1
|
Zhu Y, Chu Y, Lan Y, Wang S, Zhang Y, Liu Y, Wang X, Yu F, Ma X. Loss of Endothelial TRPC1 Induces Aortic Hypercontractility and Hypertension. Circ Res 2025. [PMID: 39912234 DOI: 10.1161/circresaha.124.325574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND The increasing prevalence of obesity-related cardiovascular diseases demands a better understanding of the contribution of different cell types to vascular function for developing new treatment strategies. Previous studies have established a fundamental role of TRPC1 (transient receptor potential channel canonical family member 1) in blood vessels. However, little is known about its functional roles within different cell types. METHODS We generated endothelial-specific TRPC1-deficient and knockin mice and analyzed their changes in vascular function under physiological and pathologically obese state. Wire myography, Ca2+ image, blood pressure measurements, RNA-sequencing analysis, liquid chromatography-mass spectrometry, immunoblotting, ELISA, luciferase reporter assay, and morphometric assessments were performed to unravel phenotype and molecular changes in response to the absence or presence of endothelial TRPC1. RESULTS Loss of endothelial TRPC1 reduced endothelial-dependent relaxation and exaggerated endothelial-dependent contraction in mouse aorta. As expected, loss of endothelial TRPC1 amplified blood pressure and decreased acetylcholine-induced intracellular Ca2+ concentration rise in the aorta. In endothelial-specific TRPC1-deficient mouse arteries, the mRNA profile identified upregulation of c-Fos. Blockade of c-Fos rescued the impaired vasomotor tone in the aorta of mice deficient in endothelial TRPC1. Endothelial TRPC1-regulated nitric oxide/endothelin-1 production is involved in vascular c-Fos expression. Moreover, knockin of endothelial TRPC1 ameliorated enhanced endothelial-dependent contraction and hypertension in obese mice which is related to alleviated endothelial endothelin-1/c-Fos production and smooth muscle contraction. CONCLUSIONS Our results identify endothelial TRPC1 as a previously unclear regulator of vascular changes and blood pressure in both physiological and pathologically obese state, and it is associated with nitric oxide/endothelin-1/c-Fos signaling.
Collapse
Affiliation(s)
- Yifei Zhu
- Wuxi School of Medicine, Jiangnan University, China
| | - Yuan Chu
- Wuxi School of Medicine, Jiangnan University, China
| | - Yihui Lan
- Wuxi School of Medicine, Jiangnan University, China
| | - Sheng Wang
- Wuxi School of Medicine, Jiangnan University, China
| | - Yizhi Zhang
- Wuxi School of Medicine, Jiangnan University, China
| | - Yuan Liu
- Wuxi School of Medicine, Jiangnan University, China
| | | | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, China
| |
Collapse
|
2
|
Rabbitt D, Villapún VM, Carter LN, Man K, Lowther M, O'Kelly P, Knowles AJ, Mottura A, Tang YT, Luerti L, Reed RC, Cox SC. Rethinking Biomedical Titanium Alloy Design: A Review of Challenges from Biological and Manufacturing Perspectives. Adv Healthc Mater 2025; 14:e2403129. [PMID: 39711273 PMCID: PMC11804846 DOI: 10.1002/adhm.202403129] [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: 08/20/2024] [Revised: 11/14/2024] [Indexed: 12/24/2024]
Abstract
Current biomedical titanium alloys have been repurposed from other industries, which has contributed to several biologically driven implant failure mechanisms. This review highlights the added value that may be gained by building an appreciation of implant biological responses at the onset of alloy design. Specifically, the fundamental mechanisms associated with immune response, angiogenesis, osseointegration and the potential threat of infection are discussed, including how elemental selection can modulate these pivotal systems. With a view to expedite inclusion of these interactions in alloy design criteria, methods to analyze these performance characteristics are also summarized. While machine learning techniques are being increasingly used to unearth complex relationships between alloying elements and material properties, much is still unknown about the correlation between composition and some bio-related properties. To bridge this gap, high-throughput methods are also reviewed to validate biological response along with cutting edge manufacturing approaches that may support rapid discovery. Taken together, this review encourages the alloy development community to rethink their approach to enable a new generation of biomedical implants intrinsically designed for a life in the body, including functionality to tackle biological challenges thereby offering improved patient outcomes.
Collapse
Affiliation(s)
- Daisy Rabbitt
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Victor M. Villapún
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Luke N. Carter
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Kenny Man
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht3508 GAThe Netherlands
- Regenerative Medicine Center UtrechtUniversity Medical Center UtrechtUtrecht3584 CTThe Netherlands
| | - Morgan Lowther
- Paihau‐Robinson Research InstituteVictoria University of WellingtonWellington5010New Zealand
| | - Paraic O'Kelly
- Center for the Accelerated Maturation of MaterialsDepartment of Materials Science and EngineeringThe Ohio State University1305 Kinnear RoadColumbusOH43212USA
| | | | - Alessandro Mottura
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
| | - Yuanbo T. Tang
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
| | - Lorenzo Luerti
- Alloyed LtdUnit 15, Oxford Industrial ParkYarntonOX5 1QUUK
| | - Roger C. Reed
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
- Department of MaterialsUniversity of OxfordParks RoadOxfordOX1 3PJUK
| | - Sophie C. Cox
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| |
Collapse
|
3
|
Santamaria JC, Chevallier J, Dutour L, Picart A, Kergaravat C, Cieslak A, Amrane M, Vincentelli R, Puthier D, Clave E, Sergé A, Cohen-Solal M, Toubert A, Irla M. RANKL treatment restores thymic function and improves T cell-mediated immune responses in aged mice. Sci Transl Med 2024; 16:eadp3171. [PMID: 39630886 DOI: 10.1126/scitranslmed.adp3171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/18/2024] [Indexed: 12/07/2024]
Abstract
Age-related thymic involution, leading to reduced T cell production, is one of the major causes of immunosenescence. This results in an increased susceptibility to cancers, infections, and autoimmunity and in reduced vaccine efficacy. Here, we identified that the receptor activator of nuclear factor κB (RANK)-RANK ligand (RANKL) axis in the thymus is altered during aging. Using a conditional transgenic mouse model, we demonstrated that endothelial cells depend on RANK signaling for their cellularity and functional maturation. Decreased RANKL availability during aging resulted in a decline in cellularity and function of both endothelial cells and thymic epithelial cells, contributing to thymic involution. We then found that, whereas RANKL neutralization in young mice mimicked thymic involution, exogenous RANKL treatment in aged mice restored thymic architecture as well as endothelial cell and epithelial cell abundance and functional properties. Consequently, RANKL improved T cell progenitor homing to the thymus and boosted T cell production. This cascade of events resulted in peripheral T cell renewal and effective antitumor and vaccine responses in aged mice. Furthermore, we conducted a proof-of-concept study that showed that RANKL stimulates endothelial cells and epithelial cells in human thymic organocultures. Overall, our findings suggest that targeting the RANK-RANKL axis through exogenous RANKL administration could represent a therapeutic strategy to rejuvenate thymic function and improve T cell immunity during aging.
Collapse
Affiliation(s)
- Jérémy C Santamaria
- Centre d'Immunologie de Marseille-Luminy, CIML, CNRS, INSERM, Aix-Marseille Université, Marseille, Turing Centre for Living Systems, 13288 Marseille Cedex 09, France
| | - Jessica Chevallier
- Centre d'Immunologie de Marseille-Luminy, CIML, CNRS, INSERM, Aix-Marseille Université, Marseille, Turing Centre for Living Systems, 13288 Marseille Cedex 09, France
| | - Léa Dutour
- Université de Paris Cité, Institut de Recherche Saint Louis, EMiLy, INSERM UMRS 1160, 75010 Paris, France
| | - Amandine Picart
- Université de Paris Cité, INSERM, UMR-S 1132 BIOSCAR, 75010 Paris, France
- Departement de Rhumatologie, Hôpital Lariboisière, AP-HP, 75010 Paris, France
| | - Camille Kergaravat
- Université de Paris Cité, Institut de Recherche Saint Louis, EMiLy, INSERM UMRS 1160, 75010 Paris, France
| | - Agata Cieslak
- Laboratoire d'Onco-Hematologie, Hôpital Necker Enfants Malades, AP-HP, 75015 Paris, France
- Université Paris Cité, CNRS, INSERM U1151, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | - Mourad Amrane
- Service de Chirurgie Cardiovasculaire, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Renaud Vincentelli
- Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257 CNRS-Aix-Marseille Université, 13288 Marseille Cedex 09, France
| | - Denis Puthier
- Theories and Approaches of Genomic Complexity (TAGC), Inserm U1090, Aix-Marseille University, 13288 Marseille Cedex 09, France
| | - Emmanuel Clave
- Université de Paris Cité, Institut de Recherche Saint Louis, EMiLy, INSERM UMRS 1160, 75010 Paris, France
| | - Arnauld Sergé
- Laboratoire Adhesion and Inflammation (LAI), CNRS, INSERM, Aix Marseille Université, Turing Centre for Living Systems, 13288 Marseille Cedex 09, France
| | - Martine Cohen-Solal
- Université de Paris Cité, INSERM, UMR-S 1132 BIOSCAR, 75010 Paris, France
- Departement de Rhumatologie, Hôpital Lariboisière, AP-HP, 75010 Paris, France
| | - Antoine Toubert
- Université de Paris Cité, Institut de Recherche Saint Louis, EMiLy, INSERM UMRS 1160, 75010 Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, 75010 Paris France
| | - Magali Irla
- Centre d'Immunologie de Marseille-Luminy, CIML, CNRS, INSERM, Aix-Marseille Université, Marseille, Turing Centre for Living Systems, 13288 Marseille Cedex 09, France
| |
Collapse
|
4
|
Peng X, Hu Y, Xu J, Chen L, Ren W, Cai W. Inverse association between serum klotho levels and C-reactive protein levels in the US population: a cross-sectional study. BMC Cardiovasc Disord 2024; 24:687. [PMID: 39614159 DOI: 10.1186/s12872-024-04375-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND The inverse relationship between serum Klotho levels and systemic inflammation, particularly C-reactive protein (CRP), has been suggested in limited studies. However, the association within a large and diverse population remains underexplored. METHODS We conducted a cross-sectional study using data from the National Health and Nutrition Examination Survey (NHANES) to investigate the association between serum Klotho levels and CRP among a nationally representative sample of the US population. Multiple linear regression analyses were performed to assess this relationship while adjusting for relevant covariates. Stratified analysis with interaction, restricted cubic splines (RCS) were employed to support the research objectives. RESULTS A total of 5901 participants had a mean age of 57.9 ± 11.0 years, with 49.4% of them being male and 50.6% of them being female. A negative association between serum Klotho and CRP was revealed in the fully adjusted model (β -0.26; 95% CI -0.41∼-0.11). When serum Klotho was taken as quartiles with Q1 as reference, the adjusted β that were lowest in Q4 were - 0.1 (95% CI -0.16∼-0.04, p-value = 0.002) in model 4, respectively. These statistics were robust in stratified analyses. CONCLUSION While our study demonstrates an inverse association between serum Klotho levels and CRP, suggesting a potential cardioprotective role of Klotho, it is important to note that our cross-sectional design does not permit the establishment of causality. Therefore, we cannot definitively conclude that increasing Klotho levels will directly reduce cardiovascular risk. Our findings do, however, highlight the need for further research to explore the potential of Klotho as a therapeutic target for cardiovascular health.
Collapse
Affiliation(s)
- Xuelan Peng
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yingjie Hu
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiarong Xu
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ling Chen
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wei Ren
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wenzhi Cai
- Department of Nursing, Shenzhen Hospital, Southern Medical University, Number 1333, Xinhu Road, Baoán District, Shenzhen, Guangdong, 518101, China.
- School of Nursing, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
5
|
Laowpanitchakorn P, Zeng J, Piantino M, Uchida K, Katsuyama M, Matsusaki M. Biofabrication of engineered blood vessels for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2330339. [PMID: 38633881 PMCID: PMC11022926 DOI: 10.1080/14686996.2024.2330339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 04/19/2024]
Abstract
To successfully engineer large-sized tissues, establishing vascular structures is essential for providing oxygen, nutrients, growth factors and cells to prevent necrosis at the core of the tissue. The diameter scale of the biofabricated vasculatures should range from 100 to 1,000 µm to support the mm-size tissue while being controllably aligned and spaced within the diffusion limit of oxygen. In this review, insights regarding biofabrication considerations and techniques for engineered blood vessels will be presented. Initially, polymers of natural and synthetic origins can be selected, modified, and combined with each other to support maturation of vascular tissue while also being biocompatible. After they are shaped into scaffold structures by different fabrication techniques, surface properties such as physical topography, stiffness, and surface chemistry play a major role in the endothelialization process after transplantation. Furthermore, biological cues such as growth factors (GFs) and endothelial cells (ECs) can be incorporated into the fabricated structures. As variously reported, fabrication techniques, especially 3D printing by extrusion and 3D printing by photopolymerization, allow the construction of vessels at a high resolution with diameters in the desired range. Strategies to fabricate of stable tubular structures with defined channels will also be discussed. This paper provides an overview of the many advances in blood vessel engineering and combinations of different fabrication techniques up to the present time.
Collapse
Affiliation(s)
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Kentaro Uchida
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation, Kadoma, Osaka, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- The Consortium for Future Innovation by Cultured Meat, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
6
|
Wang C, He J, Chen C, Luo W, Dang X, Mao L. A potential role of human esophageal cancer-related gene-4 in cardiovascular homeostasis. Gene 2024; 894:147977. [PMID: 37956966 DOI: 10.1016/j.gene.2023.147977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/10/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Human esophageal cancer related gene-4 (ECRG-4) encodes a 148-aminoacid pre-pro-peptide that can be processed tissue-dependently into multiple small peptides possessing multiple functions distinct from, similar to, or opposite to the tumor suppressor function of the full-length Ecrg4. Ecrg-4 is covalently bound to the cell surface through its signal peptide, colocalized with the innate immunity complex (TLR4-CD14-MD2), and functions as a 'sentinel' molecule in the maintenance of epithelium and leukocyte homeostasis, meaning that the presence of Ecrg-4 on the cell surface signals the maintained homeostasis, whereas the loss of Ecrg-4 due to tissue injury activates pro-inflammatory and tissue proliferative responses, and the level of Ecrg-4 gradually returns to its pre-injury level upon wound healing. Interestingly, Ecrg-4 is also highly expressed in the heart and its conduction system, endothelial cells, and vascular smooth muscle cells. Accumulating evidence has shown that Ecrg-4 is involved in cardiac rate/rhythm control, the development of atrial fibrillation, doxorubicin-induced cardiotoxicity, the ischemic response of the heart and hypoxic response in the carotid body, the pathogenesis of atherosclerosis, and likely the endemic incidence of idiopathic dilated cardiomyopathy. These preliminary discoveries suggest that Ecrg-4 may function as a 'sentinel' molecule in cardiovascular system as well. Here, we briefly review the basic characteristics of ECRG-4 as a tumor suppressor gene and its regulatory functions on inflammation and apoptosis; summarize the discoveries about its distribution in cardiovascular system and involvement in the development of CVDs, and discuss its potential as a novel therapeutic target for the maintenance of cardiovascular system homeostasis.
Collapse
Affiliation(s)
- Chaoying Wang
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Jianghui He
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Chunyue Chen
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Wenjun Luo
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Xitong Dang
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China.
| | - Liang Mao
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
7
|
AlZaim I, de Rooij LPMH, Sheikh BN, Börgeson E, Kalucka J. The evolving functions of the vasculature in regulating adipose tissue biology in health and obesity. Nat Rev Endocrinol 2023; 19:691-707. [PMID: 37749386 DOI: 10.1038/s41574-023-00893-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 09/27/2023]
Abstract
Adipose tissue is an endocrine organ and a crucial regulator of energy storage and systemic metabolic homeostasis. Additionally, adipose tissue is a pivotal regulator of cardiovascular health and disease, mediated in part by the endocrine and paracrine secretion of several bioactive products, such as adipokines. Adipose vasculature has an instrumental role in the modulation of adipose tissue expansion, homeostasis and metabolism. The role of the adipose vasculature has been extensively explored in the context of obesity, which is recognized as a global health problem. Obesity-induced accumulation of fat, in combination with vascular rarefaction, promotes adipocyte dysfunction and induces oxidative stress, hypoxia and inflammation. It is now recognized that obesity-associated endothelial dysfunction often precedes the development of cardiovascular diseases. Investigations have revealed heterogeneity within the vascular niche and dynamic reciprocity between vascular and adipose cells, which can become dysregulated in obesity. Here we provide a comprehensive overview of the evolving functions of the vasculature in regulating adipose tissue biology in health and obesity.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Laura P M H de Rooij
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bilal N Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Emma Börgeson
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
8
|
Li M, Popovic Z, Chu C, Reichetzeder C, Pommer W, Krämer BK, Hocher B. Impact of Angiopoietin-2 on Kidney Diseases. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:0. [PMID: 38306230 PMCID: PMC10826602 DOI: 10.1159/000529774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/14/2023] [Indexed: 02/04/2024]
Abstract
Background Angiopoietins (Ang) are essential angiogenic factors involved in angiogenesis, vascular maturation, and inflammation. The most studied angiopoietins, angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2), behave antagonistically to each other in vivo to sustain vascular endothelium homeostasis. While Ang-1 typically acts as the endothelium-protective mediator, its context-dependent antagonist Ang-2 can promote endothelium permeability and vascular destabilization, hence contributing to a poor outcome in vascular diseases via endothelial injury, vascular dysfunction, and microinflammation. The pathogenesis of kidney diseases is associated with endothelial dysfunction and chronic inflammation in renal diseases. Summary Several preclinical studies report overexpression of Ang-2 in renal tissues of certain kidney disease models; additionally, clinical studies show increased levels of circulating Ang-2 in the course of chronic kidney disease, implying that Ang-2 may serve as a useful biomarker in these patients. However, the exact mechanisms of Ang-2 action in renal diseases remain unclear. Key Messages We summarized the recent findings on Ang-2 in kidney diseases, including preclinical studies and clinical studies, aiming to provide a systematic understanding of the role of Ang-2 in these diseases.
Collapse
Affiliation(s)
- Mei Li
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Zoran Popovic
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Nephrology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | | | - Wolfgang Pommer
- Charité University Hospital Department of Nephrology and Internal Intensive Care Medicine, Berlin, Germany
| | - Bernhard K. Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- Center for Innate Immunoscience, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Institute of Medical Diagnostics, IMD Berlin, Berlin, Germany
| |
Collapse
|
9
|
Oliveira F, Bondareva O, Rodríguez-Aguilera JR, Sheikh BN. Cultured brain pericytes adopt an immature phenotype and require endothelial cells for expression of canonical markers and ECM genes. Front Cell Neurosci 2023; 17:1165887. [PMID: 37201162 PMCID: PMC10185779 DOI: 10.3389/fncel.2023.1165887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/05/2023] [Indexed: 05/20/2023] Open
Abstract
Pericytes (PCs) are essential components of the blood brain barrier. Brain PCs are critical for dynamically regulating blood flow, for maintaining vascular integrity and their dysregulation is associated with a myriad of disorders such as Alzheimer's disease. To understand their physiological and molecular functions, studies have increasingly focused on primary brain PC isolation and culture. Multiple methods for PC culture have been developed over the years, however, it is still unclear how primary PCs compare to their in vivo counterparts. To address this question, we compared cultured brain PCs at passage 5 and 20 to adult and embryonic brain PCs directly isolated from mouse brains via single cell RNA-seq. Cultured PCs were highly homogeneous, and were most similar to embryonic PCs, while displaying a significantly different transcriptional profile to adult brain PCs. Cultured PCs downregulated canonical PC markers and extracellular matrix (ECM) genes. Importantly, expression of PC markers and ECM genes could be improved by co-culture with brain endothelial cells, showing the importance of the endothelium in maintaining PC identity and function. Taken together, these results highlight key transcriptional differences between cultured and in vivo PCs which should be considered when performing in vitro experiments with brain PCs.
Collapse
Affiliation(s)
- Fabiana Oliveira
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, Leipzig University, Leipzig, Germany
| | - Olga Bondareva
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, Leipzig University, Leipzig, Germany
| | - Jesús Rafael Rodríguez-Aguilera
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, Leipzig University, Leipzig, Germany
| | - Bilal N. Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, Leipzig University, Leipzig, Germany
- *Correspondence: Bilal N. Sheikh,
| |
Collapse
|
10
|
Bondareva O, Rodríguez-Aguilera JR, Oliveira F, Liao L, Rose A, Gupta A, Singh K, Geier F, Schuster J, Boeckel JN, Buescher JM, Kohli S, Klöting N, Isermann B, Blüher M, Sheikh BN. Single-cell profiling of vascular endothelial cells reveals progressive organ-specific vulnerabilities during obesity. Nat Metab 2022; 4:1591-1610. [PMID: 36400935 PMCID: PMC9684070 DOI: 10.1038/s42255-022-00674-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 09/30/2022] [Indexed: 11/20/2022]
Abstract
Obesity promotes diverse pathologies, including atherosclerosis and dementia, which frequently involve vascular defects and endothelial cell (EC) dysfunction. Each organ has distinct EC subtypes, but whether ECs are differentially affected by obesity is unknown. Here we use single-cell RNA sequencing to analyze transcriptomes of ~375,000 ECs from seven organs in male mice at progressive stages of obesity to identify organ-specific vulnerabilities. We find that obesity deregulates gene expression networks, including lipid handling, metabolic pathways and AP1 transcription factor and inflammatory signaling, in an organ- and EC-subtype-specific manner. The transcriptomic aberrations worsen with sustained obesity and are only partially mitigated by dietary intervention and weight loss. For example, dietary intervention substantially attenuates dysregulation of liver, but not kidney, EC transcriptomes. Through integration with human genome-wide association study data, we further identify a subset of vascular disease risk genes that are induced by obesity. Our work catalogs the impact of obesity on the endothelium, constitutes a useful resource and reveals leads for investigation as potential therapeutic targets.
Collapse
Affiliation(s)
- Olga Bondareva
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Jesús Rafael Rodríguez-Aguilera
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Fabiana Oliveira
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Longsheng Liao
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Alina Rose
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Anubhuti Gupta
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Kunal Singh
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Florian Geier
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
| | - Jenny Schuster
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
| | - Jes-Niels Boeckel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, University of Leipzig, Leipzig, Germany
| | - Joerg M Buescher
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Shrey Kohli
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig, Germany
| | - Bilal N Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany.
- Medical Faculty, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
11
|
McDaniels JM, Shetty AC, Rousselle TV, Bardhi E, Maluf DG, Mas VR. The cellular landscape of the normal kidney allograft: Main players balancing the alloimmune response. FRONTIERS IN TRANSPLANTATION 2022; 1:988238. [PMID: 38994377 PMCID: PMC11235379 DOI: 10.3389/frtra.2022.988238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/22/2022] [Indexed: 07/13/2024]
Abstract
Despite recent advances made in short-term outcomes; minimal improvements have been observed in long-term kidney transplantation outcomes. Due to an imbalance between organ transplant availability and patient waiting list, expanding kidney allograft longevity is a critical need in the field. Prior studies have either focused on early ischemic and immunological conditions affecting kidney allografts (e.g., delayed graft function, acute rejection) or late stage chronic injury when interventions are no longer feasible. However, studies characterizing kidney allografts with normal function by its cellular distribution, cell-cell interactions, and associated molecular pathways are lacking. Herein, we used single nuclei RNA-sequencing to uncover the cellular landscape and transcriptome of the normal kidney allograft. We profiled 40,950 nuclei from seven human kidney biopsies (normal native, N = 3; normal allograft, N = 4); normal allograft protocol biopsies were collected ≥15-months post-transplant. A total of 17 distinct cell clusters were identified with proximal tubules (25.70 and 21.01%), distal tubules (15.22 and 18.20%), and endothelial cells (EC) (4.26 and 9.94%) constituting the major cell populations of normal native and normal allograft kidneys, respectively. A large proportion of cycling cells from normal native kidneys were in G1-phase (43.96%) whereas cells from normal allograft were predominantly in S-phase (32.69%). This result suggests that transcriptional differences between normal native and normal allograft biopsies are dependent on the new host environment, immunosuppression, and injury-affliction. In the normal allograft, EC-specific genes upregulated metabolism, the immune response, and cellular growth, emphasizing their role in maintaining homeostasis during the ongoing alloreactive stress response. Immune cells, including B (2.81%), macrophages (24.96%), monocytes (15.29%), natural killer (NK) (12.83%), neutrophils (8.44%), and T cells (14.41%, were increased in normal allografts despite lack of histological or clinical evidence of acute rejection. Phenotypic characterization of immune cell markers supported lymphocyte activation and proinflammatory cytokines signaling pathways (i.e., IL-15, IL-32). The activation of B, NK, and T cells reveals potential immune cells underlying subclinical inflammation and repair. These single nuclei analyses provide novel insights into kidney and immune cell associated signaling pathways that portray kidney grafts with normal allograft function beyond 2-years post-transplant, revealing a novel perspective in understanding long-term allograft graft survival.
Collapse
Affiliation(s)
- Jennifer M McDaniels
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| | - Amol C Shetty
- Institute for Genome Sciences, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Thomas V Rousselle
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| | - Elissa Bardhi
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| | - Daniel G Maluf
- Program in Transplantation, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Valeria R Mas
- Surgical Sciences Division, Department of Surgery, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
12
|
Li Q, Ouyang X, Lin J. The impact of periodontitis on vascular endothelial dysfunction. Front Cell Infect Microbiol 2022; 12:998313. [PMID: 36118034 PMCID: PMC9480849 DOI: 10.3389/fcimb.2022.998313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/15/2022] [Indexed: 11/26/2022] Open
Abstract
Periodontitis, an oral inflammatory disease, originates from periodontal microbiota dysbiosis which is associated with the dysregulation of host immunoinflammatory response. This chronic infection is not only harmful to oral health but is also a risk factor for the onset and progress of various vascular diseases, such as hypertension, atherosclerosis, and coronary arterial disease. Vascular endothelial dysfunction is the initial key pathological feature of vascular diseases. Clarifying the association between periodontitis and vascular endothelial dysfunction is undoubtedly a key breakthrough for understanding the potential relationship between periodontitis and vascular diseases. However, there is currently a lack of an updated review of their relationship. Therefore, we aim to focus on the implications of periodontitis in vascular endothelial dysfunction in this review.
Collapse
Affiliation(s)
- Qian Li
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiangying Ouyang
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
- *Correspondence: Xiangying Ouyang, ; Jiang Lin,
| | - Jiang Lin
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- *Correspondence: Xiangying Ouyang, ; Jiang Lin,
| |
Collapse
|
13
|
Wenninger N, Bernhart C, Kappaun W, Kollau A, Kalcher K, Ortner A. High-performance amperometric determination of nitric oxide released by endothelial cells using flow injection analysis. Talanta 2022. [DOI: 10.1016/j.talanta.2022.123810] [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]
|
14
|
Liu Z, Andraska E, Akinbode D, Mars W, Alvidrez RIM. LRP1 in the Vascular Wall. CURRENT PATHOBIOLOGY REPORTS 2022. [DOI: 10.1007/s40139-022-00231-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
15
|
Namiguchi K, Sakaue T, Okazaki M, Kanno K, Komoda Y, Shikata F, Kurata M, Ota N, Kubota Y, Kurobe H, Nishimura T, Masumoto J, Higashiyama S, Izutani H. Unique Angiogenesis From Cardiac Arterioles During Pericardial Adhesion Formation. Front Cardiovasc Med 2022; 8:761591. [PMID: 35187100 PMCID: PMC8852280 DOI: 10.3389/fcvm.2021.761591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/22/2021] [Indexed: 11/22/2022] Open
Abstract
Objectives The molecular mechanisms underlying post-operative pericardial adhesions remain poorly understood. We aimed to unveil the temporal molecular and cellular mechanisms underlying tissue dynamics during adhesion formation, including inflammation, angiogenesis, and fibrosis. Methods and Results We visualized cell-based tissue dynamics during pericardial adhesion using histological evaluations. To determine the molecular mechanism, RNA-seq was performed. Chemical inhibitors were administered to confirm the molecular mechanism underlying adhesion formation. A high degree of adhesion formation was observed during the stages in which collagen production was promoted. Histological analyses showed that arterioles excessively sprouted from pericardial tissues after the accumulation of neutrophils on the heart surface in mice as well as humans. The combination of RNA-seq and histological analyses revealed that hyperproliferative endothelial and smooth muscle cells with dedifferentiation appeared in cytokine-exposed sprouting vessels and adhesion tissue but not in quiescent vessels in the heart. SMAD2/3 and ERK activation was observed in sprouting vessels. The simultaneous abrogation of PI3K/ERK or TGF-β/MMP9 signaling significantly decreased angiogenic sprouting, followed by inhibition of adhesion formation. Depleting MMP9-positive neutrophils shortened mice survival and decreased angiogenic sprouting and fibrosis in the adhesion. Our data suggest that TGF-β/matrix metalloproteinase-dependent tissue remodeling and PI3K/ERK signaling activation might contribute to unique angiogenesis with dedifferentiation of vascular smooth muscle cells from the contractile to the synthetic phenotype for fibrosis in the pericardial cavity. Conclusions Our findings provide new insights in developing prevention strategies for pericardial adhesions by targeting the recruitment of vascular cells from heart tissues.
Collapse
Affiliation(s)
- Kenji Namiguchi
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Tomohisa Sakaue
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Japan
- *Correspondence: Tomohisa Sakaue
| | - Mikio Okazaki
- Department of General Thoracic Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kaho Kanno
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yuhei Komoda
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Fumiaki Shikata
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Mie Kurata
- Department of Pathology, Division of Analytical Pathology, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Pathology, Proteo-Science Center, Toon, Japan
| | - Noritaka Ota
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Hirotsugu Kurobe
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takashi Nishimura
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Junya Masumoto
- Department of Pathology, Division of Analytical Pathology, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Pathology, Proteo-Science Center, Toon, Japan
| | - Shigeki Higashiyama
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Japan
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Molecular and Cellular Biology, Research Center, Osaka International Cancer Institute, Osaka, Japan
| | - Hironori Izutani
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Toon, Japan
- Hironori Izutani
| |
Collapse
|
16
|
Hemodynamics Challenges for the Navigation of Medical Microbots for the Treatment of CVDs. MATERIALS 2021; 14:ma14237402. [PMID: 34885556 PMCID: PMC8658690 DOI: 10.3390/ma14237402] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 01/03/2023]
Abstract
Microbots have been considered powerful tools in minimally invasive medicine. In the last few years, the topic has been highly studied by researchers across the globe to further develop the capabilities of microbots in medicine. One of many applications of these devices is performing surgical procedures inside the human circulatory system. It is expected that these microdevices traveling along the microvascular system can remove clots, deliver drugs, or even look for specific cells or regions to diagnose and treat. Although many studies have been published about this subject, the experimental influence of microbot morphology in hemodynamics of specific sites of the human circulatory system is yet to be explored. There are numerical studies already considering some of human physiological conditions, however, experimental validation is vital and demands further investigations. The roles of specific hemodynamic variables, the non-Newtonian behavior of blood and its particulate nature at small scales, the flow disturbances caused by the heart cycle, and the anatomy of certain arteries (i.e., bifurcations and tortuosity of vessels of some regions) in the determination of the dynamic performance of microbots are of paramount importance. This paper presents a critical analysis of the state-of-the-art literature related to pulsatile blood flow around microbots.
Collapse
|
17
|
Ge W, Yan ZH, Wang L, Tan SJ, Liu J, Reiter RJ, Luo SM, Sun QY, Shen W. A hypothetical role for autophagy during the day/night rhythm-regulated melatonin synthesis in the rat pineal gland. J Pineal Res 2021; 71:e12742. [PMID: 33960014 DOI: 10.1111/jpi.12742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/12/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022]
Abstract
Melatonin is a highly conserved molecule that regulates day/night rhythms; it is associated with sleep improvement, reactive oxygen species (ROS) scavenging, anti-aging effects, and seasonal and circadian rhythms and has been a hot topic of research for decades. Using single-cell RNA sequencing, a recent study describes a single-cell transcriptome atlas for the rat pineal gland. Based on a more comprehensive analysis of the retrieved data (Mays et al., PLoS One, 2018, 13, e0205883), results from the current study unveiled the underappreciated gene regulatory network behind different cell populations in the pineal gland. More importantly, our study here characterized, for the first time, the day/night activation of autophagy flux in the rat pineal gland, indicating a potential role of autophagy in regulating melatonin synthesis in the rat pineal gland. These findings emphasized a hypothetical role of day/night autophagy in linking the biological clock with melatonin synthesis. Furthermore, ultrastructure analysis of pinealocytes provided fascinating insights into differences in their intracellular structure between daytime and nighttime. In addition, we also provide a preliminary description of cell-cell communication in the rat pineal gland. In summary, the current study unveils the day/night regulation of autophagy in the rat pineal gland, raising a potential role of autophagy in day/night-regulated melatonin synthesis.
Collapse
Affiliation(s)
- Wei Ge
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Zi-Hui Yan
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Lu Wang
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Shao-Jing Tan
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Jing Liu
- Central Laboratory of Qingdao Agricultural University, Qingdao Agricultural University, Qingdao, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health, San Antonio, TX, USA
| | - Shi-Ming Luo
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Wei Shen
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
18
|
He Z, Wang G, Wu J, Tang Z, Luo M. The molecular mechanism of LRP1 in physiological vascular homeostasis and signal transduction pathways. Biomed Pharmacother 2021; 139:111667. [PMID: 34243608 DOI: 10.1016/j.biopha.2021.111667] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023] Open
Abstract
Interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), pericytes (PCs) and macrophages (MФ), the major components of blood vessels, play a crucial role in maintaining vascular structural and functional homeostasis. Low-density lipoprotein (LDL) receptor-related protein-1 (LRP1), a transmembrane receptor protein belonging to the LDL receptor family, plays multifunctional roles in maintaining endocytosis, homeostasis, and signal transduction. Accumulating evidence suggests that LRP1 modulates vascular homeostasis mainly by regulating vasoactive substances and specific intracellular signaling pathways, including the plasminogen activator inhibitor 1 (PAI-1) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, transforming growth factor-β (TGF-β) signaling pathway and vascular endothelial growth factor (VEGF) signaling pathway. The aim of the present review is to focus on recent advances in the discovery and mechanism of vascular homeostasis regulated by LRP1-dependent signaling pathways. These recent discoveries expand our understanding of the mechanisms controlling LRP1 as a target for studies on vascular complications.
Collapse
Affiliation(s)
- Zhaohui He
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Zonghao Tang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
19
|
Delicate Role of PD-L1/PD-1 Axis in Blood Vessel Inflammatory Diseases: Current Insight and Future Significance. Int J Mol Sci 2020; 21:ijms21218159. [PMID: 33142805 PMCID: PMC7663405 DOI: 10.3390/ijms21218159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint molecules are the antigen-independent generator of secondary signals that aid in maintaining the homeostasis of the immune system. The programmed death ligand-1 (PD-L1)/PD-1 axis is one among the most extensively studied immune-inhibitory checkpoint molecules, which delivers a negative signal for T cell activation by binding to the PD-1 receptor. The general attributes of PD-L1's immune-suppressive qualities and novel mechanisms on the barrier functions of vascular endothelium to regulate blood vessel-related inflammatory diseases are concisely reviewed. Though targeting the PD-1/PD-L1 axis has received immense recognition-the Nobel Prize in clinical oncology was awarded in the year 2018 for this discovery-the use of therapeutic modulating strategies for the PD-L1/PD-1 pathway in chronic inflammatory blood vessel diseases is still limited to experimental models. However, studies using clinical specimens that support the role of PD-1 and PD-L1 in patients with underlying atherosclerosis are also detailed. Of note, delicate balances in the expression levels of PD-L1 that are needed to preserve T cell immunity and to curtail acute as well as chronic infections in underlying blood vessel diseases are discussed. A significant link exists between altered lipid and glucose metabolism in different cells and the expression of PD-1/PD-L1 molecules, and its possible implications on vascular inflammation are justified. This review summarizes the most recent insights concerning the role of the PD-L1/PD-1 axis in vascular inflammation and, in addition, provides an overview exploring the novel therapeutic approaches and challenges of manipulating these immune checkpoint proteins, PD-1 and PD-L1, for suppressing blood vessel inflammation.
Collapse
|