1
|
Xia T, Pan Z, Wan H, Li Y, Mao G, Zhao J, Zhang F, Pan S. Mechanisms of mechanical stimulation in the development of respiratory system diseases. Am J Physiol Lung Cell Mol Physiol 2024; 327:L724-L739. [PMID: 39316681 DOI: 10.1152/ajplung.00122.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
During respiration, mechanical stress can initiate biological responses that impact the respiratory system. Mechanical stress plays a crucial role in the development of the respiratory system. However, pathological mechanical stress can impact the onset and progression of respiratory diseases by influencing the extracellular matrix and cell transduction processes. In this article, we explore the mechanisms by which mechanical forces communicate with and influence cells. We outline the basic knowledge of respiratory mechanics, elucidating the important role of mechanical stimulation in influencing respiratory system development and differentiation from a microscopic perspective. We also explore the potential mechanisms of mechanical transduction in the pathogenesis and development of respiratory diseases such as asthma, lung injury, pulmonary fibrosis, and lung cancer. Finally, we look forward to new research directions in cellular mechanotransduction, aiming to provide fresh insights for future therapeutic research on respiratory diseases.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoxin Wan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongsen Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guocai Mao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fangbiao Zhang
- Department of Cardiothoracic Surgery, Lishui Municipal Central Hospital, Lishui, People's Republic of China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
2
|
Han H, Jia H, Wang YF, Song JP. Cardiovascular adaptations and pathological changes induced by spaceflight: from cellular mechanisms to organ-level impacts. Mil Med Res 2024; 11:68. [PMID: 39334239 PMCID: PMC11429428 DOI: 10.1186/s40779-024-00570-3] [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: 03/04/2024] [Accepted: 09/01/2024] [Indexed: 09/30/2024] Open
Abstract
The advancement in extraterrestrial exploration has highlighted the crucial need for studying how the human cardiovascular system adapts to space conditions. Human development occurs under the influence of gravity, shielded from space radiation by Earth's magnetic field, and within an environment characterized by 24-hour day-night cycles resulting from Earth's rotation, thus deviating from these conditions necessitates adaptive responses for survival. With upcoming manned lunar and Martian missions approaching rapidly, it is essential to understand the impact of various stressors induced by outer-space environments on cardiovascular health. This comprehensive review integrates insights from both actual space missions and simulated experiments on Earth, to analyze how microgravity, space radiation, and disrupted circadian affect cardiovascular well-being. Prolonged exposure to microgravity induces myocardial atrophy and endothelial dysfunction, which may be exacerbated by space radiation. Mitochondrial dysfunction and oxidative stress emerge as key underlying mechanisms along with disturbances in ion channel perturbations, cytoskeletal damage, and myofibril changes. Disruptions in circadian rhythms caused by factors such as microgravity, light exposure, and irregular work schedules, could further exacerbate cardiovascular issues. However, current research tends to predominantly focus on disruptions in the core clock gene, overlooking the multifactorial nature of circadian rhythm disturbances in space. Future space missions should prioritize targeted prevention strategies and early detection methods for identifying cardiovascular risks, to preserve astronaut health and ensure mission success.
Collapse
Affiliation(s)
- Han Han
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yi-Fan Wang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jiang-Ping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
3
|
Yang J, Cui Y, Zhao J, Tang S, Wang A, Wang J, Chen Y, Luo J, Wang G, Yan J, Du J, Wang J. Simulated microgravity-induced dysregulation of cerebrospinal fluid immune homeostasis by disrupting the blood-cerebrospinal fluid barrier. Brain Behav 2024; 14:e3648. [PMID: 39262161 PMCID: PMC11391017 DOI: 10.1002/brb3.3648] [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: 09/26/2023] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND The blood-cerebrospinal fluid barrier (BCSFB) comprises the choroid plexus epithelia. It is important for brain development, maintenance, function, and especially for maintaining immune homeostasis in the cerebrospinal fluid (CSF). Although previous studies have shown that the peripheral immune function of the body is impaired upon exposure to microgravity, no studies have reported changes in immune cells and cytokines in the CSF that reflect neuroimmune status. The purpose of this study is to investigate the alterations in cerebrospinal fluid (CSF) immune homeostasis induced by microgravity and its mechanisms. This research is expected to provide basic data for brain protection of astronauts during spaceflight. METHODS The proportions of immune cells in the CSF and peripheral blood (PB) of SMG rats were analyzed using flow cytometry. Immune function was evaluated by measuring cytokine concentrations using the Luminex method. The histomorphology and ultrastructure of the choroid plexus epithelia were determined. The concentrations of intercellular junction proteins in choroid plexus epithelial cells, including vascular endothelial-cadherin (VE-cadherin), zonula occludens 1 (ZO-1), Claudin-1 and occludin, were detected using western blotting and immunofluorescence staining to characterize BCSFB injury. RESULTS We found that SMG caused significant changes in the proportion of CD4 and CD8 T cells in the CSF and a significant increase in the levels of cytokines (GRO/KC, IL-18, MCP-1, and RANTES). In the PB, there was a significant decrease in the proportion of T cells and NKT cells and a significant increase in cytokine levels (GRO/KC, IL-18, MCP-1, and TNF-α). Additionally, we observed that the trends in immune markers in the PB and CSF were synchronized within specific SMG durations, suggesting that longer SMG periods (≥21 days) have a more pronounced impact on immune markers. Furthermore, 21d-SMG resulted in ultrastructural disruption and downregulated expression of intercellular junction proteins in rat choroid plexus epithelial cells. CONCLUSIONS We found that SMG disrupts the BCSFB and affects the CSF immune homeostasis. This study provides new insights into the health protection of astronauts during spaceflight.
Collapse
Affiliation(s)
- Jing Yang
- Beijing Tong Ren HospitalCapital Medical UniversityBeijingChina
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
- Department of NeurologyAerospace Center HospitalBeijingChina
| | - Yaoyuan Cui
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
| | - Juan Zhao
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
| | - Shiyi Tang
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
| | - Anqing Wang
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
| | - Junxiao Wang
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
| | - Yu Chen
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
| | - Jilong Luo
- Institute of Medical TechnologyPeking University Health Science CenterBeijingChina
| | - Guan Wang
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
| | - Junhao Yan
- Department of Anatomy, Histology and Embryology, School of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Jichen Du
- Aerospace Medical CenterAerospace Center HospitalBeijingChina
- Institute of Medical TechnologyPeking University Health Science CenterBeijingChina
| | - Jiawei Wang
- Beijing Tong Ren HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
4
|
Gros A, Furlan FM, Rouglan V, Favereaux A, Bontempi B, Morel JL. Physical exercise restores adult neurogenesis deficits induced by simulated microgravity. NPJ Microgravity 2024; 10:69. [PMID: 38906877 PMCID: PMC11192769 DOI: 10.1038/s41526-024-00411-6] [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: 11/03/2023] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
Cognitive impairments have been reported in astronauts during spaceflights and documented in ground-based models of simulated microgravity (SMG) in animals. However, the neuronal causes of these behavioral effects remain largely unknown. We explored whether adult neurogenesis, known to be a crucial plasticity mechanism supporting memory processes, is altered by SMG. Adult male Long-Evans rats were submitted to the hindlimb unloading model of SMG. We studied the proliferation, survival and maturation of newborn cells in the following neurogenic niches: the subventricular zone (SVZ)/olfactory bulb (OB) and the dentate gyrus (DG) of the hippocampus, at different delays following various periods of SMG. SMG exposure for 7 days, but not shorter periods of 6 or 24 h, resulted in a decrease of newborn cell proliferation restricted to the DG. SMG also induced a decrease in short-term (7 days), but not long-term (21 days), survival of newborn cells in the SVZ/OB and DG. Physical exercise, used as a countermeasure, was able to reverse the decrease in newborn cell survival observed in the SVZ and DG. In addition, depending on the duration of SMG periods, transcriptomic analysis revealed modifications in gene expression involved in neurogenesis. These findings highlight the sensitivity of adult neurogenesis to gravitational environmental factors during a transient period, suggesting that there is a period of adaptation of physiological systems to this new environment.
Collapse
Affiliation(s)
- Alexandra Gros
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
- Centre National d'Etudes Spatiales, F-75001, Paris, France
| | - Fandilla Marie Furlan
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
- Department of Genetics & Evolution, 30 Quai Ernest-Ansermet, 1205, Geneva, Switzerland
| | - Vanessa Rouglan
- CNRS, IINS, UMR 5297, University Bordeaux, F-33000, Bordeaux, France
| | | | - Bruno Bontempi
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
| | - Jean-Luc Morel
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France.
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France.
| |
Collapse
|
5
|
Jogdand A, Landolina M, Chen Y. Organs in orbit: how tissue chip technology benefits from microgravity, a perspective. FRONTIERS IN LAB ON A CHIP TECHNOLOGIES 2024; 3:1356688. [PMID: 38915901 PMCID: PMC11195915 DOI: 10.3389/frlct.2024.1356688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Tissue chips have become one of the most potent research tools in the biomedical field. In contrast to conventional research methods, such as 2D cell culture and animal models, tissue chips more directly represent human physiological systems. This allows researchers to study therapeutic outcomes to a high degree of similarity to actual human subjects. Additionally, as rocket technology has advanced and become more accessible, researchers are using the unique properties offered by microgravity to meet specific challenges of modeling tissues on Earth; these include large organoids with sophisticated structures and models to better study aging and disease. This perspective explores the manufacturing and research applications of microgravity tissue chip technology, specifically investigating the musculoskeletal, cardiovascular, and nervous systems.
Collapse
Affiliation(s)
- Aditi Jogdand
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| | - Maxwell Landolina
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
6
|
Li Z, Wu J, Zhao T, Wei Y, Xu Y, Liu Z, Li X, Chen X. Microglial activation in spaceflight and microgravity: potential risk of cognitive dysfunction and poor neural health. Front Cell Neurosci 2024; 18:1296205. [PMID: 38425432 PMCID: PMC10902453 DOI: 10.3389/fncel.2024.1296205] [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: 09/18/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Due to the increased crewed spaceflights in recent years, it is vital to understand how the space environment affects human health. A lack of gravitational force is known to risk multiple physiological functions of astronauts, particularly damage to the central nervous system (CNS). As innate immune cells of the CNS, microglia can transition from a quiescent state to a pathological state, releasing pro-inflammatory cytokines that contribute to neuroinflammation. There are reports indicating that microglia can be activated by simulating microgravity or exposure to galactic cosmic rays (GCR). Consequently, microglia may play a role in the development of neuroinflammation during spaceflight. Prolonged spaceflight sessions raise concerns about the chronic activation of microglia, which could give rise to various neurological disorders, posing concealed risks to the neural health of astronauts. This review summarizes the risks associated with neural health owing to microglial activation and explores the stressors that trigger microglial activation in the space environment. These stressors include GCR, microgravity, and exposure to isolation and stress. Of particular focus is the activation of microglia under microgravity conditions, along with the proposal of a potential mechanism.
Collapse
Affiliation(s)
- Zihan Li
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Jiarui Wu
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Tianyuan Zhao
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Yiyun Wei
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Yajing Xu
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Xiaoqiong Li
- School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Xuechai Chen
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
7
|
Kazmi S, Farajdokht F, Meynaghizadeh-Zargar R, Sadigh-Eteghad S, Pasokh A, Farzipour M, Farazi N, Hamblin MR, Mahmoudi J. Transcranial photobiomodulation mitigates learning and memory impairments induced by hindlimb unloading in a mouse model of microgravity exposure by suppression of oxidative stress and neuroinflammation signaling pathways. Brain Res 2023; 1821:148583. [PMID: 37717889 DOI: 10.1016/j.brainres.2023.148583] [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: 04/05/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
Prolonged microgravity exposure causes cognitive impairment. Evidence shows that oxidative stress and neuroinflammation are involved in the causation. Here, we explore the effectiveness of transcranial near-infrared photobiomodulation (PBM) on cognitive deficits in a mouse model of simulated microgravity. 24 adult male C57BL/6 mice were assigned into three groups (8 in each); control, hindlimb unloading (HU), and HU + PBM groups. After surgery to fit the suspension fixing, the animals were housed either in HU cages or in their normal cage for 14 days. The mice in the HU + PBM group received PBM (810 nm laser, 10 Hz, 8 J/cm2) once per day for 14 days. Spatial learning and memory were assessed in the Lashley III maze and hippocampus tissue samples were collected to assess oxidative stress markers and protein expression of brain-derived neurotrophic factor (BDNF), nuclear factor erythroid 2-related factor 2 (Nrf2), Sirtuin 1 (Sirt1), and Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Behavioral testing showed that the PBM-treated animals had a shorter latency time to find the target and fewer errors than the HU group. PBM decreased hippocampal lipid peroxidation while increasing antioxidant defense systems (glutathione peroxidase, superoxide dismutase, and total antioxidant capacity) compared to HU mice. PBM increased protein expression of Sirt1, Nrf2, and BDNF while decreasing NF-κB compared to HU mice. Our findings suggested that the protective effect of PBM against HU-induced cognitive impairment involved the activation of the Sirt1/Nrf2 signaling pathway, up-regulation of BDNF, and reduction of neuroinflammation and oxidative stress in the hippocampus.
Collapse
Affiliation(s)
- Sareh Kazmi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Pasokh
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Mohammad Farzipour
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narmin Farazi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Troncoso F, Sandoval H, Ibañez B, López-Espíndola D, Bustos F, Tapia JC, Sandaña P, Escudero-Guevara E, Nualart F, Ramírez E, Powers R, Vatish M, Mistry HD, Kurlak LO, Acurio J, Escudero C. Reduced Brain Cortex Angiogenesis in the Offspring of the Preeclampsia-Like Syndrome. Hypertension 2023; 80:2559-2571. [PMID: 37767691 DOI: 10.1161/hypertensionaha.123.21756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Children from pregnancies affected by preeclampsia have an increased risk of cognitive and behavioral alterations via unknown pathophysiology. We tested the hypothesis that preeclampsia generated reduced brain cortex angiogenesis in the offspring. METHODS The preeclampsia-like syndrome (PELS) mouse model was generated by administering the nitric oxide inhibitor NG-nitroarginine methyl ester hydrochloride. Confirmatory experiments were done using 2 additional PELS models. While in vitro analysis used mice and human brain endothelial cells exposed to serum of postnatal day 5 pups or umbilical plasma from preeclamptic pregnancies, respectively. RESULTS We report significant reduction in the area occupied by blood vessels in the motor and somatosensory brain cortex of offspring (postnatal day 5) from PELS compared with uncomplicated control offspring. These data were confirmed using 2 additional PELS models. Furthermore, circulating levels of critical proangiogenic factors, VEGF (vascular endothelial growth factor), and PlGF (placental growth factor) were lower in postnatal day 5 PELS. Also we found lower VEGF receptor 2 (KDR [kinase insert domain-containing receptor]) levels in mice and human endothelial cells exposed to the serum of postnatal day 5 PELS or fetal plasma of preeclamptic pregnancies, respectively. These changes were associated with lower in vitro angiogenic capacity, diminished cell migration, larger F-actin filaments, lower number of filopodia, and lower protein levels of F-actin polymerization regulators in brain endothelial cells exposed to serum or fetal plasma of offspring from preeclampsia. CONCLUSIONS Offspring from preeclampsia exhibited diminished brain cortex angiogenesis, associated with lower circulating VEGF/PlGF/KDR protein levels, impaired brain endothelial migration, and dysfunctional assembly of F-actin filaments. These alterations may predispose to structural and functional alterations in long-term brain development.
Collapse
Affiliation(s)
- Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Hermes Sandoval
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Belén Ibañez
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E., F.B.)
- Group of Research and Innovation in Vascular Health, Chillan, Chile (D.L.-E., C.E.)
| | - Francisca Bustos
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E., F.B.)
| | - Juan Carlos Tapia
- Stem Cells and Neuroscience Center, School of Medicine, University of Talca, Chile (J.C.T.)
| | - Pedro Sandaña
- Anatomopatholy Unit, Hospital Clinico Herminda Martin, Chillan, Chile (P.S.)
| | - Esthefanny Escudero-Guevara
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepcion, Chile (F.N., E.R.)
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile (F.N.)
| | - Eder Ramírez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepcion, Chile (F.N., E.R.)
| | - Robert Powers
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, PA (R.P.)
| | - Manu Vatish
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, England (M.V.)
| | - Hiten D Mistry
- Division of Women and Children's Health, School of Life Course and Population Sciences, King's College London, United Kingdom (H.D.M.)
| | - Lesia O Kurlak
- Stroke Trials Unit, School of Medicine, University of Nottingham, United Kingdom (L.O.K.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
- Group of Research and Innovation in Vascular Health, Chillan, Chile (D.L.-E., C.E.)
| |
Collapse
|
9
|
Simulated Microgravity Alters P-Glycoprotein Efflux Function and Expression via the Wnt/β-Catenin Signaling Pathway in Rat Intestine and Brain. Int J Mol Sci 2023; 24:ijms24065438. [PMID: 36982513 PMCID: PMC10049079 DOI: 10.3390/ijms24065438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
The drug efflux transporter permeability glycoprotein (P-gp) plays an important role in oral drug absorption and distribution. Under microgravity (MG), the changes in P-gp efflux function may alter the efficacy of oral drugs or lead to unexpected effects. Oral drugs are currently used to protect and treat multisystem physiological damage caused by MG; whether P-gp efflux function changes under MG remains unclear. This study aimed to investigate the alteration of P-gp efflux function, expression, and potential signaling pathway in rats and cells under different simulated MG (SMG) duration. The altered P-gp efflux function was verified by the in vivo intestinal perfusion and the brain distribution of P-gp substrate drugs. Results showed that the efflux function of P-gp was inhibited in the 7 and 21 day SMG-treated rat intestine and brain and 72 h SMG-treated human colon adenocarcinoma cells and human cerebral microvascular endothelial cells. P-gp protein and gene expression levels were continually down-regulated in rat intestine and up-regulated in rat brain by SMG. P-gp expression was regulated by the Wnt/β-catenin signaling pathway under SMG, verified by a pathway-specific agonist and inhibitor. The elevated intestinal absorption and brain distribution of acetaminophen levels also confirmed the inhibited P-gp efflux function in rat intestine and brain under SMG. This study revealed that SMG alters the efflux function of P-gp and regulates the Wnt/β-catenin signaling pathway in the intestine and the brain. These findings may be helpful in guiding the use of P-gp substrate drugs during spaceflight.
Collapse
|
10
|
Dubayle D, Vanden-Bossche A, Peixoto T, Morel JL. Hypergravity Increases Blood-Brain Barrier Permeability to Fluorescent Dextran and Antisense Oligonucleotide in Mice. Cells 2023; 12:cells12050734. [PMID: 36899870 PMCID: PMC10000817 DOI: 10.3390/cells12050734] [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: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
The earliest effect of spaceflight is an alteration in vestibular function due to microgravity. Hypergravity exposure induced by centrifugation is also able to provoke motion sickness. The blood-brain barrier (BBB) is the crucial interface between the vascular system and the brain to ensure efficient neuronal activity. We developed experimental protocols of hypergravity on C57Bl/6JRJ mice to induce motion sickness and reveal its effects on the BBB. Mice were centrifuged at 2× g for 24 h. Fluorescent dextrans with different sizes (40, 70 and 150 kDa) and fluorescent antisense oligonucleotides (AS) were injected into mice retro-orbitally. The presence of fluorescent molecules was revealed by epifluorescence and confocal microscopies in brain slices. Gene expression was evaluated by RT-qPCR from brain extracts. Only the 70 kDa dextran and AS were detected in the parenchyma of several brain regions, suggesting an alteration in the BBB. Moreover, Ctnnd1, Gja4 and Actn1 were upregulated, whereas Jup, Tjp2, Gja1, Actn2, Actn4, Cdh2 and Ocln genes were downregulated, specifically suggesting a dysregulation in the tight junctions of endothelial cells forming the BBB. Our results confirm the alteration in the BBB after a short period of hypergravity exposure.
Collapse
Affiliation(s)
- David Dubayle
- CNRS, INCC, UMR 8002, Université Paris Cité, F-75006 Paris, France
- Correspondence: (D.D.); (J.-L.M.)
| | - Arnaud Vanden-Bossche
- INSERM, SAINBIOSE U1059, Mines Saint-Etienne, Université Jean Monnet Saint-Étienne, F-42023 Saint-Étienne, France
| | - Tom Peixoto
- University Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Jean-Luc Morel
- University Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
- University Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
- Correspondence: (D.D.); (J.-L.M.)
| |
Collapse
|
11
|
Li C, Pan Y, Tan Y, Wang Y, Sun X. PINK1-Dependent Mitophagy Reduced Endothelial Hyperpermeability and Cell Migration Capacity Under Simulated Microgravity. Front Cell Dev Biol 2022; 10:896014. [PMID: 35874841 PMCID: PMC9300855 DOI: 10.3389/fcell.2022.896014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
The effect of cardiovascular dysfunction including orthostatic intolerance and disability on physical exercise is one of the health problems induced by long-term spaceflight astronauts face. As an important part of vascular structure, the vascular endothelium, uniquely sensitive to mechanical force, plays a pivotal role in coordinating vascular functions. Our study found that simulated microgravity induced PINK1-dependent mitophagy in human umbilical vein endothelial cells (HUVECs). Here, we explored the underlying mechanism of mitophagy induction. The ER stress induced by proteostasis failure in HUVECs promoted the Ca2+ transfer from ER to mitochondria, resulting in mitochondria Ca2+ overload, decreased mitochondrial membrane potential, mitochondria fission, and accumulation of Parkin and p62 in mitochondria and mitophagy under simulated microgravity. Moreover, we assumed that mitophagy played a vital role in functional changes in endothelial cells under simulated microgravity. Using mdivi-1 and PINK1 knockdown, we found that NLRP3 inflammasome activation was enhanced after mitophagy was inhibited. The NLRP3 inflammasome contributed to endothelial hyperpermeability and cellular migration by releasing IL-1β. Thus, mitophagy inhibited cell migration ability and hyperpermeability in HUVECs exposed to clinostat-simulated microgravity. Collectively, we here clarify the mechanism of mitophagy induction by simulated microgravity in vitro and demonstrate the relationship between mitophagy and vascular endothelial functional changes including cellular migration and permeability. This study deepens the understanding of vascular functional changes under microgravity.
Collapse
Affiliation(s)
- Chengfei Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yikai Pan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yingjun Tan
- China Astronaut Research and Training Center, Beijing, China
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiqing Sun, , Yongchun Wang,
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiqing Sun, , Yongchun Wang,
| |
Collapse
|
12
|
Liu H, Zhang X, Liu Y, Xin N, Deng Y, Li Y. Semen Ziziphi Spinosae attenuates blood-brain barrier dysfunction induced by lipopolysaccharide by targeting the FAK-DOCK180-Rac1-WAVE2-Arp3 signaling pathway. NPJ Sci Food 2022; 6:27. [PMID: 35655066 PMCID: PMC9163036 DOI: 10.1038/s41538-022-00142-6] [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: 01/12/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022] Open
Abstract
Semen Ziziphi Spinosae (SZS) has been extensively used in the daily diet as a functional food for neuroprotective health-benefit in China for many years. However, the neuroprotective mechanism of SZS associated with blood–brain barrier (BBB) integrity remains unexplored. The present study suggests SZS could protect against lipopolysaccharide (LPS)-induced BBB dysfunction. Proteomics indicate that 135 proteins in rat brain are significantly altered by SZS. These differentially expressed proteins are mainly clustered into cell–cell adhesion and adherens junctions, which are closely related with BBB integrity. SZS reversed LPS-induces BBB breakdown by activating the FAK-DOCK180-Rac1-WAVE2-Arp3 pathway. Molecular docking between signaling pathway proteins and identified SZS components in rat plasma reveals that 6”‘-feruloylspinosin, spinosin, and swertisin strongly binds to signaling proteins at multiple amino acid sites. These novel findings suggest a health benefit of SZS in prevention of cerebral diseases and contributes to the further application of SZS as a functional food.
Collapse
Affiliation(s)
- Huayan Liu
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, China
| | - Xin Zhang
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, China
| | - Yujiao Liu
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, China
| | - Nian Xin
- BIT&GS Technologies Co. Ltd, 100074, Beijing, China
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, China.
| | - Yujuan Li
- School of Life Science, Beijing Institute of Technology, 100081, Beijing, China.
| |
Collapse
|
13
|
Zeng Y, Du X, Yao X, Qiu Y, Jiang W, Shen J, Li L, Liu X. Mechanism of cell death of endothelial cells regulated by mechanical forces. J Biomech 2021; 131:110917. [PMID: 34952348 DOI: 10.1016/j.jbiomech.2021.110917] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022]
Abstract
Cell death of endothelial cells (ECs) is a common devastating consequence of various vascular-related diseases. Atherosclerosis, hypertension, sepsis, diabetes, cerebral ischemia and cardiac ischemia/reperfusion injury, and chronic kidney disease remain major causes of morbidity and mortality worldwide, in which ECs are constantly subjected to a great amount of dynamic changed mechanical forces including shear stress, extracellular matrix stiffness, mechanical stretch and microgravity. A thorough understanding of the regulatory mechanisms by which the mechanical forces controlled the cell deaths including apoptosis, autophagy, and pyroptosis is crucial for the development of new therapeutic strategies. In the present review, experimental and clinical data highlight that nutrient depletion, oxidative stress, tumor necrosis factor-α, high glucose, lipopolysaccharide, and homocysteine possess cytotoxic effects in many tissues and induce apoptosis of ECs, and that sphingosine-1-phosphate protects ECs. Nevertheless, EC apoptosis in the context of those artificial microenvironments could be enhanced, reduced or even reversed along with the alteration of patterns of shear stress. An appropriate level of autophagy diminishes EC apoptosis to some extent, in addition to supporting cell survival upon microenvironment challenges. The intervention of pyroptosis showed a profound effect on atherosclerosis. Further cell and animal studies are required to ascertain whether the alterations in the levels of cell deaths and their associated regulatory mechanisms happen at local lesion sites with considerable mechanical force changes, for preventing senescence and cell deaths in the vascular-related diseases.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Xiaoqiang Du
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinghong Yao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Qiu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|