.0001) due to reduced nitric oxide (NO) bioavailability. Old mice had greater arterial reactive oxygen species (ROS) production and oxidative stress (higher nitrotyrosine) associated with greater nicotinamide adenine dinucleotide phosphate oxidase (oxidant enzyme) and lower superoxide dismutase 1 and 2 (antioxidant enzymes); ex vivo administration of Tempol (antioxidant) restored EDD to young levels, indicating ROS-mediated suppression of EDD. Old animals also had greater aortic stiffness as indicated by higher aortic pulse wave velocity (PWV, 434 ± 9 vs. 346 ± 5 cm/s, P < 0.0001) due to greater intrinsic aortic wall stiffness associated with lower elastin levels and higher collagen, advanced glycation end products (AGEs), and proinflammatory cytokine abundance. In old mice, apigenin restored EDD (96% ± 2%) by increasing NO bioavailability, normalized arterial ROS, oxidative stress, and antioxidant expression, and abolished ROS inhibition of EDD. Moreover, apigenin prevented foam cell formation in vitro (initiating step in atherosclerosis) and mitigated age-associated aortic stiffening (PWV 373 ± 5 cm/s) by normalizing aortic intrinsic wall stiffness, collagen, elastin, AGEs, and inflammation. Thus, apigenin is a promising therapeutic for arterial aging.NEW & NOTEWORTHY Our study provides novel evidence that oral apigenin supplementation can reverse two clinically important indicators of arterial dysfunction with age, namely, vascular endothelial dysfunction and large elastic artery stiffening, and prevents foam cell formation in an established cell culture model of early atherosclerosis. Importantly, our results provide extensive insight into the biological mechanisms of apigenin action, including increased nitric oxide bioavailability, normalization of age-related increases in arterial ROS production and oxidative stress, reversal of age-associated aortic intrinsic mechanical wall stiffening and adverse remodeling of the extracellular matrix, and suppression of vascular inflammation. Given that apigenin is commercially available as a dietary supplement in humans, these preclinical findings provide the experimental basis for future translational studies assessing the potential of apigenin to treat arterial dysfunction and reduce cardiovascular disease risk with aging.
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - David A Hutton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Nicholas S VanDongen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Brian P Ziemba
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Abigail G Casso
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Nathan T Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Amanda N Mercer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, California.,Lawrence Berkley National Laboratory, Berkeley, California
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, California
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
47
|
Otsubo A, Miyazato M, Oshiro T, Kimura R, Matsuo T, Miyata Y, Sakai H. Age-associated bladder and urethral coordination impairment and changes in urethral oxidative stress in rats. Life Sci 2021; 279:119690. [PMID: 34111460 DOI: 10.1016/j.lfs.2021.119690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/22/2021] [Accepted: 05/30/2021] [Indexed: 10/24/2022]
Abstract
AIMS We examined age-associated changes in bladder and urethral coordination involving the nitric oxide (NO)/soluble guanylyl cyclase (sGC) system, which induces urethral smooth muscle relaxation, and urethral ischemic/oxidative stress changes in rats. MAIN METHODS Sixteen female Sprague-Dawley rats were divided into young (3 months old) and middle-aged (12-15 months old) groups. Urethral activity was evaluated by simultaneously recording intravesical pressure under isovolumetric conditions and urethral perfusion pressure (UPP) under urethane anesthesia. Sodium nitroprusside (SNP, 0.1 mg/kg), an NO donor, and BAY 41-2272, a novel NO-independent stimulator of sGC (0.1 mg/kg), were administered intravenously to both groups. N-nitro-l-arginine methyl ester hydrochloride (l-NAME, 100 mg/kg) was also injected intravenously, to inhibit NO synthase activity in both groups. Staining for the ischemic marker, hypoxia-inducible factor-1α (HIF-1α), and the oxidative stress markers, 8-hydroxy-2'-deoxyguanosine (8-OHdG) and malondialdehyde (MDA), was performed on tissue sections of the urethra, in both groups. KEY FINDINGS Baseline UPP and UPP changes were significantly lower in middle-aged rats than in young rats. After administration of SNP and BAY 41-2272, baseline UPP and UPP nadir were significantly decreased in both groups. After administration of l-NAME, UPP change/bladder contraction amplitude in young rats was still lower than at baseline but was completely restored to control levels in middle-aged rats. Immunoreactivity of HIF-1α, 8-OHdG, and MDA was higher in middle-aged rats than in young rats. SIGNIFICANCE Age-associated ischemic and oxidative stress in the urethra might be correlated with impairment of the NO/sGC system and with coordination of the bladder and urethra.
Collapse
Affiliation(s)
- Asato Otsubo
- Department of Urology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Minoru Miyazato
- Department of Systems Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.
| | - Takuma Oshiro
- Department of Urology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Ryu Kimura
- Department of Urology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Tomohiro Matsuo
- Department of Urology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yasuyoshi Miyata
- Department of Urology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hideki Sakai
- Department of Urology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
48
|
Mahmoud M, Mayer M, Cancel LM, Bartosch AM, Mathews R, Tarbell JM. The glycocalyx core protein Glypican 1 protects vessel wall endothelial cells from stiffness-mediated dysfunction and disease. Cardiovasc Res 2021; 117:1592-1605. [PMID: 32647868 PMCID: PMC8152694 DOI: 10.1093/cvr/cvaa201] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/22/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Arterial stiffness is an underlying risk factor and a hallmark of cardiovascular diseases. The endothelial cell (EC) glycocalyx is a glycan rich surface layer that plays a key role in protecting against EC dysfunction and vascular disease. However, the mechanisms by which arterial stiffness promotes EC dysfunction and vascular disease are not fully understood, and whether the mechanism involves the protective endothelial glycocalyx is yet to be determined. We hypothesized that endothelial glycocalyx protects the endothelial cells lining the vascular wall from dysfunction and disease in response to arterial stiffness. METHODS AND RESULTS Cells cultured on polyacrylamide (PA) gels of substrate stiffness 10 kPa (mimicking the subendothelial stiffness of aged, unhealthy arteries) showed a significant inhibition of glycocalyx expression compared to cells cultured on softer PA gels (2.5 kPa, mimicking the subendothelial stiffness of young, healthy arteries). Specifically, gene and protein analyses revealed that a glycocalyx core protein Glypican 1 was inhibited in cells cultured on stiff PA gels. These cells had enhanced endothelial cell dysfunction as determined by enhanced cell inflammation (enhanced inflammatory gene expression, monocyte adhesion, and inhibited nitric oxide expression), proliferation, and EndMT. Removal of Glypican 1 using gene-specific silencing with siRNA or gene overexpression using a plasmid revealed that Glypican 1 is required to protect against stiffness-mediated endothelial cell dysfunction. Consistent with this, using a model of age-mediated stiffness, older mice exhibited a reduced expression of Glypican 1 and enhanced endothelial cell dysfunction compared to young mice. Glypican 1 gene deletion in knockout mice (GPC1-/-) exacerbated endothelial dysfunction in young mice, which normally had high endothelial expression, but not in old mice that normally expressed low levels. Endothelial cell dysfunction was exacerbated in young, but not aged, Glypican 1 knockout mice (GPC1-/-). CONCLUSION Arterial stiffness promotes EC dysfunction and vascular disease at least partly through the suppression of the glycocalyx protein Glypican 1. Glypican 1 contributes to the protection against endothelial cell dysfunction and vascular disease in endothelial cells.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Mariya Mayer
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Limary M Cancel
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Anne Marie Bartosch
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Rick Mathews
- Oregon Health & Science University, School of Medicine, Portland, OR, USA
| | - John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| |
Collapse
|
49
|
Kawarazaki W, Fujita T. Kidney and epigenetic mechanisms of salt-sensitive hypertension. Nat Rev Nephrol 2021; 17:350-363. [PMID: 33627838 DOI: 10.1038/s41581-021-00399-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
Dietary salt intake increases blood pressure (BP) but the salt sensitivity of BP differs between individuals. The interplay of ageing, genetics and environmental factors, including malnutrition and stress, contributes to BP salt sensitivity. In adults, obesity is often associated with salt-sensitive hypertension. The children of women who experience malnutrition during pregnancy are at increased risk of developing obesity, diabetes and salt-sensitive hypertension as adults. Similarly, the offspring of mice that are fed a low-protein diet during pregnancy develop salt-sensitive hypertension in association with aberrant DNA methylation of the gene encoding type 1A angiotensin II receptor (AT1AR) in the hypothalamus, leading to upregulation of hypothalamic AT1AR and renal sympathetic overactivity. Ageing is also associated with salt-sensitive hypertension. In aged mice, promoter methylation leads to reduced kidney production of the anti-ageing factor Klotho and a decrease in circulating soluble Klotho. In the setting of Klotho deficiency, salt-induced activation of the vascular Wnt5a-RhoA pathway leads to ageing-associated salt-sensitive hypertension, potentially as a result of reduced renal blood flow and increased peripheral resistance. Thus, kidney mechanisms and aberrant DNA methylation of certain genes are involved in the development of salt-sensitive hypertension during fetal development and old age. Three distinct paradigms of epigenetic memory operate on different timescales in prenatal malnutrition, obesity and ageing.
Collapse
Affiliation(s)
- Wakako Kawarazaki
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan. .,School of Medicine, Shinshu University, Matsumoto, Japan. .,Research Center for Social Systems, Shinshu University, Matsumoto, Japan.
| |
Collapse
|
50
|
Shafer AT, Beason-Held L, An Y, Williams OA, Huo Y, Landman BA, Caffo BS, Resnick SM. Default mode network connectivity and cognition in the aging brain: the effects of age, sex, and APOE genotype. Neurobiol Aging 2021; 104:10-23. [PMID: 33957555 DOI: 10.1016/j.neurobiolaging.2021.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/04/2021] [Accepted: 03/24/2021] [Indexed: 01/18/2023]
Abstract
The default mode network (DMN) overlaps with regions showing early Alzheimer's Disease (AD) pathology. Age, sex, and apolipoprotein E ɛ4 are the predominant risk factors for developing AD. How these risk factors interact to influence DMN connectivity and connectivity-cognition relationships before the onset of impairment remains unknown. Here, we examined these issues in 475 cognitively normal adults, targeting total DMN connectivity, its anticorrelated network (acDMN), and the DMN-hippocampal component. There were four main findings. First, in the ɛ3 homozygous group, lower DMN and acDMN connectivity was observed with age. Second, sex and ɛ4 modified the relationship between age and connectivity for the DMN and hippocampus with ɛ4 vs. ɛ3 males showing sustained or higher connectivity with age. Third, in the ɛ3 group, age and sex modified connectivity-cognition relationships with the oldest participants having the most differential patterns due to sex. Fourth, ɛ4 carriers with lower connectivity had poorer cognitive performance. Taken together, our results show the three predominant risk factors for AD interact to influence brain function and function-cognition relationships.
Collapse
Affiliation(s)
- Andrea T Shafer
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD.
| | - Lori Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD
| | - Owen A Williams
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD
| | - Yuankai Huo
- Department of Electrical Engineering & Computer Science, Vanderbilt University, Nashville, TN
| | - Bennett A Landman
- Department of Electrical Engineering & Computer Science, Vanderbilt University, Nashville, TN
| | - Brian S Caffo
- Department of Biostatistics, Johns Hopkins School of Public Health, Baltimore, MD
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD.
| |
Collapse
|