1
|
Riddell A, Flynn A, Bergugnat H, Dowsett L, Miller A. SDMA as a marker and mediator in cerebrovascular disease. Clin Sci (Lond) 2024; 138:1305-1323. [PMID: 39391895 PMCID: PMC11479986 DOI: 10.1042/cs20241021] [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: 06/06/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024]
Abstract
Symmetric dimethylarginine (SDMA) is a methylated derivative of arginine, generated by all cells as a by-product of cellular metabolism and eliminated via the kidney. For many years SDMA has been considered inert and of little biological significance. However, a growing body of evidence now suggests this view is outdated and that circulating SDMA levels may, in fact, be intricately linked to endothelial dysfunction and vascular risk. In this review, we specifically examine SDMA within the context of cerebrovascular disease, with a particular focus on ischaemic stroke. We first discuss pre-clinical evidence supporting the notion that SDMA has effects on nitric oxide signalling, inflammation, oxidative stress, and HDL function. We then appraise the most recent clinical studies that explore the relationship between circulating SDMA and cerebrovascular risk factors, such as chronic kidney disease, hypertension, atrial fibrillation, and atherosclerosis, exploring whether any associations may arise due to the existence of shared risk factors. Finally, we consider the evidence that elevated circulating SDMA is linked to poor outcomes following ischaemic and haemorrhagic stroke. We draw upon pre-clinical insights into SDMA function to speculate how SDMA may not only be a marker of cerebrovascular disease but could also directly influence cerebrovascular pathology, and we highlight the pressing need for more mechanistic pre-clinical studies alongside adequately powered, longitudinal clinical studies to fully evaluate SDMA as a marker/mediator of disease.
Collapse
Affiliation(s)
- Alexandra Riddell
- British Heart Foundation Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Arun Flynn
- British Heart Foundation Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Hugo Bergugnat
- British Heart Foundation Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Laura B. Dowsett
- British Heart Foundation Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Alyson A. Miller
- British Heart Foundation Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
2
|
Ji M, Ran X, Zuo H, Zhang Q. Novel Insights into the Kallikrein-Kinin System in Fulminant Myocarditis: Physiological Basis and Potential Therapeutic Advances. J Inflamm Res 2024; 17:7347-7360. [PMID: 39429854 PMCID: PMC11490248 DOI: 10.2147/jir.s488237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Fulminant myocarditis (FM) is characterized by rapid cardiac deterioration often instigated by an inflammatory cytokine storm. The kallikrein-kinin system (KKS) is a metabolic cascade known for releasing vasoactive kinins, such as bradykinin-related peptides, possessing diverse pharmacological activities that include inflammation, regulation of vascular permeability, endothelial barrier dysfunction, and blood pressure modulation. The type 1 and type 2 bradykinin receptors (B1R and B2R), integral components of the KKS system, mediate the primary biological effects of kinin peptides. This review aims to offer a comprehensive overview of the primary mechanisms of the KKS in FM, including an examination of the structural components, regulatory activation, and downstream signaling pathways of the KKS. Furthermore, it explores the involvement of the tissue kallikrein/B1R/inducible nitric oxide synthase (TK/B1R/iNOS) pathway in myocyte dysfunction, modulation of the immune response, and preservation of endothelial barrier integrity. The potential therapeutic advances targeting the inhibition of the KKS in managing FM will be discussed, providing valuable insights for the development of clinical treatment strategies.
Collapse
Affiliation(s)
- Mengmeng Ji
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xiao Ran
- Department of Critical-Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Houjuan Zuo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
3
|
Ribatti D. The crossroad between tumor and endothelial cells. Clin Exp Med 2024; 24:227. [PMID: 39325128 PMCID: PMC11427519 DOI: 10.1007/s10238-024-01490-1] [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: 07/24/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Endothelial cells are critical in tumor development, and the specific targeting of endothelial cells offers a potent means to effectively impede angiogenesis and suppress the growth of tumors. Tumor endothelial cells are responsible for the loss of anticancer immunity, the so-called endothelial anergy, i.e., the unresponsiveness of tumor endothelial cells to pro-inflammatory stimulation, not allowing adhesion of immune cells to the endothelium. Endothelial cells downregulate antigen presentation and recruitment of immune cells, contributing to immunosuppression. Targeting endothelial cells may assist in improving the immune effect of immune cells in tumor microenvironment.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Università Degli Studi Di Bari, Piazza Giulio Cesare 11, 70125, Bari, Italy.
| |
Collapse
|
4
|
Visagie JL, Aruwajoye GS, van der Sluis R. Pharmacokinetics of aspirin: evaluating shortcomings in the literature. Expert Opin Drug Metab Toxicol 2024:1-14. [PMID: 39092921 DOI: 10.1080/17425255.2024.2386368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Aspirin is known for its therapeutic benefits in preventing strokes and relieving pain. However, it is toxic to some individuals, and the biological mechanisms causing toxicity are unknown. Limited literature is available on the role of glycine conjugation as the principal pathway in aspirin detoxification. Previous studies have quantified this two-step enzyme reaction as a singular enzymatic process. Consequently, the individual contributions of these enzymes to the kinetics remain unclear. AREAS COVERED This review summarized the available information on the pharmacokinetics and detoxification of aspirin by the glycine conjugation pathway. Literature searches were conducted using Google Scholar and the academic journal databases accessible through the North-West University Library. Furthermore, the factors affecting interindividual variation in aspirin metabolism and what is known regarding aspirin toxicity were discussed. EXPERT OPINION The greatest drawback in understanding the pharmacokinetics of aspirin is the limited information available on the substrate preference of the xenobiotic ligase (ACSM) responsible for activating salicylate to salicyl-CoA. Furthermore, previous pharmacokinetic studies did not consider the contribution of other substrates from the diet or genetic variants, to the detoxification rate of glycine conjugation. Impaired glycine conjugation might contribute to adverse health effects seen in Reye's syndrome and cancer.
Collapse
Affiliation(s)
- Jacobus Lukas Visagie
- Focus Area for Human Metabolomics, North-West University, Potchefstroom, South Africa
| | | | - Rencia van der Sluis
- Focus Area for Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
5
|
Pavan AR, Terroni B, Dos Santos JL. Endothelial dysfunction in Sickle Cell Disease: Strategies for the treatment. Nitric Oxide 2024; 149:7-17. [PMID: 38806107 DOI: 10.1016/j.niox.2024.05.003] [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: 02/15/2024] [Revised: 04/15/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
Sickle Cell Anemia (SCA), is an inherited hemoglobinopathy characterized by the presence of an abnormal hemoglobin (HbS), being the most prevalent sickle cell disease (SCD). SCA is characterized by vascular endothelial dysfunction, which contributes significantly to various clinical conditions, including but not limited to pulmonary hypertension, priapism, cutaneous leg ulceration, and stroke. The pathophysiology of endothelial dysfunction (ED) in SCA is a multifaceted process involving a chronic inflammatory and hypercoagulable state. Key factors include hemolysis-associated elements like reduced arginine and nitric oxide (NO) availability, elevated levels of vascular adhesion molecules, the uncoupling effect of NO synthase, heightened arginase activity, an environment characterized by oxidative stress with the production of reactive oxygen and nitrogen species, and occurrences of ischemia-reperfusion injury, along with apolipoprotein A-1 depletion. The urgency for novel interventions addressing ED is evident. Presently, there is a focus on investigating small molecules that disrupt the arginine-nitric oxide pathway, exhibiting anti-inflammatory and antioxidant properties while diminishing levels of cellular and vascular adhesion molecules. In this mini-review article, we delve into the progress made in strategies for treating ED in SCD with the aim of cultivating insights for drug design.
Collapse
Affiliation(s)
- Aline Renata Pavan
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, Brazil.
| | - Barbara Terroni
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Brazil
| | | |
Collapse
|
6
|
Catella J, Guillot N, Nader E, Skinner S, Poutrel S, Hot A, Connes P, Fromy B. Controversies in the pathophysiology of leg ulcers in sickle cell disease. Br J Haematol 2024; 205:61-70. [PMID: 38867511 DOI: 10.1111/bjh.19584] [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: 12/22/2023] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Patients with sickle cell disease (SCD) often experience painful vaso-occlusive crises and chronic haemolytic anaemia, as well as various acute and chronic complications, such as leg ulcers. Leg ulcers are characterized by their unpredictability, debilitating pain and prolonged healing process. The pathophysiology of SCD leg ulcers is not well defined. Known risk factors include male gender, poor social conditions, malnutrition and a lack of compression therapy when oedema occurs. Leg ulcers typically start with spontaneous pain, followed by induration, hyperpigmentation, blister formation and destruction of the epidermis. SCD is characterized by chronic haemolysis, increased oxidative stress and decreased nitric oxide bioavailability, which promote ischaemia and inflammation and consequently impair vascular function in the skin. This cutaneous vasculopathy, coupled with venostasis around the ankle, creates an ideal environment for local vaso-occlusive crises, which can result in the development of leg ulcers that resemble arterial ulcers. Following the development of the ulcer, healing is hindered as a result of factors commonly observed in venous ulceration, including venous insufficiency, oedema and impaired angiogenesis. All of these factors are modulated by genetic factors. However, our current understanding of these genetic factors remains limited and does not yet enable us to accurately predict ulceration susceptibility.
Collapse
Affiliation(s)
- Judith Catella
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305), CNRS/Université Claude Bernard Lyon 1, Lyon, France
| | - Nicolas Guillot
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Elie Nader
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Sarah Skinner
- Clinical Research and Epidemiology Unit, Montpellier University, Montpellier, France
| | - Solène Poutrel
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Arnaud Hot
- Service de Médecine Interne et Vasculaire, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Philippe Connes
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), Sorbonne, Paris, France
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Equipe "Biologie Vasculaire et du Globule Rouge", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Berengère Fromy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI UMR 5305), CNRS/Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
7
|
Dos Passos RR, Santos CV, Priviero F, Briones AM, Tostes RC, Webb RC, Bomfim GF. Immunomodulatory Activity of Cytokines in Hypertension: A Vascular Perspective. Hypertension 2024; 81:1411-1423. [PMID: 38686582 PMCID: PMC11168883 DOI: 10.1161/hypertensionaha.124.21712] [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] [Indexed: 05/02/2024]
Abstract
Cytokines play a crucial role in the structure and function of blood vessels in hypertension. Hypertension damages blood vessels by mechanisms linked to shear forces, activation of the renin-angiotensin-aldosterone and sympathetic nervous systems, oxidative stress, and a proinflammatory milieu that lead to the generation of neoantigens and damage-associated molecular patterns, ultimately triggering the release of numerous cytokines. Damage-associated molecular patterns are recognized by PRRs (pattern recognition receptors) and activate inflammatory mechanisms in endothelial cells, smooth muscle cells, perivascular nerves, and perivascular adipose tissue. Activated vascular cells also release cytokines and express factors that attract macrophages, dendritic cells, and lymphocytes to the blood vessels. Activated and differentiated T cells into Th1, Th17, and Th22 in secondary lymphoid organs migrate to the vessels, releasing specific cytokines that further contribute to vascular dysfunction and remodeling. This chronic inflammation alters the profile of endothelial and smooth muscle cells, making them dysfunctional. Here, we provide an overview of how cytokines contribute to hypertension by impacting the vasculature. Furthermore, we explore clinical perspectives about the modulation of cytokines as a potential therapeutic intervention to specifically target hypertension-linked vascular dysfunction.
Collapse
Affiliation(s)
- Rinaldo R Dos Passos
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
| | - Cintia V Santos
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil (C.V.S., R.C.T.)
| | - Fernanda Priviero
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Biomedical Engineering, College of Engineering and Computing (F.P., R.C.W.), University of South Carolina, Columbia
| | - Ana M Briones
- Department of Pharmacology, Facultad de Medicina, Universidad Autónoma de Madrid, Spain (A.M.B.)
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain (A.M.B.)
- CIBER Cardiovascular, Madrid, Spain (A.M.B.)
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil (C.V.S., R.C.T.)
| | - R Clinton Webb
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Biomedical Engineering, College of Engineering and Computing (F.P., R.C.W.), University of South Carolina, Columbia
| | - Gisele F Bomfim
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- NUPADS - Health Education and Research Center, Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil (G.F.B.)
| |
Collapse
|
8
|
Baek KI, Ryu K. Role of Flow-Sensitive Endothelial Genes in Atherosclerosis and Antiatherogenic Therapeutics Development. J Cardiovasc Transl Res 2024; 17:609-623. [PMID: 38010480 DOI: 10.1007/s12265-023-10463-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease that is the underlying cause of cardiovascular disease which initiates from endothelial dysfunction from genetic and environmental risk factors, including biomechanical forces: blood flow. Endothelial cells (ECs) lining the inner arterial wall regions exposed to disturbed flow are prone to atherosclerosis development, whereas the straight regions exposed to stable flow are spared from the disease. These flow patterns induce genome- and epigenome-wide changes in gene expression in ECs. Through the sweeping changes in gene expression, disturbed flow reprograms ECs from athero-protected cell types under the stable flow condition to pro-atherogenic cell conditions. The pro-atherogenic changes induced by disturbed flow, in combination with additional risk factors such as hypercholesterolemia, lead to the progression of atherosclerosis. The flow-sensitive genes and proteins are critical in understanding the mechanisms and serve as novel targets for antiatherogenic therapeutics.
Collapse
Affiliation(s)
- Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kitae Ryu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Department of Biotechnology, The University of Suwon, 17, Wauan-Gil, Bongdam-Eup, Hwaseong-Si, Gyeonggi-Do, 18323, Republic of Korea.
| |
Collapse
|
9
|
Hong SG, Ashby JW, Kennelly JP, Wu M, Steel M, Chattopadhyay E, Foreman R, Tontonoz P, Tarling EJ, Turowski P, Gallagher-Jones M, Mack JJ. Mechanosensitive membrane domains regulate calcium entry in arterial endothelial cells to protect against inflammation. J Clin Invest 2024; 134:e175057. [PMID: 38771648 PMCID: PMC11213468 DOI: 10.1172/jci175057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/10/2024] [Indexed: 05/23/2024] Open
Abstract
Endothelial cells (ECs) in the descending aorta are exposed to high laminar shear stress, and this supports an antiinflammatory phenotype. High laminar shear stress also induces flow-aligned cell elongation and front-rear polarity, but whether these are required for the antiinflammatory phenotype is unclear. Here, we showed that caveolin-1-rich microdomains polarize to the downstream end of ECs that are exposed to continuous high laminar flow. These microdomains were characterized by high membrane rigidity, filamentous actin (F-actin), and raft-associated lipids. Transient receptor potential vanilloid (TRPV4) ion channels were ubiquitously expressed on the plasma membrane but mediated localized Ca2+ entry only at these microdomains where they physically interacted with clustered caveolin-1. These focal Ca2+ bursts activated endothelial nitric oxide synthase within the confines of these domains. Importantly, we found that signaling at these domains required both cell body elongation and sustained flow. Finally, TRPV4 signaling at these domains was necessary and sufficient to suppress inflammatory gene expression and exogenous activation of TRPV4 channels ameliorated the inflammatory response to stimuli both in vitro and in vivo. Our work revealed a polarized mechanosensitive signaling hub in arterial ECs that dampened inflammatory gene expression and promoted cell resilience.
Collapse
Affiliation(s)
- Soon-Gook Hong
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| | | | - John P. Kennelly
- Molecular Biology Institute
- Department of Pathology and Laboratory Medicine, and
| | - Meigan Wu
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| | | | | | - Rob Foreman
- Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, California, USA
| | - Peter Tontonoz
- Molecular Biology Institute
- Department of Pathology and Laboratory Medicine, and
| | | | - Patric Turowski
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Marcus Gallagher-Jones
- Correlated Imaging, Rosalind Franklin Institute, Harwell Science & Innovation Campus, Didcot, United Kingdom
| | - Julia J. Mack
- Department of Medicine, Division of Cardiology
- Molecular Biology Institute
| |
Collapse
|
10
|
Jerez HE, Simioni YR, Ghosal K, Morilla MJ, Romero EL. Cholesterol nanoarchaeosomes for alendronate targeted delivery as an anti-endothelial dysfunction agent. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:517-534. [PMID: 38774586 PMCID: PMC11106671 DOI: 10.3762/bjnano.15.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024]
Abstract
Sodium alendronate (ALN) is a very hydrosoluble and poorly permeable molecule used as an antiresorptive agent and with vascular anticalcifying capacity. Loaded into targeted nanovesicles, its anti-inflammatory activity may be amplified towards extra-osseous and noncalcified target cells, such as severely irritated vascular endothelium. Here cytotoxicity, mitochondrial membrane potential, ATP content, and membrane fluidity of human endothelial venous cells (HUVECs) were determined after endocytosis of ALN-loaded nanoarchaeosomes (nanoARC-Chol(ALN), made of polar lipids from Halorubrum tebenquichense: cholesterol 7:3 w/w, 166 ± 5 nm, 0.16 ± 0.02 PDI, -40.8 ± 5.4 mV potential, 84.7 ± 21 µg/mg ALN/total lipids, TL). The effect of nanoARC-Chol(ALN) was further assessed on severely inflamed HUVECs. To that aim, HUVECs were grown on a porous barrier on top of a basal compartment seeded either with macrophages or human foam cells. One lighter and one more pronounced inflammatory context was modelled by adding lipopolysaccharide (LPS) to the apical or the apical and basal compartments. The endocytosis of nanoARC-Chol(ALN), was observed to partly reduce the endothelial-mesenchymal transition of HUVECs. Besides, while 10 mg/mL dexamethasone, 7.6 mM free ALN and ALN-loaded liposomes failed, 50 μg/mL TL + 2.5 μg/mL ALN (i.e., nanoARC-Chol(ALN)) reduced the IL-6 and IL-8 levels by, respectively, 75% and 65% in the mild and by, respectively, 60% and 40% in the pronounced inflammation model. This is the first report showing that the endocytosis of nanoARC-Chol(ALN) by HUVECs magnifies the anti-inflammatory activity of ALN even under conditions of intense irritation, not only surpassing that of free ALN but also that of dexamethasone.
Collapse
Affiliation(s)
- Horacio Emanuel Jerez
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| | - Yamila Roxana Simioni
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| | - Kajal Ghosal
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja Subodh Chandra Mallick Rd., Jadavpur, Kolkata 700032, West Bengal, India
| | - Maria Jose Morilla
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| | - Eder Lilia Romero
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| |
Collapse
|
11
|
Mol N, Priya A, Singh AK, Mago P, Shalimar, Ray AK. "Unravelling the impacts of climatic heat events on cardiovascular health in animal models". ENVIRONMENTAL RESEARCH 2024; 248:118315. [PMID: 38301760 DOI: 10.1016/j.envres.2024.118315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/03/2024]
Abstract
Climate change has led to an increase in high ambient temperatures, causing extreme heat events worldwide. According to the World Meteorological Organization (WMO), July 2023 marked a historic milestone as the Earth reached its hottest recorded temperature, precisely hitting the critical threshold of 1.5 °C set by the Paris Agreement. This distressing development led to a stark warning from the United Nations, signaling the dawn of what they call "an era of global boiling". The increasing global temperatures can result in high heat stress which leads to various physiological and biochemical alterations in the human body. Given that cardiovascular diseases (CVDs) are a leading cause of morbidity and mortality globally, heat events exacerbate this public health issue. While clinical and in-vitro studies have suggested a range of pathophysiological and biochemical mechanisms underlying the body's response to heat stress, the complex nature of organ-system level interactions makes precise investigation challenging. To address this knowledge gap effectively, the use of animal models exposed to acute or chronic heat stress can be invaluable. These models can closely replicate the multifaceted effects observed in humans during heat stress conditions. Despite extensive independent reviews, limited focus has been shed on the high heat-induced cardiovascular complications and their mechanisms, particularly utilizing animal models. Therefore, in this comprehensive review, we highlight the crucial biomarkers altered during heat stress, contributing significantly to various CVDs. We explore potential mechanisms underlying heat-induced cardiovascular dysfunction and damage, delving into various animal models. While traditional rodent models are commonly employed, we also examine less conventional models, including ruminants, broilers, canines, and primates. Furthermore, we delve into various potential therapeutic approaches and preventive measures. These insights hold significant promise for the development of more effective clinical interventions against the effects of heat stress on the human cardiovascular system.
Collapse
Affiliation(s)
- Nidhi Mol
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Anjali Priya
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Payal Mago
- Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, New Delhi, India; Campus of Open Learning, University of Delhi, New Delhi, India
| | - Shalimar
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| |
Collapse
|
12
|
Xu Y, Miller CP, Tykodi SS, Akilesh S, Warren EH. Signaling crosstalk between tumor endothelial cells and immune cells in the microenvironment of solid tumors. Front Cell Dev Biol 2024; 12:1387198. [PMID: 38726320 PMCID: PMC11079179 DOI: 10.3389/fcell.2024.1387198] [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: 02/16/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
Tumor-associated endothelial cells (TECs) are crucial mediators of immune surveillance and immune escape in the tumor microenvironment (TME). TECs driven by angiogenic growth factors form an abnormal vasculature which deploys molecular machinery to selectively promote the function and recruitment of immunosuppressive cells while simultaneously blocking the entry and function of anti-tumor immune cells. TECs also utilize a similar set of signaling regulators to promote the metastasis of tumor cells. Meanwhile, the tumor-infiltrating immune cells further induce the TEC anergy by secreting pro-angiogenic factors and prevents further immune cell penetration into the TME. Understanding the complex interactions between TECs and immune cells will be needed to successfully treat cancer patients with combined therapy to achieve vasculature normalization while augmenting antitumor immunity. In this review, we will discuss what is known about the signaling crosstalk between TECs and tumor-infiltrating immune cells to reveal insights and strategies for therapeutic targeting.
Collapse
Affiliation(s)
- Yuexin Xu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Chris P. Miller
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Scott S. Tykodi
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Kidney Research Institute, University of Washington, Seattle, WA, United States
| | - Edus H. Warren
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, United States
| |
Collapse
|
13
|
Sherratt SCR, Mason RP, Libby P, Steg PG, Bhatt DL. Do patients benefit from omega-3 fatty acids? Cardiovasc Res 2024; 119:2884-2901. [PMID: 38252923 PMCID: PMC10874279 DOI: 10.1093/cvr/cvad188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/11/2023] [Accepted: 09/26/2023] [Indexed: 01/24/2024] Open
Abstract
Omega-3 fatty acids (O3FAs) possess beneficial properties for cardiovascular (CV) health and elevated O3FA levels are associated with lower incident risk for CV disease (CVD.) Yet, treatment of at-risk patients with various O3FA formulations has produced disparate results in large, well-controlled and well-conducted clinical trials. Prescription formulations and fish oil supplements containing low-dose mixtures of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have routinely failed to prevent CV events in primary and secondary prevention settings when added to contemporary care, as shown most recently in the STRENGTH and OMEMI trials. However, as observed in JELIS, REDUCE-IT, and RESPECT-EPA, EPA-only formulations significantly reduce CVD events in high-risk patients. The CV mechanism of action of EPA, while certainly multifaceted, does not depend solely on reductions of circulating lipids, including triglycerides (TG) and LDL, and event reduction appears related to achieved EPA levels suggesting that the particular chemical and biological properties of EPA, as compared to DHA and other O3FAs, may contribute to its distinct clinical efficacy. In vitro and in vivo studies have shown different effects of EPA compared with DHA alone or EPA/DHA combination treatments, on atherosclerotic plaque morphology, LDL and membrane oxidation, cholesterol distribution, membrane lipid dynamics, glucose homeostasis, endothelial function, and downstream lipid metabolite function. These findings indicate that prescription-grade, EPA-only formulations provide greater benefit than other O3FAs formulations tested. This review summarizes the clinical findings associated with various O3FA formulations, their efficacy in treating CV disease, and their underlying mechanisms of action.
Collapse
Affiliation(s)
- Samuel C R Sherratt
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Elucida Research LLC, Beverly, MA, USA
| | - R Preston Mason
- Elucida Research LLC, Beverly, MA, USA
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ph Gabriel Steg
- Université Paris-Cité, INSERM_UMR1148/LVTS, FACT (French Alliance for Cardiovascular Trials), Assistance Publique–Hôpitaux de Paris, Hôpital Bichat, Paris, France
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, NewYork 10029-5674, NY, USA
| |
Collapse
|
14
|
Leone P, Malerba E, Susca N, Favoino E, Perosa F, Brunori G, Prete M, Racanelli V. Endothelial cells in tumor microenvironment: insights and perspectives. Front Immunol 2024; 15:1367875. [PMID: 38426109 PMCID: PMC10902062 DOI: 10.3389/fimmu.2024.1367875] [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: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
The tumor microenvironment is a highly complex and dynamic mixture of cell types, including tumor, immune and endothelial cells (ECs), soluble factors (cytokines, chemokines, and growth factors), blood vessels and extracellular matrix. Within this complex network, ECs are not only relevant for controlling blood fluidity and permeability, and orchestrating tumor angiogenesis but also for regulating the antitumor immune response. Lining the luminal side of vessels, ECs check the passage of molecules into the tumor compartment, regulate cellular transmigration, and interact with both circulating pathogens and innate and adaptive immune cells. Thus, they represent a first-line defense system that participates in immune responses. Tumor-associated ECs are involved in T cell priming, activation, and proliferation by acting as semi-professional antigen presenting cells. Thus, targeting ECs may assist in improving antitumor immune cell functions. Moreover, tumor-associated ECs contribute to the development at the tumor site of tertiary lymphoid structures, which have recently been associated with enhanced response to immune checkpoint inhibitors (ICI). When compared to normal ECs, tumor-associated ECs are abnormal in terms of phenotype, genetic expression profile, and functions. They are characterized by high proliferative potential and the ability to activate immunosuppressive mechanisms that support tumor progression and metastatic dissemination. A complete phenotypic and functional characterization of tumor-associated ECs could be helpful to clarify their complex role within the tumor microenvironment and to identify EC specific drug targets to improve cancer therapy. The emerging therapeutic strategies based on the combination of anti-angiogenic treatments with immunotherapy strategies, including ICI, CAR T cells and bispecific antibodies aim to impact both ECs and immune cells to block angiogenesis and at the same time to increase recruitment and activation of effector cells within the tumor.
Collapse
Affiliation(s)
- Patrizia Leone
- Internal Medicine Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Eleonora Malerba
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Aldo Moro University of Bari, Bari, Italy
| | - Nicola Susca
- Internal Medicine Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Elvira Favoino
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Giuliano Brunori
- Centre for Medical Sciences, University of Trento and Nephrology and Dialysis Division, Santa Chiara Hospital, Provincial Health Care Agency (APSS), Trento, Italy
| | - Marcella Prete
- Internal Medicine Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Vito Racanelli
- Centre for Medical Sciences, University of Trento and Internal Medicine Division, Santa Chiara Hospital, Provincial Health Care Agency (APSS), Trento, Italy
| |
Collapse
|
15
|
Loscalzo J. Nitric oxide in vascular biology: elegance in complexity. J Clin Invest 2024; 134:e176747. [PMID: 38357929 PMCID: PMC10866648 DOI: 10.1172/jci176747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
|
16
|
Kaplish D, Vagha JD, Meshram RJ, Lohiya S. A Comprehensive Review of Inhaled Nitric Oxide Therapy: Current Trends, Challenges, and Future Directions. Cureus 2024; 16:e53558. [PMID: 38445143 PMCID: PMC10913844 DOI: 10.7759/cureus.53558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 02/04/2024] [Indexed: 03/07/2024] Open
Abstract
This comprehensive review explores the multifaceted landscape of inhaled nitric oxide (iNO) therapy, tracing its historical evolution, mechanisms of action, clinical applications, challenges, and future directions. The nitric oxide signaling pathway, characterized by vasodilatory effects and anti-inflammatory properties, forms the foundation of iNO's therapeutic efficacy. Clinical applications are found in neonatal respiratory distress syndrome, pulmonary hypertension, and acute respiratory distress syndrome, showcasing its versatility. However, challenges, including cost considerations, technical intricacies, safety concerns, and resistance, highlight the nuanced landscape surrounding iNO therapy. Implications for clinical practice underscore the need for a tailored and evidence-based approach, considering individual patient characteristics and indications. Recommendations for future research emphasize ongoing exploration, novel indications, and the development of targeted therapies. In conclusion, this review positions iNO as a dynamic and adaptable intervention, poised to reshape therapeutic strategies and enhance patient outcomes in critical care.
Collapse
Affiliation(s)
- Divyanshi Kaplish
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Jayant D Vagha
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Revat J Meshram
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Sham Lohiya
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| |
Collapse
|
17
|
Reddy H, Javvaji CK, Malali S, Kumar S, Acharya S, Toshniwal S. Navigating the Cytokine Storm: A Comprehensive Review of Chemokines and Cytokines in Sepsis. Cureus 2024; 16:e54275. [PMID: 38496165 PMCID: PMC10944554 DOI: 10.7759/cureus.54275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 02/11/2024] [Indexed: 03/19/2024] Open
Abstract
This comprehensive review thoroughly explores the intricate relationship between chemokines, cytokines, and the cytokine storm in sepsis, offering a nuanced understanding of the molecular mechanisms underpinning this life-threatening syndrome. Beginning with examining sepsis stages and immune response dynamics, the review emphasizes the dysregulation leading to the cytokine storm, where pro- and anti-inflammatory cytokines disrupt the delicate immune equilibrium. Delving into chemokines, the discussion encompasses subfamilies, receptors, and functions, highlighting their critical roles in immune cell migration and activation during sepsis. The implications for clinical practice are substantial, suggesting avenues for targeted diagnostics and therapeutic interventions. The review identifies areas for future research, including the search for novel biomarkers, deeper insights into cytokine regulation, and the pursuit of personalized medicine approaches. This comprehensive exploration aims to guide clinicians, researchers, and policymakers in navigating the complexities of sepsis, fostering a foundation for transformative advancements in understanding and managing this formidable clinical challenge.
Collapse
Affiliation(s)
- Harshitha Reddy
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Chaitanya Kumar Javvaji
- Pediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Suprit Malali
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sunil Kumar
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sourya Acharya
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Saket Toshniwal
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
18
|
Kang P, Dong P. CircMETTL14(11)S upregulated METTL14 and induced CXCR4 to aggravate endothelial inflammation and atherosclerosis. Int Immunopharmacol 2024; 126:110979. [PMID: 37972448 DOI: 10.1016/j.intimp.2023.110979] [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: 02/21/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 11/19/2023]
Abstract
Endothelial inflammatory response can induce a variety of cardiovascular diseases, including atherosclerosis (AS). As a member of the m6A methyltransferase family, methyltransferase like 14 (METTL14) was reported to propel endothelial inflammation and aggravate AS. In this study, qRT-PCR and western blot analyses were performed to detect the RNA and protein levels of genes. To analyze the cyclic structure and localization of circMETTL14(11)S, agarose gel electrophoresis, subcellular fractionation and FISH assays were conducted. The role of circMETTL14(11)S on endothelial inflammation was exposed by monocyte adhesion assay. Luciferase reporter, chromatin immunoprecipitation (ChIP), pull-down and RNA binding protein immunoprecipitation (RIP) assays were conducted to explore the mechanism of circMETTL14(11)S on endothelial inflammation and AS. We found that circMETTL14(11)S (hsa_circ_0125169) expressed highly in TNF-α-induced endothelial inflammation and positively regulated the expression of METTL14 in human umbilical vein endothelial cells (HUVECs). CircMETTL14(11)S facilitated endothelial inflammation of HUVECs by METTL14. Based on the nuclear location, circMETTL14(11)S was found to activate METTL14 transcription via cooperating with SRY-box transcription factor 2 (SOX2). METTL14 accelerated the m6A methylation and stabilization of C-X-C motif chemokine receptor 4 (CXCR4) mRNA. Further, the facilitation of circMETTL14(11)S/METTL14/CXCR4 on TNF-α-induced endothelial inflammation of HUVECs was verified. Collectively, circMETTL14(11)S/METTL14/CXCR4 axis aggravated endothelial inflammation and AS.
Collapse
Affiliation(s)
- Pinfang Kang
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Peng Dong
- Department of Cardiovascular Medicine, Affiliated Hospital of Hangzhou Normal University, Clinical School of Medicine, Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Hangzhou 310015, China.
| |
Collapse
|
19
|
Sienel RI, Mamrak U, Biller J, Roth S, Zellner A, Parakaw T, Khambata RS, Liesz A, Haffner C, Ahluwalia A, Seker BF, Plesnila N. Inhaled nitric oxide suppresses neuroinflammation in experimental ischemic stroke. J Neuroinflammation 2023; 20:301. [PMID: 38102677 PMCID: PMC10725028 DOI: 10.1186/s12974-023-02988-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
Ischemic stroke is a major global health issue and characterized by acute vascular dysfunction and subsequent neuroinflammation. However, the relationship between these processes remains elusive. In the current study, we investigated whether alleviating vascular dysfunction by restoring vascular nitric oxide (NO) reduces post-stroke inflammation. Mice were subjected to experimental stroke and received inhaled NO (iNO; 50 ppm) after reperfusion. iNO normalized vascular cyclic guanosine monophosphate (cGMP) levels, reduced the elevated expression of intercellular adhesion molecule-1 (ICAM-1), and returned leukocyte adhesion to baseline levels. Reduction of vascular pathology significantly reduced the inflammatory cytokines interleukin-1β (Il-1β), interleukin-6 (Il-6), and tumor necrosis factor-α (TNF-α), within the brain parenchyma. These findings suggest that vascular dysfunction is responsible for leukocyte adhesion and that these processes drive parenchymal inflammation. Reversing vascular dysfunction may therefore emerge as a novel approach to diminish neuroinflammation after ischemic stroke and possibly other ischemic disorders.
Collapse
Affiliation(s)
- Rebecca I Sienel
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
| | - Uta Mamrak
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
| | - Janina Biller
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
| | - Andreas Zellner
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
| | - Tipparat Parakaw
- William Harvey Research Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- William Harvey Research Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christof Haffner
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Burcu F Seker
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Klinikum der Universität München and Ludwig Maximilian University (LMU) Munich, Feodor-Lynen Str. 17, 81377, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
20
|
Chang A, Martin KA, Colvin M, Bellumkonda L. Role of ascorbic acid in cardiac allograft vasculopathy. Clin Transplant 2023; 37:e15153. [PMID: 37792313 DOI: 10.1111/ctr.15153] [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: 06/08/2023] [Revised: 09/04/2023] [Accepted: 09/22/2023] [Indexed: 10/05/2023]
Abstract
PURPOSE OF THE REVIEW Cardiac allograft vasculopathy (CAV) is a progressive fibroproliferative disease which occurs after heart transplantation and is associated with significant long-term morbidity and mortality. Currently available strategies including statins, mammalian target of rapamycin (mTOR) inhibitors, and revascularization, have limited overall effectiveness in treating this pathology once the disease process is established. mTOR inhibitors, while effective when used early in the disease process, are not well tolerated, and hence not routinely used in post-transplant care. RECENT DATA Recent work on rodent models have given us a novel mechanistic understanding of effects of ascorbic acid in preventing CAV. TET methyl cytosine dioxygenase2 (TET2) reduces vascular smooth muscle cell (VSMC) apoptosis and intimal thickening. TET2 is repressed by interferon γ (IFNγ) in the setting of CAV. Ascorbic acid has been shown to promote TET2 activity and attenuate allograft vasculopathy in animal models and CAV progression in a small clinical trial. SUMMARY CAV remains a challenging disease process and needs better preventative strategies. Ascorbic acid improves endothelial dysfunction, reduces reactive oxygen species, and prevents development of intimal hyperplasia by preventing smooth muscle cell apoptosis and hyperproliferation. Further large-scale randomized control studies of ascorbic acid are needed to establish the role in routine post-transplant management.
Collapse
Affiliation(s)
- Alyssa Chang
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathleen A Martin
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Monica Colvin
- Division of Cardiology, Department of Medicine, Yale University, New Haven, Connecticut, USA
| | - Lavanya Bellumkonda
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
Nguyen TD, Rahman NT, Sessa WC, Lee MY. Endothelial nitric oxide synthase (eNOS) S1176 phosphorylation status governs atherosclerotic lesion formation. Front Cardiovasc Med 2023; 10:1279868. [PMID: 38034389 PMCID: PMC10683645 DOI: 10.3389/fcvm.2023.1279868] [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: 08/18/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Objective We have previously demonstrated the in vivo importance of the Akt-eNOS substrate-kinase relationship, as defective postnatal angiogenesis characteristic of global Akt1-null mice is rescued when bred to 'gain-of-function' eNOS S1176D mutant mice. While multiple studies support the vascular protective role of endothelial NO generation, the causal role of Akt1-dependent eNOS S1176 phosphorylation during atherosclerotic plaque formation is not yet clear. Approach and results We herein bred congenic 'loss-of-function' eNOS S1176A and 'gain-of-function' eNOS S1176D mutant mice to the exacerbated atherogenic Akt1-/-; ApoE-/- double knockout mice to definitively test the importance of Akt-mediated eNOS S1176 phosphorylation during atherogenesis. We find that a single amino acid substitution at the eNOS S1176 phosphorylation site yields divergent effects on atherosclerotic plaque formation, as an eNOS phospho-mimic aspartate (D) substitution at S1176 leads to favorable lipid profiles and decreased indices of atherosclerosis, even when on a proatherogenic Akt1 global deletion background. Conversely, mice harboring an unphosphorylatable mutation to alanine (S1176A) result in increased plasma lipids, increased lesion formation and cellular apoptosis, phenocopying the physiological consequence of eNOS deletion and/or impaired enzyme function. Furthermore, gene expression analyses of whole aortas indicate a combinatorial detriment from NO deficiency and Western Diet challenge, as 'loss-of-function' eNOS S1176A mice on a Western Diet present a unique expression pattern indicative of augmented T-cell activity when compared to eNOS S1176D mice. Conclusions By using genetic epistasis approaches, we conclusively demonstrate that Akt-mediated eNOS S1176 phosphorylation and subsequent eNOS activation remains to be the most physiologically relevant method of NO production to promote athero-protective effects.
Collapse
Affiliation(s)
- Tung D. Nguyen
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago School of Medicine, Chicago, IL, United States
| | - Nur-Taz Rahman
- Bioinformatics Support Group, Yale University Cushing/Whitney Medical Library, New Haven, CT, United States
| | - William C. Sessa
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Monica Y. Lee
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago School of Medicine, Chicago, IL, United States
| |
Collapse
|
22
|
Dugbartey GJ. Therapeutic benefits of nitric oxide in lung transplantation. Biomed Pharmacother 2023; 167:115549. [PMID: 37734260 DOI: 10.1016/j.biopha.2023.115549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023] Open
Abstract
Lung transplantation is an evolutionary procedure from its experimental origin in the twentieth century and is now recognized as an established and routine life-saving intervention for a variety of end-stage pulmonary diseases refractory to medical management. Despite the success and continuous refinement in lung transplantation techniques, the widespread application of this important life-saving intervention is severely hampered by poor allograft quality offered from donors-after-brain-death. This has necessitated the use of lung allografts from donors-after-cardiac-death (DCD) as an additional source to expand the pool of donor lungs. Remarkably, the lung exhibits unique properties that may make it ideally suitable for DCD lung transplantation. However, primary graft dysfunction (PGD), allograft rejection and other post-transplant complications arising from unavoidable ischemia-reperfusion injury (IRI) of transplanted lungs, increase morbidity and mortality of lung transplant recipients annually. In the light of this, nitric oxide (NO), a selective pulmonary vasodilator, has been identified as a suitable agent that attenuates lung IRI and prevents PGD when administered directly to lung donors prior to donor lung procurement, or to recipients during and after transplantation, or administered indirectly by supplementing lung preservation solutions. This review presents a historical account of clinical lung transplantation and discusses the lung as an ideal organ for DCD. Next, the author highlights IRI and its clinical effects in lung transplantation. Finally, the author discusses preservation solutions suitable for lung transplantation, and the protective effects and mechanisms of NO in experimental and clinical lung transplantation.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Accra College of Medicine, Magnolia St, JVX5+FX9, East Legon, Accra, Ghana.
| |
Collapse
|
23
|
Atamer Y, Sarac S, Asık HK, Sahbaz T. Serum paraoxonase activities, nitric oxide, and malondialdehyde levels are altered in patients with primary fibromyalgia syndrome. Ir J Med Sci 2023; 192:2541-2547. [PMID: 36689069 DOI: 10.1007/s11845-023-03280-2] [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: 10/04/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND Fibromyalgia patients who are exposed to extreme oxidative stress may face more severe clinical features or oxidative stress may be increased by the severity of the disease. AIM The purpose of these investigation were to determine serum paraoxonase activities (PON-1) and nitric oxide (NO) activities and malondialdehyde (MDA) level in fibromyalgia and whether there were any associations between these enzymes activities, MDA level, and clinical parameters. METHODS The study groups were consisted of 30 primer fibromyalgia patients and 30 healthy subjects. Clinical findings, pain severity, functional disability, general health status, anxiety, and depression assessed, and serum PON-1 activity, MDA, and NO levels were measured. RESULTS The primer fibromyalgia group had significantly higher MDA, low density lipoprotein-cholesterol (LDL-C), and decreased PON-1 activity, NO, and high density lipoprotein-cholesterol (HDL-C) with respect to controls. The paraoxonase activity was negatively correlated with MDA, LDL-C, Visual Analog Scale (VAS), Fibromyalgia Impact Questionnaire score (FIQ score), tender point score, age, and BDI score, while positively correlated with NO and HDL-C. MDA level was positively correlated with VAS, FIQ score, tender point score, age, and negatively correlated with NO level. CONCLUSION These results suggest that FMS patients have an alteration in levels of MDA, NO, and PON-1 activities. We think that impaired oxidant/antioxidant status may affect the symptoms of the disease. Also, they may be of importance in the complex physiopathologic mechanism behind the development of FMS.
Collapse
Affiliation(s)
- Yildiz Atamer
- Department of Medical Biochemistry, Faculty of Medicine, Beykent University, Istanbul, Turkey
| | - Serdar Sarac
- Department of Physical Medicine and Rehabilitation, Private Erenköy Physical Therapy Center, Istanbul, Turkey
| | - Hatice Kubra Asık
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Beykent University, Istanbul, Turkey
| | - Tugba Sahbaz
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Beykent University, Istanbul, Turkey.
| |
Collapse
|
24
|
Kwok C, Nolan M. Cardiotoxicity of anti-cancer drugs: cellular mechanisms and clinical implications. Front Cardiovasc Med 2023; 10:1150569. [PMID: 37745115 PMCID: PMC10516301 DOI: 10.3389/fcvm.2023.1150569] [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: 01/24/2023] [Accepted: 07/17/2023] [Indexed: 09/26/2023] Open
Abstract
Cardio-oncology is an emerging field that seeks to enhance quality of life and longevity of cancer survivors. It is pertinent for clinicians to understand the cellular mechanisms of prescribed therapies, as this contributes to robust understanding of complex treatments and off-target effects, improved communication with patients, and guides long term care with the goal to minimise or prevent cardiovascular complications. Our aim is to review the cellular mechanisms of cardiotoxicity involved in commonly used anti-cancer treatments and identify gaps in literature and strategies to mitigate cardiotoxicity effects and guide future research endeavours.
Collapse
Affiliation(s)
- Cecilia Kwok
- Department of Medicine, Western Health, Melbourne, VIC, Australia
| | - Mark Nolan
- Department of Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Cardiovascular Imaging, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Pappa M, Ntouros PA, Papanikolaou C, Sfikakis PP, Souliotis VL, Tektonidou MG. Augmented oxidative stress, accumulation of DNA damage and impaired DNA repair mechanisms in thrombotic primary antiphospholipid syndrome. Clin Immunol 2023; 254:109693. [PMID: 37454866 DOI: 10.1016/j.clim.2023.109693] [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: 01/19/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Antiphospholipid syndrome (APS) is a rare autoimmune disorder with complex pathogenesis. Studies have shown that oxidative stress may contribute to APS pathophysiology. In peripheral blood mononuclear cells (PBMCs) from thrombotic Primary APS (thrPAPS) patients and age/sex-matched healthy controls (HC), as well as a control group of asymptomatic antiphospholipid antibody (aPL) positive individuals without APS (aPL+/non-APS), we examined oxidative stress, abasic (apurinic/apyrimidinic) sites, and DNA damage response (DDR)-associated parameters, including endogenous DNA damage (single- and double-strand breaks) and DNA repair mechanisms, namely nucleotide excision repair (NER) and double-strand breaks repair (DSB/R). We found that thrPAPS patients exhibited significantly higher levels of endogenous DNA damage, increased oxidative stress and abasic sites, as well as lower NER and DSB/R capacities versus HC (all P < 0.001) and versus aPL+/non-APS subjects (all P < 0.05). Our findings demonstrate that oxidative stress and decreased DNA repair mechanisms contribute to the accumulation of endogenous DNA damage in PBMCs from thrPAPS patients and, if further validated, may be exploited as therapeutic targets and potential biomarkers.
Collapse
Affiliation(s)
- Maria Pappa
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis A Ntouros
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Petros P Sfikakis
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis L Souliotis
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
26
|
Orrico F, Laurance S, Lopez AC, Lefevre SD, Thomson L, Möller MN, Ostuni MA. Oxidative Stress in Healthy and Pathological Red Blood Cells. Biomolecules 2023; 13:1262. [PMID: 37627327 PMCID: PMC10452114 DOI: 10.3390/biom13081262] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Red cell diseases encompass a group of inherited or acquired erythrocyte disorders that affect the structure, function, or production of red blood cells (RBCs). These disorders can lead to various clinical manifestations, including anemia, hemolysis, inflammation, and impaired oxygen-carrying capacity. Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the antioxidant defense mechanisms, plays a significant role in the pathophysiology of red cell diseases. In this review, we discuss the most relevant oxidant species involved in RBC damage, the enzymatic and low molecular weight antioxidant systems that protect RBCs against oxidative injury, and finally, the role of oxidative stress in different red cell diseases, including sickle cell disease, glucose 6-phosphate dehydrogenase deficiency, and pyruvate kinase deficiency, highlighting the underlying mechanisms leading to pathological RBC phenotypes.
Collapse
Affiliation(s)
- Florencia Orrico
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Sandrine Laurance
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| | - Ana C. Lopez
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Sophie D. Lefevre
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| | - Leonor Thomson
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Matias N. Möller
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Mariano A. Ostuni
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| |
Collapse
|
27
|
Gao M, Li J, Li N, Li W, Zhang S, Zhang T, Wang H, Fang Z, Yu Z, Hu G, Leng J, Yang X. Circulating hyaluronidase in early pregnancy and increased risk of gestational diabetes in Chinese pregnant women: A nested case control study. Clin Chim Acta 2023; 548:117512. [PMID: 37598741 DOI: 10.1016/j.cca.2023.117512] [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: 05/28/2023] [Revised: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND AND AIMS To explore association of serum hyaluronidase 1 (HYAL1) level in early pregnancy with gestational diabetes mellitus (GDM), and to examine interactive effects of HYAL1 with ceramides species on GDM risk. MATERIALS AND METHODS We conducted a 1:1 matched case-control study (n = 414) of pregnant women from 2010 to 2012 in Tianjin, China. Blood samples were collected at the first antenatal care visit (at a median of 10th gestational weeks). Binary conditional logistic regression and restricted cubic spline (RCS) analysis were used to examine full-range risk association between HYAL1 and GDM. Additive interactions and multiplicative interactions were employed to test interactive effects of HYAL1 with ceramides species on GDM risk. RESULTS Ln HYAL1 was linearly associated with GDM risk and the adjusted OR of HYAL1 ≥ vs. < its median for GDM was significant (1.65, 95%CI: 1.08-2.52). High HYAL1 markedly enhanced the ORs of high ceramide 18:0 for GDM from 2.31 (1.06-5.01) to 6.74 (2.85-16.0), and low ceramide 24:0 from 3.08 (1.33-7.11) to 8.15 (3.03-21.9), with significant additive interactions. CONCLUSIONS High HYAL1 in early pregnancy may increase the risk of GDM in Chinese women, possibly via enhancing the effects of high ceramide 18:0 and low ceramide 24:0 on GDM risk.
Collapse
Affiliation(s)
- Ming Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Weiqin Li
- Project Office, Tianjin Women and Children's Health Center, Tianjin 300070, China
| | - Shuang Zhang
- Project Office, Tianjin Women and Children's Health Center, Tianjin 300070, China
| | - Tao Zhang
- Project Office, Tianjin Women and Children's Health Center, Tianjin 300070, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Zhongze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, 15000 Halifax, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Junhong Leng
- Project Office, Tianjin Women and Children's Health Center, Tianjin 300070, China.
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China.
| |
Collapse
|
28
|
Kerget F, Kerget B. Can Fractional Exhaled Nitric Oxide (FeNO) Serve as a Clinical Indicator for Patients Hospitalized with Crimean-Congo Hemorrhagic Fever? Jpn J Infect Dis 2023; 76:226-232. [PMID: 37005275 DOI: 10.7883/yoken.jjid.2022.470] [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] [Indexed: 04/03/2023]
Abstract
Crimean-Congo hemorrhagic fever (CCHF), a zoonotic disease spread by infected viruses, can be a significant cause of morbidity and mortality in endemic areas. This prospective study aimed to establish the relationship between fractional exhaled nitric oxide (FeNO) levels and clinical prognosis of CCHF. The study included 85 participants: 55 patients followed up for CCHF from May to August 2022, and 30 healthy controls. FeNO levels were measured upon hospital admission and were 7.6 ± 3.3 parts per billion (ppb) in patients with mild/moderate CCHF, 2.5 ± 2.1 ppb in patients with severe CCHF, and 6.7 ± 1.7 ppb in the healthy control group. There was no statistically significant difference in FeNO levels between the control group and patients with mild/moderate CCHF (P = 0.09), whereas patients with severe CCHF had lower FeNO levels than those in the control group and patients with mild/moderate CCHF (P < 0.001 for both). FeNO measurement may offer a noninvasive and easily applied approach for predicting the clinical course and prognosis of CCHF in the early stages of the disease.
Collapse
Affiliation(s)
- Ferhan Kerget
- Department of Infectious Diseases and Clinical Microbiology, Erzurum Regional Training and Research Hospital, University of Health Sciences, Turkey
| | - Buğra Kerget
- Department of Pulmonary Diseases, Faculty of Medicine, Ataturk University, Turkey
| |
Collapse
|
29
|
Jayant G, Kuperberg S, Somnay K, Wadgaonkar R. The Role of Sphingolipids in Regulating Vascular Permeability in Idiopathic Pulmonary Fibrosis. Biomedicines 2023; 11:1728. [PMID: 37371823 DOI: 10.3390/biomedicines11061728] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 05/29/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease that causes scarring and fibrotic transformation of the lung parenchyma, resulting in the progressive loss of respiratory function and, often, death. Current treatments that target profibrotic factors can slow the rate of progression but are unable to ultimately stop it. In the past decade, many studies have shown that increased vascular permeability may be both a predictive and perpetuating factor in fibrogenesis. Consequently, there is a search for therapeutic targets to try and modulate vascular permeability in fibrotic lungs. One such class of targets that show great promise is sphingolipids. Sphingolipids are common in cell membranes and are increasingly recognized as critical to many cell signaling pathways, including those that affect the integrity of the vascular endothelial barrier. In this focused review we look at sphingolipids, particularly the sphingosine-1-phosphate (S1P) axis and its effects on vascular permeability, and how those effects may affect the pathogenesis of IPF. We further examine existing S1P modulators and their potential efficacy as therapeutics for IPF.
Collapse
Affiliation(s)
- Girish Jayant
- SUNY Downstate College of Medicine, Brooklyn, NY 11203, USA
| | | | - Kaumudi Somnay
- NY Presbyterian Hospital Queens, New York, NY 11355, USA
| | - Raj Wadgaonkar
- SUNY Downstate College of Medicine, Brooklyn, NY 11203, USA
| |
Collapse
|
30
|
Bao J, Gan X, Feng W, Li Y, Qiu Y, Zhou M, Guo J, He L. Abnormal flow pattern of low wall shear stress and high oscillatory shear index in spontaneous vertebral artery dissection with vertebral artery hypoplasia. Front Neurosci 2023; 17:1179963. [PMID: 37389359 PMCID: PMC10303804 DOI: 10.3389/fnins.2023.1179963] [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: 03/05/2023] [Accepted: 05/26/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction Spontaneous vertebral artery dissection (sVAD) might tend to develop in vertebral artery hypoplasia (VAH) with hemodynamic dysfunction and it is crucial to assess hemodynamics in sVAD with VAH to investigate this hypothesis. This retrospective study aimed to quantify hemodynamic parameters in patients with sVAD with VAH. Methods Patients who had suffered ischemic stroke due to an sVAD of VAH were enrolled in this retrospective study. The geometries of 14 patients (28 vessels) were reconstructed using Mimics and Geomagic Studio software from CT angiography (CTA). ANSYS ICEM and ANSYS FLUENT were utilized for mesh generation, set boundary conditions, solve governing equations, and perform numerical simulations. Slices were obtained at the upstream area, dissection or midstream area and downstream area of each VA. The blood flow patterns were visualized through instantaneous streamline and pressure at peak systole and late diastole. The hemodynamic parameters included pressure, velocity, time-averaged blood flow, time-averaged wall shear stress (TAWSS), oscillatory shear index (OSI), endothelial cell action potential (ECAP), relative residence time (RRT) and time-averaged nitric oxide production rate (TARNO). Results Significant focal increased velocity was present in the dissection area of steno-occlusive sVAD with VAH compared to other nondissected areas (0.910 m/s vs. 0.449 vs. 0.566, p < 0.001), while focal slow flow velocity was observed in the dissection area of aneurysmal dilatative sVAD with VAH according to velocity streamlines. Steno-occlusive sVAD with VAH arteries had a lower time-averaged blood flow (0.499 cm3/s vs. 2.268, p < 0.001), lower TAWSS (1.115 Pa vs. 2.437, p = 0.001), higher OSI (0.248 vs. 0.173, p = 0.006), higher ECAP (0.328 Pa-1 vs. 0.094, p = 0.002), higher RRT (3.519 Pa-1 vs. 1.044, p = 0.001) and deceased TARNO (104.014 nM/s vs. 158.195, p < 0.001) than the contralateral VAs. Conclusion Steno-occlusive sVAD with VAH patients had abnormal blood flow patterns of focal increased velocity, low time-averaged blood flow, low TAWSS, high OSI, high ECAP, high RRT and decreased TARNO. These results provide a good basis for further investigation of sVAD hemodynamics and support the applicability of the CFD method in testing the hemodynamic hypothesis of sVAD. More detailed hemodynamic conditions with different stages of sVAD are warranted in the future.
Collapse
Affiliation(s)
- Jiajia Bao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinling Gan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Wentao Feng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University) Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yanbo Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Qiu
- Department of Applied Mechanics, Sichuan University, Chengdu, China
| | - Muke Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Guo
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Li He
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Rajendra A, Bondonno NP, Murray K, Zhong L, Rainey-Smith SR, Gardener SL, Blekkenhorst LC, Ames D, Maruff P, Martins RN, Hodgson JM, Bondonno CP. Habitual dietary nitrate intake and cognition in the Australian Imaging, Biomarkers and Lifestyle Study of ageing: A prospective cohort study. Clin Nutr 2023; 42:1251-1259. [PMID: 37331149 DOI: 10.1016/j.clnu.2023.05.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/09/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND & AIMS Dietary nitrate improves cardiovascular health via a nitric oxide (NO) pathway. NO is key to both cardiovascular and brain health. There is also a strong association between vascular risk factors and brain health. Dietary nitrate intake could therefore be associated with better cognitive function and reduced risk of cognitive decline. This is yet to be investigated. The aim of this study was to investigate the association between habitual intake of dietary nitrate from sources where nitrate is naturally present, and cognitive function, and cognitive decline, in the presence or absence of the apolipoprotein E (APOE) ε4 allele. METHODS The study included 1254 older adult participants of the Australian Imaging, Biomarkers and Lifestyle Study of Ageing who were cognitively normal at baseline. Plant-derived, vegetable-derived, animal derived nitrate (not including meat where nitrate is an allowed additive), and total nitrate intakes were calculated from baseline food frequency questionnaires using comprehensive nitrate databases. Cognition was assessed at baseline and every 18 months over a follow-up period of 126 months using a comprehensive neuropsychological test battery. Multivariable-adjusted linear mixed effect models were used to examine the association between baseline nitrate intake and cognition over the 126 months (median [IQR] follow-up time of 36 [18-72] months), stratified by APOE ε4 carrier status. RESULTS In non APOE ε4 carriers, for every 60 mg/day higher intake of plant-derived nitrate at baseline there was an associated higher language score [β (95% CI): 0.10 (0.01, 0.19)] over 126 months, after multivariable adjustments. In APOE ε4 carriers, there was an associated better episodic recall memory [0.24 (0.08, 0.41)] and recognition memory [0.15 (0.01, 0.30)] scores. Similar associations were seen for the intakes of vegetable-derived and total nitrate. Additionally, in APOE ε4 carriers, for every 6 mg/day higher intake of animal-derived nitrate (excluding meat with nitrate as an allowed additive) at baseline there was an associated higher executive function score [β (95% CI): 1.41 (0.42, 2.39)]. We did not find any evidence of an association between dietary nitrate intake and rate of cognitive decline. CONCLUSION Our results suggest that habitual intake of dietary nitrate from sources where nitrate is naturally present impacts cognitive performance in an APOE genotype contingent manner. Further work is needed to validate our findings and understand potential mechanisms underlying the observed effects.
Collapse
Affiliation(s)
- Anjana Rajendra
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Nicola P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; The Danish Cancer Society Research Centre, Copenhagen, Denmark
| | - Kevin Murray
- School of Population and Global Health, University of Western Australia, Perth, Western Australia, Australia
| | - Liezhou Zhong
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia; Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Perth, Western Australia, Australia
| | - Samantha L Gardener
- Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia
| | - Lauren C Blekkenhorst
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - David Ames
- National Ageing Research Institute, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Victoria, Australia
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia; Cogstate Ltd, Melbourne, Victoria, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Jonathan M Hodgson
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia
| | - Catherine P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia.
| |
Collapse
|
32
|
Dutta B, Loo S, Kam A, Sze SK, Tam JP. Ginsentide TP1 Protects Hypoxia-Induced Dysfunction and ER Stress-Linked Apoptosis. Cells 2023; 12:1401. [PMID: 37408235 PMCID: PMC10216702 DOI: 10.3390/cells12101401] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Hypoxia-induced vascular endothelial dysfunction (VED) is a significant contributor to several severe human diseases, including heart disease, stroke, dementia, and cancer. However, current treatment options for VED are limited due to the lack of understanding of the underlying disease mechanisms and therapeutic leads. We recently discovered a heat-stable microprotein in ginseng, called ginsentide TP1, that has been shown to reduce vascular dysfunction in cardiovascular disease models. In this study, we use a combination of functional assays and quantitative pulsed SILAC proteomics to identify new proteins synthesized in hypoxia and to show that ginsentide TP1 provides protection for human endothelial cells against hypoxia and ER stress. Consistent with the reported findings, we also found that hypoxia activates various pathways related to endothelium activation and monocyte adhesion, which in turn, impairs nitric oxide (NO) synthase activity, reduces the bioavailability of NO, and increases the production of reactive oxygen species that contribute to VED. Additionally, hypoxia triggers endoplasmic reticulum stress and initiates apoptotic signaling pathways associated with cardiovascular pathology. Treatment with ginsentide TP1 reduced surface adhesion molecule expression, prevented activation of the endothelium and leukocyte adhesion, restored protein hemostasis, and reduced ER stress to protect against hypoxia-induced cell death. Ginsentide TP1 also restored NO signaling and bioavailability, reduced oxidative stress, and protected endothelial cells from endothelium dysfunction. In conclusion, this study shows that the molecular pathogenesis of VED induced by hypoxia can be mitigated by treatment with ginsentide TP1, which could be one of the key bioactive compounds responsible for the "cure-all" effect of ginseng. This research may lead to the development of new therapies for cardiovascular disorders.
Collapse
Affiliation(s)
- Bamaprasad Dutta
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
| | - Shining Loo
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
- Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Antony Kam
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
- Department of Health Sciences, Brock University, Niagara Region, St. Catharines, ON L2S 3A1, Canada
| | - James P. Tam
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; (B.D.); (S.L.); (A.K.); (S.K.S.)
| |
Collapse
|
33
|
Gotardo ÉMF, Brito PL, Gushiken LFS, Chweih H, Leonardo FC, Costa FF, Conran N. Molecular and cellular effects of in vivo chronic intravascular hemolysis and anti-inflammatory therapeutic approaches. Vascul Pharmacol 2023; 150:107176. [PMID: 37116732 DOI: 10.1016/j.vph.2023.107176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
Intravascular hemolysis (IVH) occurs in numerous inherited and acquired disorders, including sickle cell disease (SCD), malaria and sepsis. These diseases display unique symptoms, but often share complications, such as vasomotor dysfunction and pulmonary hypertension. Consequently, in vivo models are needed to study the effects of continuous intravascular hemolytic processes, independently of the molecular or extrinsic alteration that leads to erythrocyte destruction. We gave twice-weekly low-dose phenylhydrazine (LDPHZ) to C57BL/6 J mice for 4 weeks, and measured parameters indicative of anemia, hemoglobin-clearance pathways, inflammation and iron turnover, comparing these to those of a murine model of SCD, which displays associated IVH. LDPHZ administration provoked discreet anemia in mice and significant reticulocytosis, in association with hemoglobin/heme-clearance pathway protein depletion. Mice subjected to chronic hemolysis displayed elevated leukocyte counts and plasma levels of interleukin (IL)-1β, TNF-α, IL-6, soluble ICAM-1, endothelin-1 and anti-inflammatory IL-10, closely emulating alterations indicative of systemic inflammatory and endothelial activation in SCD, and confirming chronic IVH in itself as a serious complication. Discreet accelerations in hepatic and splenic iron turnover also occurred in LDPHZ mice, without alterations in liver damage markers. Examining the effects of two therapies on hemolysis-induced inflammation, the administration of hydroxyurea (and to a lesser extent, l-glutamine) significantly abrogated hemolytic inflammation in mice, without apparent inhibition of hemolysis. In conclusion, the isolation of chronic IVH, a common disease mechanism, using this model, may allow the study of hemolysis-specific sequelae at the cellular and systemic level, and the investigation of candidate agents that could potentially counter hemolytic inflammation.
Collapse
Affiliation(s)
- Érica M F Gotardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| | - Pâmela L Brito
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Lucas F S Gushiken
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Hanan Chweih
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Flavia C Leonardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Fernando F Costa
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Nicola Conran
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
34
|
Kamenshchikov NO, Duong N, Berra L. Nitric Oxide in Cardiac Surgery: A Review Article. Biomedicines 2023; 11:1085. [PMID: 37189703 PMCID: PMC10135597 DOI: 10.3390/biomedicines11041085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
Perioperative organ injury remains a medical, social and economic problem in cardiac surgery. Patients with postoperative organ dysfunction have increases in morbidity, length of stay, long-term mortality, treatment costs and rehabilitation time. Currently, there are no pharmaceutical technologies or non-pharmacological interventions that can mitigate the continuum of multiple organ dysfunction and improve the outcomes of cardiac surgery. It is essential to identify agents that trigger or mediate an organ-protective phenotype during cardiac surgery. The authors highlight nitric oxide (NO) ability to act as an agent for perioperative protection of organs and tissues, especially in the heart-kidney axis. NO has been delivered in clinical practice at an acceptable cost, and the side effects of its use are known, predictable, reversible and relatively rare. This review presents basic data, physiological research and literature on the clinical application of NO in cardiac surgery. Results support the use of NO as a safe and promising approach in perioperative patient management. Further clinical research is required to define the role of NO as an adjunct therapy that can improve outcomes in cardiac surgery. Clinicians also have to identify cohorts of responders for perioperative NO therapy and the optimal modes for this technology.
Collapse
Affiliation(s)
- Nikolay O. Kamenshchikov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Nicolette Duong
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Respiratory Care Service, Patient Care Services, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lorenzo Berra
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Respiratory Care Service, Patient Care Services, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
35
|
Hirschberger S, Schmid A, Kreth S. [Immunomodulation by nutritional intervention in critically ill patients]. DIE ANAESTHESIOLOGIE 2023; 72:229-244. [PMID: 36797533 PMCID: PMC9934515 DOI: 10.1007/s00101-023-01258-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 04/12/2023]
Abstract
Critically ill patients often suffer from a complex and severe immunological dysfunction. The differentiation and function of human immune cells are fundamentally controlled through metabolic processes. New concepts of immunonutrition therefore try to use enteral and parenteral nutrition to positively impact on the immune function of intensive care unit patients. This review article concisely presents the currently available evidence on the commonly used isolated supplements (anti-oxidative substances, amino acids, essential fatty acids) and difficulties related to their clinical use. The second part presents new and more comprehensive concepts of immunonutrition to influence the intestinal microbiome and to modulate the macronutrient composition. Immunonutrition of critically ill patients bears enormous potential and could become a valuable clinical tool for modulation of the immunometabolism of intensive care unit patients.
Collapse
Affiliation(s)
- Simon Hirschberger
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland
| | - Annika Schmid
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland
| | - Simone Kreth
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland.
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland.
| |
Collapse
|
36
|
Vera OD, Wulff H, Braun AP. Endothelial KCa channels: Novel targets to reduce atherosclerosis-driven vascular dysfunction. Front Pharmacol 2023; 14:1151244. [PMID: 37063294 PMCID: PMC10102451 DOI: 10.3389/fphar.2023.1151244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
Elevated levels of cholesterol in the blood can induce endothelial dysfunction, a condition characterized by impaired nitric oxide production and decreased vasodilatory capacity. Endothelial dysfunction can promote vascular disease, such as atherosclerosis, where macrophages accumulate in the vascular intima and fatty plaques form that impair normal blood flow in conduit arteries. Current pharmacological strategies to treat atherosclerosis mostly focus on lipid lowering to prevent high levels of plasma cholesterol that induce endothelial dysfunction and atherosclerosis. While this approach is effective for most patients with atherosclerosis, for some, lipid lowering is not enough to reduce their cardiovascular risk factors associated with atherosclerosis (e.g., hypertension, cardiac dysfunction, stroke, etc.). For such patients, additional strategies targeted at reducing endothelial dysfunction may be beneficial. One novel strategy to restore endothelial function and mitigate atherosclerosis risk is to enhance the activity of Ca2+-activated K+ (KCa) channels in the endothelium with positive gating modulator drugs. Here, we review the mechanism of action of these small molecules and discuss their ability to improve endothelial function. We then explore how this strategy could mitigate endothelial dysfunction in the context of atherosclerosis by examining how KCa modulators can improve cardiovascular function in other settings, such as aging and type 2 diabetes. Finally, we consider questions that will need to be addressed to determine whether KCa channel activation could be used as a long-term add-on to lipid lowering to augment atherosclerosis treatment, particularly in patients where lipid-lowering is not adequate to improve their cardiovascular health.
Collapse
Affiliation(s)
- O. Daniel Vera
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, United States
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- *Correspondence: Andrew P. Braun,
| |
Collapse
|
37
|
Modulation of Endothelial Function by TMAO, a Gut Microbiota-Derived Metabolite. Int J Mol Sci 2023; 24:ijms24065806. [PMID: 36982880 PMCID: PMC10054148 DOI: 10.3390/ijms24065806] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Endothelial function is essential in the maintenance of systemic homeostasis, whose modulation strictly depends on the proper activity of tissue-specific angiocrine factors on the physiopathological mechanisms acting at both single and multi-organ levels. Several angiocrine factors take part in the vascular function itself by modulating vascular tone, inflammatory response, and thrombotic state. Recent evidence has outlined a strong relationship between endothelial factors and gut microbiota-derived molecules. In particular, the direct involvement of trimethylamine N-oxide (TMAO) in the development of endothelial dysfunction and its derived pathological outcomes, such as atherosclerosis, has come to light. Indeed, the role of TMAO in the modulation of factors strictly related to the development of endothelial dysfunction, such as nitric oxide, adhesion molecules (ICAM-1, VCAM-1, and selectins), and IL-6, has been widely accepted. The aim of this review is to present the latest studies that describe a direct role of TMAO in the modulation of angiocrine factors primarily involved in the development of vascular pathologies.
Collapse
|
38
|
In Search of the Holy Grail: Stem Cell Therapy as a Novel Treatment of Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2023; 24:ijms24054903. [PMID: 36902332 PMCID: PMC10003723 DOI: 10.3390/ijms24054903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Heart failure, a leading cause of hospitalizations and deaths, is a major clinical problem. In recent years, the increasing incidence of heart failure with preserved ejection fraction (HFpEF) has been observed. Despite extensive research, there is no efficient treatment for HFpEF available. However, a growing body of evidence suggests stem cell transplantation, due to its immunomodulatory effect, may decrease fibrosis and improve microcirculation and therefore, could be the first etiology-based therapy of the disease. In this review, we explain the complex pathogenesis of HFpEF, delineate the beneficial effects of stem cells in cardiovascular therapy, and summarize the current knowledge concerning cell therapy in diastolic dysfunction. Furthermore, we identify outstanding knowledge gaps that may indicate directions for future clinical studies.
Collapse
|
39
|
Abstract
While neutrophils are the main effectors of protective innate immune responses, they are also key players in inflammatory pathologies. Sickle cell disease (SCD) is a genetic blood disorder in which red blood cells (RBCs) are constantly destroyed in the circulation which generates a highly inflammatory environment that culminates in vascular occlusions. Vaso-occlusion is the hallmark of SCD and a predictor of disease severity. Neutrophils initiate and propagate SCD-related vaso-occlusion through adhesive interactions with the activated and dysfunctional endothelium, sickle RBCs, and platelets, leading to acute and chronic complications that progress to irreversible organ damage and ultimately death. The use of SCD humanized mouse models, in combination with in vivo imaging techniques, has emerged as a fundamental tool to understand the dynamics of neutrophils under complex inflammatory contexts and their contribution to vascular injury in SCD. In this review, we discuss the various mechanisms by which circulating neutrophils sense and respond to the wide range of stimuli present in the blood of SCD patients and mice. We argue that the central role of neutrophils in SCD can be rationalized to develop targets for the management of clinical complications in SCD patients.
Collapse
Affiliation(s)
- Lidiane S Torres
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
40
|
Wu J, Tang L, Du M, Li C, Kou H, Jiang H, Luo W, Zhang Y, Huang Z, Liao D, Xiong W, Mei H, Hu Y. Plasma proteomic and metabolomic signatures of B-ALL patients during CAR-T cell therapy. Clin Transl Med 2023; 13:e1225. [PMID: 36938989 PMCID: PMC10026086 DOI: 10.1002/ctm2.1225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/21/2023] Open
Affiliation(s)
- Jianghua Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Mengyi Du
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Chenggong Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Haiming Kou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Huiwen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Wenjing Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yinqiang Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Zhongpei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Danying Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Wei Xiong
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| |
Collapse
|
41
|
Rajendra A, Bondonno NP, Rainey-Smith SR, Gardener SL, Hodgson JM, Bondonno CP. Potential role of dietary nitrate in relation to cardiovascular and cerebrovascular health, cognition, cognitive decline and dementia: a review. Food Funct 2022; 13:12572-12589. [PMID: 36377891 DOI: 10.1039/d2fo02427f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
There is currently no effective treatment for dementia, of which Alzheimer's disease (AD) is the most common form. It is, therefore, imperative to focus on evidence-based preventive strategies to combat this extremely debilitating chronic disease. Nitric oxide (NO) is a key signalling molecule in the cardiovascular, cerebrovascular, and central nervous systems. Vegetables rich in nitrate, such as spinach and beetroot, are an important source of NO, with beneficial effects on validated markers of cardiovascular health and an association with a lower risk of cardiovascular disease. Given the link between cardiovascular disease risk factors and dementia, together with the important role of NO in vascular health and cognition, it is important to determine whether dietary nitrate could also improve cognitive function, markers of brain health, and lower risk of dementia. This review presents an overview of NO's role in the cardiovascular, cerebrovascular, and central nervous systems; an overview of the available evidence that nitrate, through effects on NO, improves cardiovascular health; and evaluates the current evidence regarding dietary nitrate's potential role in cerebrovascular health, cognitive function, and brain health assessed via biomarkers.
Collapse
Affiliation(s)
- Anjana Rajendra
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.
| | - Nicola P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia. .,Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia.,Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Perth, Western Australia, Australia
| | - Samantha L Gardener
- Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia.,Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia
| | - Jonathan M Hodgson
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia. .,Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia
| | - Catherine P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia. .,Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia
| |
Collapse
|
42
|
Rai P, Desai PC, Ataga KI. The Evolving Landscape of Drug Therapies for Sickle Cell Disease. Hematol Oncol Clin North Am 2022; 36:1285-1312. [DOI: 10.1016/j.hoc.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
43
|
Urbanowicz T, Olasińska-Wiśniewska A, Gładki M, Jemielity M. The Significance of Simple Inflammatory Markers in Off Pump Surgery-Review. Rev Cardiovasc Med 2022; 23:400. [PMID: 39076673 PMCID: PMC11270455 DOI: 10.31083/j.rcm2312400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 07/31/2024] Open
Abstract
The inflammatory background of coronary artery disease is gaining more attention in recent times. Off pump surgery is minimally invasive type of surgical revascularization with relatively low number of applications in cardiac surgery centers worldwide that allows for perioperative inflammatory reactions minimalization. The simple inflammatory markers (neutrophil to lymphocyte ratio (NLR), monocyte to lymphocyte ratio (MLR), platelets to lymphocyte ratio (PLR), systemic inflammatory index (SII), systemic inflammatory response index (SIRI), aggregate index of systemic inflammation (AISI)) possess a clinically significant impact on patients' prognosis and may help to improve patients' long-term results. The review presents the current knowledge regarding their utility in clinical practice. Assessment of inflammatory indices obtained from whole blood count analysis allows to indicate those patients who need scrupulous follow-up due to predicted worse long-term survival. Perioperative measurement and analysis of simple whole blood counts is inexpensive and easily available and may improve the results of surgical revascularization by better identification of patients at higher risk of worse outcomes.
Collapse
Affiliation(s)
- Tomasz Urbanowicz
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-848 Poznan, Poland
| | - Anna Olasińska-Wiśniewska
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-848 Poznan, Poland
| | - Marcin Gładki
- Pediatric Cardiac Surgery Department, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Marek Jemielity
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-848 Poznan, Poland
- Pediatric Cardiac Surgery Department, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| |
Collapse
|
44
|
Rizwan H, Kumar S, Kumari G, Pal A. High glucose-induced increasing reactive nitrogen species accumulation triggered mitochondrial dysfunction, inflammation, and apoptosis in keratinocytes. Life Sci 2022; 312:121208. [PMID: 36427546 DOI: 10.1016/j.lfs.2022.121208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022]
Abstract
Growing evidence indicates that skin injuries are a common complication of diabetes. However, the cellular and molecular mechanisms of high glucose (HG) environment trigger nitrosative stress-mediated inflammation and apoptosis in keratinocytes remains unknown. Here we investigated whether reactive nitrogen species (RNS) induced by HG environment restrain antioxidant activity, and mitochondrial dysfunction leading to inflammation, and apoptosis via stress signaling pathways in keratinocytes. Our results established that the HG environment enhanced the production of nitric oxide (NO) and peroxynitrite anion (ONOO-) by inducible NO synthase (iNOS) in keratinocytes. Overproduction of RNS in HG environment suppress the antioxidants activity leading to mitochondrial dysfunction, characterized by loss of mitochondrial membrane potential (ΔΨm), increase in mitochondrial mass, decrease in mitochondrial transcription factor A(TFAM), increase in mitochondrial DNA (mtDNA) displacement loop (D-loop) and decrease in glycolytic flux concentration, which was attenuated by pharmacological inhibitors of NO/ONOO-, Nω-Nitro-l-argininemethyl ester hydrochloride (NAME)/hydralazine hydrochloride (Hyd.HCl). Excess production of RNS in HG environment restrained 8-oxoguanine DNA glycosylase-1 (OGG1) expression and increased 8-hydroxydeoxyguanosine (8-OHdG) accumulations in DNA were regulated by NO or ONOO-. Further, HG-induced RNA production caused an increase in the production of inflammatory mediators accompanied by activation of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3 signaling cascade, lipid peroxidation (LPO), and protein carbonylation (PC) reactions followed by breakdown the cell-cell communication and apoptosis. Pre-treatment of cell with NAME/Hyd.HCl, diminished the expression of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3, inflammatory mediators, and attenuated apoptosis in keratinocytes. Together, our results indicated that excess production of RNS in HG environment triggered inflammation and apoptosis, mediated by activation of ERK1/2MAPK/Akt/tuberin-mTOR/IRF3 signaling cascades in keratinocytes.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Sonu Kumar
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Golden Kumari
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India
| | - Arttatrana Pal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India; Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar 845401, India.
| |
Collapse
|
45
|
Jia X, Zhu Y, Qi Y, Zheng R, Lin L, Hu C, Zhang Y, Wu X, Qi H, Wei R, Zhang J, Xu M, Xu Y, Wang T, Zhao Z, Chen Y, Bi Y, Wang W, Li M, Lu J. Association between triglyceride glucose index and carotid intima-media thickness in obese and nonobese adults. J Diabetes 2022; 14:596-605. [PMID: 36071605 PMCID: PMC9512765 DOI: 10.1111/1753-0407.13312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/13/2022] [Accepted: 08/23/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The triglyceride glucose (TyG) index is closely associated with subclinical atherosclerosis. However, the association remains inconclusive among obese and nonobese individuals. METHODS This prospective study was conducted in 5751 adults with normal carotid intima-media thickness (CIMT) at baseline. We divided the population into four groups based on the TyG index, which was calculated by the following formula: Ln (fasting triglycerides [mg/dL] × fasting glucose [mg/dL]/2). Information on CIMT was acquired by ultrasonography. Incident elevated CIMT was defined as IMT values greater than 0.9 mm at follow-up. Odds ratios (ORs) and 95% confidence intervals (CIs) of the associations between TyG index and elevated CIMT were estimated using multivariable logistic regression models. RESULTS After a median follow-up of 4.3 years, 722 (12.6%) individuals had progressed to elevated CIMT. Compared with the second quartile of the TyG index, the first and fourth quartile both conferred higher risks of elevated CIMT after adjusting for potential confounders. In the total population, the ORs for the first and fourth quartile were 1.29 (95% CI, 1.00-1.66) and 1.42 (95% CI, 1.11-1.83), respectively. Restricted cubic splines demonstrated an approximately U-shaped association between TyG index and elevated CIMT among the total and nonobese adults (P for nonlinearity <.05), but not in those with general or abdominal obesity. CONCLUSIONS A U-shaped association was observed between TyG index and elevated CIMT only among nonobese Chinese adults.
Collapse
Affiliation(s)
- Xiaojing Jia
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuanyue Zhu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Qi
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lin Lin
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chunyan Hu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xueyan Wu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hongyan Qi
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ran Wei
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min Xu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yu Xu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tiange Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuhong Chen
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yufang Bi
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weiqing Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mian Li
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jieli Lu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
46
|
Cardoso TC, Rocha MA, Monteiro MMLV, Alves VS, Savio LEB, Silva CLM. The blockage of downstream P2Y 2 receptor signaling inhibits the prostate cancer cell adhesion to endothelial cells. Life Sci 2022; 306:120793. [PMID: 35850244 DOI: 10.1016/j.lfs.2022.120793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/20/2022] [Accepted: 07/06/2022] [Indexed: 11/26/2022]
Abstract
AIMS Prostate cancer is the second most frequently malignancy in men worldwide. Most deaths are caused by metastasis, and tumor cell dissemination involves the interaction with endothelial cells. However, the endothelial cell signaling involved in such interaction is not entirely understood. The tumor microenvironment contains extracellular ATP, an endogenous agonist of the purinergic P2Y2 receptor (P2Y2R). P2Y2R signaling changes endothelial cell phenotype, which may be relevant to cancer pathophysiology. Therefore, we hypothesized that P2Y2R activation could favor the metastatic prostate cancer cells adhesion to endothelial cells. MAIN METHODS For adhesion assays, confluent endothelial cells EA.hy926 were treated with P2Y2R agonists before adding and imaging stained DU-145 cells. Alternatively, fluorescent probes and antibodies were used to determine intracellular endothelial Ca2+, nitric oxide (NO), and flow cytometry assays. KEY FINDINGS Endothelial P2Y2R activation with ATP, UTP, or the selective agonist 2-thio-UTP increased DU-145 cell adhesion to EA.hy926 cells. This effect required endothelial cell Ca2+ mobilization and relied on the endothelial expression of VCAM-1 and ICAM-1. Conversely, inhibiting this proadhesive endothelial phenotype could impair DU-145 cell adhesion. To evaluate this, we chose atorvastatin based on its notable improvement of endothelial cell dysfunction. Atorvastatin blocked UTP-induced DU-145 cell adhesion to endothelial cell monolayer in a NO-dependent manner, unveiling a P2Y2R and NO signaling crosstalk. SIGNIFICANCE Endothelial P2Y2R signaling contributes to the adhesion of metastatic prostate cancer cells suggesting that the downstream signaling blockade by statins could be a putative mechanism to reduce prostate cancer metastasis.
Collapse
Affiliation(s)
- Tassya Cataldi Cardoso
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Marianna Araujo Rocha
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Matheus M L V Monteiro
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Vinícius Santos Alves
- Laboratório de Imunofisiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Luiz Eduardo Baggio Savio
- Laboratório de Imunofisiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Claudia Lucia Martins Silva
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil.
| |
Collapse
|
47
|
Chaar D, Dumont BL, Vulesevic B, Neagoe PE, Räkel A, White M, Sirois MG. Neutrophils and Circulating Inflammatory Biomarkers in Diabetes Mellitus and Heart Failure With Preserved Ejection Fraction. Am J Cardiol 2022; 178:80-88. [PMID: 35811144 DOI: 10.1016/j.amjcard.2022.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/13/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by low-grade chronic inflammation, which could be exacerbated by type 2 diabetes mellitus (DM). We hypothesized that neutrophils in patients with DM and patients with HFpEF with/without DM contribute to low-grade inflammation through the release of pro-inflammatory cytokines. Venous blood was withdrawn from patients with DM (n = 22), HFpEF (n = 15), HFpEF with DM (n = 13), and healthy controls (CTL) (n = 21). Levels of circulating cytokines and in vitro cytokines released by isolated neutrophils were assessed by enzyme-linked immunosorbent assay. Compared with CTL, there was a significant decrease in circulating nitric oxide in patients with DM (p ≤0.001), HFpEF (p ≤0.05), and HFpEF with DM (p ≤0.001) up to 44%. Circulating soluble intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 levels increased (up to 2.5-fold and 1.9-fold, respectively; p ≤0.001) in patients with HFpEF and patients with HFpEF and DM, whereas soluble E-selectin only increased in patients with HFpEF and DM (1.4-fold, p ≤0.001). Circulating vascular endothelial growth factor levels were similar in CTL and patients with DM but were decreased in patients with HFpEF with/without DM (up to 94%; p ≤0.001). Circulating C-reactive protein, interleukin (IL)-8, IL-6, and IL-receptor antagonist increased in all patient groups with a maximum of 3.3-fold, 4.7-fold, 4.8-fold, and 1.6-fold, respectively, in patients with HFpEF and patients with DM. In vitro, lipopolysaccharide increased neutrophils IL-6 release from HFpEF with DM (3.7-fold; p ≤0.001), and IL-8 release from DM and HFpEF with DM versus CTL (2.8-fold and 10.1-fold, respectively; p ≤0.001). IL-1 receptor antagonist and vascular endothelial growth factor release from HFpEF neutrophils significantly decreased up to 87.0% and 92.2%, respectively, versus CTL. Neutrophils from patients with DM and HFpEF release more cytokines than CTL. This increase in pro-inflammatory status may explain the greater event rate in patients with HFpEF and DM.
Collapse
Affiliation(s)
- Diana Chaar
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Benjamin L Dumont
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
| | - Branka Vulesevic
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | | | - Agnès Räkel
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Research Center, Center Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Michel White
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Martin G Sirois
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
48
|
Rathjen T, Kunkemoeller B, Cederquist CT, Wang X, Lockhart SM, Patti JC, Willenbrock H, Olsen GS, Povlsen GK, Beck HC, Rasmussen LM, Li Q, Park K, King GL, Rask-Madsen C. Endothelial Cell Insulin Signaling Regulates CXCR4 (C-X-C Motif Chemokine Receptor 4) and Limits Leukocyte Adhesion to Endothelium. Arterioscler Thromb Vasc Biol 2022; 42:e217-e227. [PMID: 35652755 PMCID: PMC9371472 DOI: 10.1161/atvbaha.122.317476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND An activated, proinflammatory endothelium is a key feature in the development of complications of obesity and type 2 diabetes and can be caused by insulin resistance in endothelial cells. METHODS We analyzed primary human endothelial cells by RNA sequencing to discover novel insulin-regulated genes and used endothelial cell culture and animal models to characterize signaling through CXCR4 (C-X-C motif chemokine receptor 4) in endothelial cells. RESULTS CXCR4 was one of the genes most potently regulated by insulin, and this was mediated by PI3K (phosphatidylinositol 3-kinase), likely through FoxO1, which bound to the CXCR4 promoter. CXCR4 mRNA in CD31+ cells was 77% higher in mice with diet-induced obesity compared with lean controls and 37% higher in db/db mice than db/+ controls, consistent with upregulation of CXCR4 in endothelial cell insulin resistance. SDF-1 (stromal cell-derived factor-1)-the ligand for CXCR4-increased leukocyte adhesion to cultured endothelial cells. This effect was lost after deletion of CXCR4 by gene editing while 80% of the increase was prevented by treatment of endothelial cells with insulin. In vivo microscopy of mesenteric venules showed an increase in leukocyte rolling after intravenous injection of SDF-1, but most of this response was prevented in transgenic mice with endothelial overexpression of IRS-1 (insulin receptor substrate-1). CONCLUSIONS Endothelial cell insulin signaling limits leukocyte/endothelial cell interaction induced by SDF-1 through downregulation of CXCR4. Improving insulin signaling in endothelial cells or inhibiting endothelial CXCR4 may reduce immune cell recruitment to the vascular wall or tissue parenchyma in insulin resistance and thereby help prevent several vascular complications.
Collapse
Affiliation(s)
- Thomas Rathjen
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.).,Novo Nordisk A/S, Måløv, Denmark (T.R., H.W., G.S.O., G.K.P.)
| | - Britta Kunkemoeller
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Carly T Cederquist
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Xuanchun Wang
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Sam M Lockhart
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - James C Patti
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | | | | | | | | | | | - Qian Li
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Kyoungmin Park
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - George L King
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Christian Rask-Madsen
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| |
Collapse
|
49
|
Shearston K, Tan JTM, Cochran BJ, Rye KA. Inhibition of Vascular Inflammation by Apolipoprotein A-IV. Front Cardiovasc Med 2022; 9:901408. [PMID: 35845068 PMCID: PMC9279673 DOI: 10.3389/fcvm.2022.901408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
Background Apolipoprotein (apo) A-IV, the third most abundant apolipoprotein in human high density lipoproteins (HDLs), inhibits intestinal and systemic inflammation. This study asks if apoA-IV also inhibits acute vascular inflammation. Methods Inflammation was induced in New Zealand White rabbits by placing a non-occlusive silastic collar around the common carotid artery. A single 1 mg/kg intravenous infusion of lipid-free apoA-IV or saline (control) was administered to the animals 24 h before collar insertion. The animals were euthanised 24 h post-collar insertion. Human coronary artery cells (HCAECs) were pre-incubated with reconstituted HDLs containing apoA-IV complexed with phosphatidylcholine, (A-IV)rHDLs, then activated by incubation with tumour necrosis factor (TNF)-α. Cell surface vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in the TNF-α-activated HCAECs was quantified by flow cytometry. VCAM-1, ICAM-1 and 3β-hydroxysteroid-Δ24 reductase (DHCR24) mRNA levels were quantified by real time PCR. Results Apolipoprotein ApoA-IV treatment significantly decreased collar-induced endothelial expression of VCAM-1, ICAM-1 and neutrophil infiltration into the arterial intima by 67.6 ± 9.9% (p < 0.01), 75.4 ± 6.9% (p < 0.01) and 74.4 ± 8.5% (p < 0.05), respectively. It also increased endothelial expression of DHCR24 by 2.6-fold (p < 0.05). Pre-incubation of HCAECs with (A-IV)rHDLs prior to stimulation with TNF-α inhibited VCAM-1 and ICAM-1 protein levels by 62.2 ± 12.1% and 33.7 ± 5.7%, respectively. VCAM-1 and ICAM-1 mRNA levels were decreased by 55.8 ± 7.2% and 49.6 ± 7.9%, respectively, while DHCR24 mRNA expression increased by threefold. Transfection of HCAECs with DHCR24 siRNA attenuated the anti-inflammatory effects of (A-IV)rHDLs. Pre-incubation of TNF-α-activated HCAECs with (A-IV)rHDLs also inhibited nuclear translocation of the p65 subunit of nuclear factor-κB (NF-κB), and decreased IκBα phosphorylation. Conclusion These results indicate that apoA-IV inhibits vascular inflammation in vitro and in vivo by inhibiting NF-κB activation in a DHCR24-dependent manner.
Collapse
Affiliation(s)
- Kate Shearston
- Lipid Research Group, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Joanne T. M. Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Blake J. Cochran
- Lipid Research Group, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Kerry-Anne Rye,
| |
Collapse
|
50
|
Steudler J, Ecott T, Ivan DC, Bouillet E, Walthert S, Berve K, Dick TP, Engelhardt B, Locatelli G. Autoimmune neuroinflammation triggers mitochondrial oxidation in oligodendrocytes. Glia 2022; 70:2045-2061. [PMID: 35762739 PMCID: PMC9546135 DOI: 10.1002/glia.24235] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/04/2022] [Accepted: 06/16/2022] [Indexed: 11/06/2022]
Abstract
Oligodendrocytes (ODCs) are myelinating cells of the central nervous system (CNS) supporting neuronal survival. Oxidants and mitochondrial dysfunction have been suggested as the main causes of ODC damage during neuroinflammation as observed in multiple sclerosis (MS). Nonetheless, the dynamics of this process remain unclear, thus hindering the design of neuroprotective therapeutic strategies. To decipher the spatio-temporal pattern of oxidative damage and dysfunction of ODC mitochondria in vivo, we created a novel mouse model in which ODCs selectively express the ratiometric H2 O2 biosensor mito-roGFP2-Orp1 allowing for quantification of redox changes in their mitochondria. Using 2-photon imaging of the exposed spinal cord, we observed significant mitochondrial oxidation in ODCs upon induction of the MS model experimental autoimmune encephalomyelitis (EAE). This redox change became already apparent during the preclinical phase of EAE prior to CNS infiltration of inflammatory cells. Upon clinical EAE development, mitochondria oxidation remained detectable and was associated with a significant impairment in organelle density and morphology. These alterations correlated with the proximity of ODCs to inflammatory lesions containing activated microglia/macrophages. During the chronic progression of EAE, ODC mitochondria maintained an altered morphology, but their oxidant levels decreased to levels observed in healthy mice. Taken together, our study implicates oxidative stress in ODC mitochondria as a novel pre-clinical sign of MS-like inflammation and demonstrates that evolving redox and morphological changes in mitochondria accompany ODC dysfunction during neuroinflammation.
Collapse
Affiliation(s)
- Jasmin Steudler
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Timothy Ecott
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Daniela C Ivan
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Elisa Bouillet
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | - Kristina Berve
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | |
Collapse
|