1
|
Cai Y, Yang S, Zhao J, Zheng G, Han Y, Zhang Y, Qin Y, Yang C, Xiong Q, Chu X, Ju C, Yin H, Shi Y, Jiang F, Yong H, Zhu Y. Mechanism Exploration of Dietary Supplement Astaxanthin on Improving Atherosclerosis through an Integrated Strategy Encompassing Artificial Intelligence Virtual Screening and Experimental Validation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11265-11287. [PMID: 40265257 DOI: 10.1021/acs.jafc.4c11894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Atherosclerosis (AS) is a major and common pathological basis of ischemic intestinal infarction, myocardial infarction, stroke, renal failure, and other highly lethal and disabling diseases. Current pharmacological interventions (e.g., statins) often cause adverse effects, limiting their long-term use. Natural compounds, with their multitarget efficacy and superior safety profiles, have emerged as promising alternatives for AS treatment. As a potent antioxidant carotenoid, astaxanthin exhibits unique therapeutic potential by simultaneously targeting inflammation, oxidative stress, and lipid metabolism, which are key drivers of AS pathogenesis. This study will systematically decipher astaxanthin's therapeutic mechanisms through an integrative strategy encompassing artificial intelligence virtual screening and experimental validation. Notably, five proteins, including CTSD, DPP4, FABP5, ITGAL, and MMP9, were identified as core targets for astaxanthin intervention in AS via network pharmacology and machine learning. Meanwhile, the results from molecular dynamic simulations confirmed that these core targets can stable binding with astaxanthin. Furthermore, in vitro experiments further validated astaxanthin can inhibit foam cell formation, restore redox balance, and suppress inflammation. Moreover, a close correlation has been found between them. These findings position astaxanthin as a multitarget natural agent to combat AS, addressing both efficacy advantage and safety concerns of current therapies.
Collapse
Affiliation(s)
- Yisa Cai
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, P. R. China
- Translational Institute for Cancer Pain, Chongming Hospital Affiliated to Shanghai University of Health & Medicine Sciences (Xinhua Hospital Chongming Branch), Shanghai 202155, P. R. China
| | - Shiyan Yang
- Department of Internal Medicine, Huaian Hospital Affiliated to Xuzhou Medical University, Huai'an 223002, Jiangsu, P. R. China
| | - Jiajiang Zhao
- Yunnan Hongqingfu Biotechnology Co., LTD., Kunming 650000, Yunnan, P. R. China
| | - Guangzhen Zheng
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, P. R. China
| | - Yun Han
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, Shandong, P. R. China
| | - Yuhan Zhang
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, P. R. China
| | - Yiyuan Qin
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, P. R. China
| | - Chao Yang
- Translational Institute for Cancer Pain, Chongming Hospital Affiliated to Shanghai University of Health & Medicine Sciences (Xinhua Hospital Chongming Branch), Shanghai 202155, P. R. China
| | - Qingping Xiong
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, P. R. China
| | - Xinyi Chu
- Yunnan Hongqingfu Biotechnology Co., LTD., Kunming 650000, Yunnan, P. R. China
| | - Chunhan Ju
- Yunnan Hongqingfu Biotechnology Co., LTD., Kunming 650000, Yunnan, P. R. China
| | - Huixia Yin
- Yunnan Hongqingfu Biotechnology Co., LTD., Kunming 650000, Yunnan, P. R. China
| | - Yingying Shi
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, P. R. China
| | - Feng Jiang
- Translational Institute for Cancer Pain, Chongming Hospital Affiliated to Shanghai University of Health & Medicine Sciences (Xinhua Hospital Chongming Branch), Shanghai 202155, P. R. China
| | - Hui Yong
- Department of Cardiology, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an 223000, Jiangsu, P. R. China
| | - Yong Zhu
- Jiangsu Key Laboratory of Regional Specific Resource Pharmaceutical Transformation, Huaiyin Institute of Technology, Huai'an 223003, Jiangsu, P. R. China
| |
Collapse
|
2
|
Marjan T, Lafuente-Gómez N, Rampal A, Mooney DJ, Peyton SR, Qazi TH. Cell-Instructive Biomaterials with Native-Like Biochemical Complexity. Annu Rev Biomed Eng 2025; 27:185-209. [PMID: 39874600 PMCID: PMC12045723 DOI: 10.1146/annurev-bioeng-120823-020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Biochemical signals in native tissue microenvironments instruct cell behavior during many biological processes ranging from developmental morphogenesis and tissue regeneration to tumor metastasis and disease progression. The detection and characterization of these signals using spatial and highly resolved quantitative methods have revealed their existence as matricellular proteins in the matrisome, some of which are bound to the extracellular matrix while others are freely diffusing. Including these biochemical signals in engineered biomaterials can impart enhanced functionality and native-like complexity, ultimately benefiting efforts to understand, model, and treat various diseases. In this review, we discuss advances in characterizing, mimicking, and harnessing biochemical signals in developing advanced engineered biomaterials. An overview of the diverse forms in which these biochemical signals exist and their effects on intracellular signal transduction is also provided. Finally, we highlight the application of biochemically complex biomaterials in the three broadly defined areas of tissue regeneration, immunoengineering, and organoid morphogenesis.
Collapse
Affiliation(s)
- Tuba Marjan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Nuria Lafuente-Gómez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Akaansha Rampal
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| |
Collapse
|
3
|
Tetz V, Kardava K, Vecherkovskaya M, Khodadadi-Jamayran A, Tsirigos A, Tetz G. Regulating white blood cell activity through the novel Universal Receptive System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631232. [PMID: 39896476 PMCID: PMC11785007 DOI: 10.1101/2025.01.06.631232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The understanding of the mechanisms that control key features of immune cells in various disease contexts remains limited, and few techniques are available for manipulating immune cells. Thus, discovering novel strategies for regulating immune cells is essential for gaining insight into their roles in health and disease. In this study, we investigated the potential of the recently described Universal Receptive System to regulate human immune cell functions. This was achieved for the first time by specifically targeting newly discovered surface-bound DNA and RNA-based receptors on leukocytes and generating "Leukocyte-Tells." This approach upregulated numerous genes related to immune cell signaling, migration, endocytosis, and phagocytosis pathways. The antimicrobial and anticancer activities of Leukocyte-Tells exceeded the activity of control leukocytes in vitro . In some settings, such as in antibiofilm experiments, the Leukocyte-Tells showed up to 1,000,000-fold higher activities than control leukocytes. Our findings reveal, for the first time, that the Universal Receptive System can orchestrate fundamental properties of immune cells, including enhanced antimicrobial and anti-tumor activities. This novel approach offers a new avenue for understanding the biology and regulation of white blood cells.
Collapse
|
4
|
Ubogu EE. Animal models of immune-mediated demyelinating polyneuropathies. Autoimmunity 2024; 57:2361745. [PMID: 38850571 PMCID: PMC11215812 DOI: 10.1080/08916934.2024.2361745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/13/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Immune-mediated demyelinating polyneuropathies (IMDPs) are rare disorders in which dysregulated adaptive immune responses cause peripheral nerve demyelinating inflammation and axonal injury in susceptible individuals. Despite significant advances in understanding IMDP pathogenesis guided by patient data and representative mammalian models, specific therapies are lacking. Significant knowledge gaps in IMDP pathogenesis still exist, e.g. precise antigen(s) and mechanisms that initially trigger immune system activation and identification of large population disease susceptibility factors. The initial directional cues for antigen-specific effector or autoreactive leukocyte trafficking into peripheral nerves are also unknown. An overview of current animal models, with emphasis on the experimental autoimmune neuritis and spontaneous autoimmune peripheral polyneuropathy models, is provided. Insights on the initial directional cues for peripheral nerve tissue specific autoimmunity using a novel Major Histocompatibility Complex class II conditional knockout mouse strain are also discussed, suggesting an essential research tool to study cell- and time-dependent adaptive immunity in autoimmune diseases.
Collapse
Affiliation(s)
- Eroboghene E Ubogu
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
5
|
Bauer L, Alkotub B, Ballmann M, Hasanzadeh Kafshgari M, Rammes G, Multhoff G. Cannabidiol (CBD) Protects Lung Endothelial Cells from Irradiation-Induced Oxidative Stress and Inflammation In Vitro and In Vivo. Cancers (Basel) 2024; 16:3589. [PMID: 39518030 PMCID: PMC11544820 DOI: 10.3390/cancers16213589] [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: 09/23/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Objective: Radiotherapy, which is commonly used for the local control of thoracic cancers, also induces chronic inflammatory responses in the microvasculature of surrounding normal tissues such as the lung and heart that contribute to fatal radiation-induced lung diseases (RILDs) such as pneumonitis and fibrosis. In this study, we investigated the potential of cannabidiol (CBD) to attenuate the irradiation damage to the vasculature. Methods: We investigated the ability of CBD to protect a murine endothelial cell (EC) line (H5V) and primary lung ECs isolated from C57BL/6 mice from irradiation-induced damage in vitro and lung ECs (luECs) in vivo, by measuring the induction of oxidative stress, DNA damage, apoptosis (in vitro), and induction of inflammatory and pro-angiogenic markers (in vivo). Results: We demonstrated that a non-lethal dose of CBD reduces the irradiation-induced oxidative stress and early apoptosis of lung ECs by upregulating the expression of the cytoprotective mediator heme-oxygenase-1 (HO-1). The radiation-induced increased expression of inflammatory (ICAM-2, MCAM) and pro-angiogenic (VE-cadherin, Endoglin) markers was significantly reduced by a continuous daily treatment of C57BL/6 mice with CBD (i.p. 20 mg/kg body weight), 2 weeks before and 2 weeks after a partial irradiation of the lung (less than 20% of the lung volume) with 16 Gy. Conclusions: CBD has the potential to improve the clinical outcome of radiotherapy by reducing toxic side effects on the microvasculature of the lung.
Collapse
Affiliation(s)
- Lisa Bauer
- Department of Radiation Oncology, TUM School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany;
- Radiation Immuno-Oncology Group, Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany;
| | - Bayan Alkotub
- Institute of Biological and Medical Imaging (IBMI), Helmholtz Zentrum München (HMGU), 85764 Neuherberg, Germany;
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany
| | - Markus Ballmann
- Department of Anesthesiology and Intensive Care Medicine, TUM School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany; (M.B.); (G.R.)
| | - Morteza Hasanzadeh Kafshgari
- Radiation Immuno-Oncology Group, Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany;
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine, TUM School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany; (M.B.); (G.R.)
| | - Gabriele Multhoff
- Department of Radiation Oncology, TUM School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany;
- Radiation Immuno-Oncology Group, Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, University Hospital of the Technical University of Munich (TUM), 81675 Munich, Germany;
| |
Collapse
|
6
|
Ubogu EE, Conner JA, Wang Y, Yadav D, Saunders TL. Development of a major histocompatibility complex class II conditional knockout mouse to study cell-specific and time-dependent adaptive immune responses in peripheral nerves. Muscle Nerve 2024; 70:420-433. [PMID: 38922958 DOI: 10.1002/mus.28193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION/AIMS The precise relationship between molecular mimicry and tissue-specific autoimmunity is unknown. Major histocompatibility complex (MHC) class II antigen presenting cell-CD4+ T-cell receptor complex interactions are necessary for adaptive immunity. This study aimed to determine the role of endoneurial endothelial cell MHC class II in autoimmune polyneuropathy. METHODS Cryopreserved Guillain-Barré syndrome (GBS) patient sural nerve biopsies and sciatic nerves from the severe murine experimental autoimmune neuritis (sm-EAN) GBS model were studied. Cultured conditional ready MHC Class II antigen A-alpha chain (H2-Aa) embryonic stem cells were used to generate H2-Aaflox/+ C57BL/6 mice. Mice were backcrossed and intercrossed to the SJL background to generate H2-Aaflox/flox SJL mice, bred with hemizygous Tamoxifen-inducible von Willebrand factor Cre recombinase (vWF-iCre/+) SJL mice to generate H2-Aaflox/flox; vWF-iCre/+ mice to study microvascular endothelial cell adaptive immune responses. Sm-EAN was induced in Tamoxifen-treated H2-Aaflox/flox; vWF-iCre/+, H2-Aaflox/flox; +/+, H2-Aa+/+; vWF-iCre/+ and untreated H2-Aaflox/flox; vWF-iCre/+ adult female SJL mice. Neurobehavioral, electrophysiological and histopathological assessments were performed at predefined time points. RESULTS Endoneurial endothelial cell MHC class II expression was observed in normal and inflamed human and mouse peripheral nerves. Tamoxifen-treated H2-Aaflox/flox; vWF-iCre/+ mice were resistant to sm-EAN despite extensive MHC class II expression in lymphoid and non-lymphoid tissues. DISCUSSION A conditional MHC class II knockout mouse to study cell- and time-dependent adaptive immune responses in vivo was developed. Initial studies show microvascular endothelial cell MHC class II expression is necessary for peripheral nerve specific autoimmunity, as advocated by human in vitro adaptive immunity and ex vivo transplant rejection studies.
Collapse
Affiliation(s)
- Eroboghene E Ubogu
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama, Birmingham, Alabama, USA
| | - Jeremy A Conner
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama, Birmingham, Alabama, USA
| | - Yimin Wang
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama, Birmingham, Alabama, USA
| | - Dinesh Yadav
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama, Birmingham, Alabama, USA
| | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
7
|
Di Petrillo A, Pintus F, Floris S, Tuberoso CIG, de Almeida ABABS, Giorno TBS, Fernandes PD, Boylan F. Effects of Euphorbia characias subsp. characias flower extracts on nociceptive pain and acute inflammatory models in mice. Fitoterapia 2024; 176:106002. [PMID: 38729245 DOI: 10.1016/j.fitote.2024.106002] [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: 09/20/2023] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/12/2024]
Abstract
Pain and inflammation are major health issues worldwide, leading to negative consequences. Despite several drugs being available to manage these conditions, their effectiveness can be limited by cost, adverse reactions, and potential tolerance and dependence with long-term use. Euphorbia characias traditionally used in folk medicine for its diverse biological activities - including antiproliferative, antimicrobial, and anti-inflammatory effects - has not been extensively studied in vivo for its analgesic and anti-inflammatory properties. In this study, the antinociceptive and anti-inflammatory properties of the water and ethanolic extracts of E. characias flowers (ECAEFl and ECEEFl) were evaluated using various models. Both extracts significantly reduced paw licking time in a formalin-induced paw licking model, with ECAEFl specifically targeting and ECEEFl affecting both the neurogenic and inflammatory phases. Additionally, in the carrageenan-induced cell migration model, both extracts showed a significant decrease in leukocyte migration, protein extravasation and nitric oxide levels, further demostrating their anti-inflammatory activity. High-Resolution HPLC-ESI-QTOF-MS-MS and HPLC-PDA analysis characterized the chemical composition of the extracts, identifying a significant presence of phenolic compounds, particularly quercetin and its derivatives, which likely contribute to the observed biological activities. These findings highlight the potential of E. characias extracts as natural sources of compounds with antinociceptive and anti-inflammatory properties. Further investigations are needed to elucidate the underlying mechanisms and explore their therapeutic potential in pain and inflammation-related disorders.
Collapse
Affiliation(s)
- Amalia Di Petrillo
- Department of Medical Science and Public Health, Gastroenterology Unit, University of Cagliari, Cittadella Universitaria di Monserrato, 09042 Monserrato, Cagliari, Italy.
| | - Francesca Pintus
- Department of Life and Environmental Sciences, University of Cagliari, SS 554-Bivio per Sestu, Monserrato, 09042, Cagliari, Italy.
| | - Sonia Floris
- Department of Life and Environmental Sciences, University of Cagliari, SS 554-Bivio per Sestu, Monserrato, 09042, Cagliari, Italy.
| | - Carlo Ignazio Giovanni Tuberoso
- Department of Life and Environmental Sciences, University of Cagliari, SS 554-Bivio per Sestu, Monserrato, 09042, Cagliari, Italy.
| | | | - Thais Biondino Sardella Giorno
- Laboratório de Farmacologia da Dor e da Inflamação, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil
| | - Patricia Dias Fernandes
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Fabio Boylan
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
8
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [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: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
9
|
Maina JN. A critical assessment of the cellular defences of the avian respiratory system: are birds in general and poultry in particular relatively more susceptible to pulmonary infections/afflictions? Biol Rev Camb Philos Soc 2023; 98:2152-2187. [PMID: 37489059 DOI: 10.1111/brv.13000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/01/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023]
Abstract
In commercial poultry farming, respiratory diseases cause high morbidities and mortalities, begetting colossal economic losses. Without empirical evidence, early observations led to the supposition that birds in general, and poultry in particular, have weak innate and adaptive pulmonary defences and are therefore highly susceptible to injury by pathogens. Recent findings have, however, shown that birds possess notably efficient pulmonary defences that include: (i) a structurally complex three-tiered airway arrangement with aerodynamically intricate air-flow dynamics that provide efficient filtration of inhaled air; (ii) a specialised airway mucosal lining that comprises air-filtering (ciliated) cells and various resident phagocytic cells such as surface and tissue macrophages, dendritic cells and lymphocytes; (iii) an exceptionally efficient mucociliary escalator system that efficiently removes trapped foreign agents; (iv) phagocytotic atrial and infundibular epithelial cells; (v) phagocytically competent surface macrophages that destroy pathogens and injurious particulates; (vi) pulmonary intravascular macrophages that protect the lung from the vascular side; and (vii) proficiently phagocytic pulmonary extravasated erythrocytes. Additionally, the avian respiratory system rapidly translocates phagocytic cells onto the respiratory surface, ostensibly from the subepithelial space and the circulatory system: the mobilised cells complement the surface macrophages in destroying foreign agents. Further studies are needed to determine whether the posited weak defence of the avian respiratory system is a global avian feature or is exclusive to poultry. This review argues that any inadequacies of pulmonary defences in poultry may have derived from exacting genetic manipulation(s) for traits such as rapid weight gain from efficient conversion of food into meat and eggs and the harsh environmental conditions and severe husbandry operations in modern poultry farming. To reduce pulmonary diseases and their severity, greater effort must be directed at establishment of optimal poultry housing conditions and use of more humane husbandry practices.
Collapse
Affiliation(s)
- John N Maina
- Department of Zoology, University of Johannesburg, Auckland Park Campus, Kingsway Avenue, Johannesburg, 2006, South Africa
| |
Collapse
|
10
|
Cleuren A, Molema G. Organotypic heterogeneity in microvascular endothelial cell responses in sepsis-a molecular treasure trove and pharmacological Gordian knot. Front Med (Lausanne) 2023; 10:1252021. [PMID: 38020105 PMCID: PMC10665520 DOI: 10.3389/fmed.2023.1252021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
In the last decades, it has become evident that endothelial cells (ECs) in the microvasculature play an important role in the pathophysiology of sepsis-associated multiple organ dysfunction syndrome (MODS). Studies on how ECs orchestrate leukocyte recruitment, control microvascular integrity and permeability, and regulate the haemostatic balance have provided a wealth of knowledge and potential molecular targets that could be considered for pharmacological intervention in sepsis. Yet, this information has not been translated into effective treatments. As MODS affects specific vascular beds, (organotypic) endothelial heterogeneity may be an important contributing factor to this lack of success. On the other hand, given the involvement of ECs in sepsis, this heterogeneity could also be leveraged for therapeutic gain to target specific sites of the vasculature given its full accessibility to drugs. In this review, we describe current knowledge that defines heterogeneity of organ-specific microvascular ECs at the molecular level and elaborate on studies that have reported EC responses across organ systems in sepsis patients and animal models of sepsis. We discuss hypothesis-driven, single-molecule studies that have formed the basis of our understanding of endothelial cell engagement in sepsis pathophysiology, and include recent studies employing high-throughput technologies. The latter deliver comprehensive data sets to describe molecular signatures for organotypic ECs that could lead to new hypotheses and form the foundation for rational pharmacological intervention and biomarker panel development. Particularly results from single cell RNA sequencing and spatial transcriptomics studies are eagerly awaited as they are expected to unveil the full spatiotemporal signature of EC responses to sepsis. With increasing awareness of the existence of distinct sepsis subphenotypes, and the need to develop new drug regimen and companion diagnostics, a better understanding of the molecular pathways exploited by ECs in sepsis pathophysiology will be a cornerstone to halt the detrimental processes that lead to MODS.
Collapse
Affiliation(s)
- Audrey Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Grietje Molema
- Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
11
|
Pineda-Castillo SA, Acar H, Detamore MS, Holzapfel GA, Lee CH. Modulation of Smooth Muscle Cell Phenotype for Translation of Tissue-Engineered Vascular Grafts. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:574-588. [PMID: 37166394 PMCID: PMC10618830 DOI: 10.1089/ten.teb.2023.0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Translation of small-diameter tissue-engineered vascular grafts (TEVGs) for the treatment of coronary artery disease (CAD) remains an unfulfilled promise. This is largely due to the limited integration of TEVGs into the native vascular wall-a process hampered by the insufficient smooth muscle cell (SMC) infiltration and extracellular matrix deposition, and low vasoactivity. These processes can be promoted through the judicious modulation of the SMC toward a synthetic phenotype to promote remodeling and vascular integration; however, the expression of synthetic markers is often accompanied by a decrease in the expression of contractile proteins. Therefore, techniques that can precisely modulate the SMC phenotypical behavior could have the potential to advance the translation of TEVGs. In this review, we describe the phenotypic diversity of SMCs and the different environmental cues that allow the modulation of SMC gene expression. Furthermore, we describe the emerging biomaterial approaches to modulate the SMC phenotype in TEVG design and discuss the limitations of current techniques. In addition, we found that current studies in tissue engineering limit the analysis of the SMC phenotype to a few markers, which are often the characteristic of early differentiation only. This limited scope has reduced the potential of tissue engineering to modulate the SMC toward specific behaviors and applications. Therefore, we recommend using the techniques presented in this review, in addition to modern single-cell proteomics analysis techniques to comprehensively characterize the phenotypic modulation of SMCs. Expanding the holistic potential of SMC modulation presents a great opportunity to advance the translation of living conduits for CAD therapeutics.
Collapse
Affiliation(s)
- Sergio A. Pineda-Castillo
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
| | - Handan Acar
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Michael S. Detamore
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- Department of Structural Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Chung-Hao Lee
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
12
|
Lörchner H, Cañes Esteve L, Góes ME, Harzenetter R, Brachmann N, Gajawada P, Günther S, Doll N, Pöling J, Braun T. Neutrophils for Revascularization Require Activation of CCR6 and CCL20 by TNFα. Circ Res 2023; 133:592-610. [PMID: 37641931 DOI: 10.1161/circresaha.123.323071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Activation of immune-inflammatory pathways involving TNFα (tumor necrosis factor alpha) signaling is critical for revascularization and peripheral muscle tissue repair after ischemic injury. However, mechanisms of TNFα-driven inflammatory cascades directing recruitment of proangiogenic immune cells to sites of ischemia are unknown. METHODS Muscle tissue revascularization after permanent femoral artery ligation was monitored in mutant mice by laser Doppler imaging and light sheet fluorescence microscopy. TNFα-mediated signaling and the role of the CCL20 (C-C motif chemokine ligand 20)-CCR6 (C-C chemokine receptor 6) axis for formation of new vessels was studied in vitro and in vivo using bone marrow transplantation, flow cytometry, as well as biochemical and molecular biological techniques. RESULTS TNFα-mediated activation of TNFR (tumor necrosis factor receptor) 1 but not TNFR2 was found to be required for postischemic muscle tissue revascularization. Bone marrow-derived CCR6+ neutrophil granulocytes were identified as a previously undescribed TNFα-induced population of proangiogenic neutrophils, characterized by increased expression of VEGFA (vascular endothelial growth factor A). Mechanistically, postischemic activation of TNFR1 induced expression of the CCL20 in vascular cells and promoted translocation of the CCL20 receptor CCR6 to the cell surface of neutrophils, essentially conditioning VEGFA-expressing proangiogenic neutrophils for CCL20-dependent recruitment to sites of ischemia. Moreover, impaired revascularization of ischemic peripheral muscle tissue in diabetic mice was associated with reduced numbers of proangiogenic neutrophils and diminished CCL20 expression. Administration of recombinant CCL20 enhanced recruitment of proangiogenic neutrophils and improved revascularization of diabetic ischemic skeletal muscles, which was sustained by sequential treatment with fluvastatin. CONCLUSIONS We demonstrate that site-specific activation of the CCL20-CCR6 axis via TNFα recruits proangiogenic VEGFA-expressing neutrophils to sites of ischemic injury for initiation of muscle tissue revascularization. The findings provide an attractive option for tissue revascularization, particularly under diabetic conditions.
Collapse
Affiliation(s)
- Holger Lörchner
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
- German Centre for Cardiovascular Research (DZHK), Frankfurt am Main, Germany (H.L., J.P.)
| | - Laia Cañes Esteve
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Maria Elisa Góes
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Roxanne Harzenetter
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Nathalie Brachmann
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Praveen Gajawada
- Department of Cardiac Surgery, Kerckhoff Heart Center, Bad Nauheim, Germany (P.G.)
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| | - Nicolas Doll
- Schüchtermann-Klinik, Bad Rothenfelde, Germany (N.D., J.P.)
| | - Jochen Pöling
- Schüchtermann-Klinik, Bad Rothenfelde, Germany (N.D., J.P.)
- German Centre for Cardiovascular Research (DZHK), Frankfurt am Main, Germany (H.L., J.P.)
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (H.L., L.C.E., M.E.G., R.H., N.B., S.G., T.B.)
| |
Collapse
|
13
|
Ubogu EE, Conner JA, Wang Y, Yadav D, Saunders TL. Development of a major histocompatibility complex class II conditional knockout mouse to study cell-specific and time-dependent adaptive immune responses in peripheral nerves. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550421. [PMID: 37546875 PMCID: PMC10402085 DOI: 10.1101/2023.07.24.550421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Introduction Major histocompatibility complex (MHC) class II professional antigen presenting cell-naïve CD4+ T cell interactions via the T-cell receptor complex are necessary for adaptive immunity. MHC class II upregulation in multiple cell types occurs in human autoimmune polyneuropathy patient biopsies, necessitating studies to ascertain cellular signaling pathways required for tissue-specific autoimmunity. Methods Cryopreserved Guillain-Barré syndrome (GBS) patient sural nerve biopsies and sciatic nerves from the severe murine experimental autoimmune neuritis (sm-EAN) GBS model were studied. Cultured conditional ready MHC Class II antigen A-alpha chain (H2-Aa) embryonic stem cells were used to generate H2-Aa flox/+ C57BL/6 mice. Mice were backcrossed and intercrossed to the SJL background to generate H2-Aa flox/flox SJL mice, bred with hemizygous Tamoxifen-inducible von Willebrand factor Cre recombinase (vWF-iCre/+) SJL mice to generate H2-Aa flox/flox ; vWF-iCre/+ to study microvascular endothelial cell adaptive immune responses. Sm-EAN was induced in adult female SJL Tamoxifen-treated H2-Aa flox/flox ; vWF-iCre/+ mice and H2-Aa flox/flox ; +/+ littermate controls. Neurobehavioral, electrophysiological and histopathological assessments were performed at predefined time points. Results Endoneurial endothelial cell MHC class II expression was observed in normal and inflamed human and mouse peripheral nerves. Adult female Tamoxifen-treated H2-Aa flox/flox ; vWF-iCre/+ did not develop sm-EAN despite extensive MHC class II expression in lymphoid and non-lymphoid tissues. Discussion A conditional MHC class II knockout mouse to study cell- and time-dependent adaptive immune responses in vivo is developed. Initial studies show microvascular endothelial cell MHC class II expression is necessary for peripheral nerve specific autoimmunity, as advocated by human in vitro adaptive immunity and ex vivo transplant rejection studies.
Collapse
|
14
|
Jakovija A, Chtanova T. Skin immunity in wound healing and cancer. Front Immunol 2023; 14:1060258. [PMID: 37398649 PMCID: PMC10312005 DOI: 10.3389/fimmu.2023.1060258] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/24/2023] [Indexed: 07/04/2023] Open
Abstract
The skin is the body's largest organ. It serves as a barrier to pathogen entry and the first site of immune defense. In the event of a skin injury, a cascade of events including inflammation, new tissue formation and tissue remodeling contributes to wound repair. Skin-resident and recruited immune cells work together with non-immune cells to clear invading pathogens and debris, and guide the regeneration of damaged host tissues. Disruption to the wound repair process can lead to chronic inflammation and non-healing wounds. This, in turn, can promote skin tumorigenesis. Tumors appropriate the wound healing response as a way of enhancing their survival and growth. Here we review the role of resident and skin-infiltrating immune cells in wound repair and discuss their functions in regulating both inflammation and development of skin cancers.
Collapse
Affiliation(s)
- Arnolda Jakovija
- Immunity Theme, Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent’s School of Medicine, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Tatyana Chtanova
- Immunity Theme, Garvan Institute of Medical Research, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
15
|
Bosseboeuf E, Chikh A, Chaker AB, Mitchell TP, Vignaraja D, Rajendrakumar R, Khambata RS, Nightingale TD, Mason JC, Randi AM, Ahluwalia A, Raimondi C. Neuropilin-1 interacts with VE-cadherin and TGFBR2 to stabilize adherens junctions and prevent activation of endothelium under flow. Sci Signal 2023; 16:eabo4863. [PMID: 37220183 PMCID: PMC7614756 DOI: 10.1126/scisignal.abo4863] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Linear and disturbed flow differentially regulate gene expression, with disturbed flow priming endothelial cells (ECs) for a proinflammatory, atheroprone expression profile and phenotype. Here, we investigated the role of the transmembrane protein neuropilin-1 (NRP1) in ECs exposed to flow using cultured ECs, mice with an endothelium-specific knockout of NRP1, and a mouse model of atherosclerosis. We demonstrated that NRP1 was a constituent of adherens junctions that interacted with VE-cadherin and promoted its association with p120 catenin, stabilizing adherens junctions and inducing cytoskeletal remodeling in alignment with the direction of flow. We also showed that NRP1 interacted with transforming growth factor-β (TGF-β) receptor II (TGFBR2) and reduced the plasma membrane localization of TGFBR2 and TGF-β signaling. NRP1 knockdown increased the abundance of proinflammatory cytokines and adhesion molecules, resulting in increased leukocyte rolling and atherosclerotic plaque size. These findings describe a role for NRP1 in promoting endothelial function and reveal a mechanism by which NRP1 reduction in ECs may contribute to vascular disease by modulating adherens junction signaling and promoting TGF-β signaling and inflammation.
Collapse
Affiliation(s)
- Emy Bosseboeuf
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Anissa Chikh
- Molecular and Clinical Sciences Research Institute, St. George’s, University of London, London SW17 0RE, UK
| | - Ahmed Bey Chaker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Tom P. Mitchell
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Dhilakshani Vignaraja
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Ridhi Rajendrakumar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Rayomand S. Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Thomas D. Nightingale
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre for Microvascular Research, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Justin C. Mason
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Anna M. Randi
- Vascular Sciences, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Claudio Raimondi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Centre of Cardiovascular Medicine and Devices, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
16
|
Stierschneider A, Neuditschko B, Colleselli K, Hundsberger H, Herzog F, Wiesner C. Comparative and Temporal Characterization of LPS and Blue-Light-Induced TLR4 Signal Transduction and Gene Expression in Optogenetically Manipulated Endothelial Cells. Cells 2023; 12:697. [PMID: 36899833 PMCID: PMC10000987 DOI: 10.3390/cells12050697] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
In endothelial cells (ECs), stimulation of Toll-like receptor 4 (TLR4) by the endotoxin lipopolysaccharide (LPS) induces the release of diverse pro-inflammatory mediators, beneficial in controlling bacterial infections. However, their systemic secretion is a main driver of sepsis and chronic inflammatory diseases. Since distinct and rapid induction of TLR4 signaling is difficult to achieve with LPS due to the specific and non-specific affinity to other surface molecules and receptors, we engineered new light-oxygen-voltage-sensing (LOV)-domain-based optogenetic endothelial cell lines (opto-TLR4-LOV LECs and opto-TLR4-LOV HUVECs) that allow fast, precise temporal, and reversible activation of TLR4 signaling pathways. Using quantitative mass-spectrometry, RT-qPCR, and Western blot analysis, we show that pro-inflammatory proteins were not only expressed differently, but also had a different time course when the cells were stimulated with light or LPS. Additional functional assays demonstrated that light induction promoted chemotaxis of THP-1 cells, disruption of the EC monolayer and transmigration. In contrast, ECs incorporating a truncated version of the TLR4 extracellular domain (opto-TLR4 ΔECD2-LOV LECs) revealed high basal activity with fast depletion of the cell signaling system upon illumination. We conclude that the established optogenetic cell lines are well suited to induce rapid and precise photoactivation of TLR4, allowing receptor-specific studies.
Collapse
Affiliation(s)
- Anna Stierschneider
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria
| | - Benjamin Neuditschko
- Institute Krems Bioanalytics, IMC University of Applied Sciences, 3500 Krems, Austria
| | - Katrin Colleselli
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria
| | - Harald Hundsberger
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria
| | - Franz Herzog
- Institute Krems Bioanalytics, IMC University of Applied Sciences, 3500 Krems, Austria
| | - Christoph Wiesner
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria
| |
Collapse
|
17
|
Torres-Rêgo M, Aquino-Vital AKSD, Cavalcanti FF, Rocha EEA, Daniele-Silva A, Furtado AA, Silva DPD, Ururahy MAG, Silveira ER, Fernandes-Pedrosa MDF, Araújo RM. Phytochemical analysis and preclinical toxicological, antioxidant, and anti-inflammatory evaluation of hydroethanol extract from the roots of Harpalyce brasiliana Benth (Leguminosae). JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115364. [PMID: 35551979 DOI: 10.1016/j.jep.2022.115364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/28/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Harpalyce brasiliana Benth (Leguminosae) is a shrub endemic to Brazil, popularly known as "snake's root." This species is used in folk medicine for the treatment of inflammation and snakebites. However, up to now there is no scientific research to justify its popular use. The study aimed to characterize the phytochemical profile of the hydroethanol extract from the roots of H. brasiliana (Hb), to evaluate its antioxidant and anti-inflammatory potential, as well as to investigate its cytotoxicity and acute toxicity. MATERIALS AND METHODS The extract was obtained by maceration method using a solution of ethanol:water (70: 30, v/v). The phytochemical profile was obtained by liquid chromatography coupled to mass spectrometry. The cytotoxicity of extract (31-2000 μg/mL) was evaluated in vitro, by the 3-methyl-[4-5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) method using murine macrophage and fibroblast cell lines (RAW 247.6 and 3T3, respectively) and by the hemolytic assay. For the in vivo acute toxicity, the extract (2000 mg/kg) was administered and after 14 days the weight (body and organs) and hematological and biochemical parameters were analyzed. Chemical free radical scavenging effect of the extract (125-2000 μg/mL) was investigated through diphenylpicryl hydrazine reduction, total antioxidant capacity, reducing power, hydroxyl radical scavenging, and iron and copper chelating assays. In vitro anti-inflammatory effect of the extract (125, 500, and 2000 μg/mL) was demonstrated through of nitric oxide (NO) analyzed in lipopolysaccharides stimulated RAW 264.7 cells. In vivo anti-inflammatory activities were evaluated in carrageenan-induced paw edema and zymosan-air-pouch models, with gavage administration (post-treatment) of extract at 100, 200, and 400 mg/kg. For the first animal model, the anti-edematogenic activity and myeloperoxidase (MPO) levels were investigated, while in the zymosan-air-pouch model the leukocyte number, MPO, total protein and pro-inflammatory cytokine (IL-1β, IL-6, and TNF-α) levels were quantified. In addition, the oxidative parameters such as malondialdehyde (MDA) and reduced glutathione (GSH) were determined. RESULTS The phytochemical profile revealed the presence of 20 compounds, mainly prenylated and geranylated pterocarpans. The extract demonstrated no cytotoxicity in erythrocytes, macrophages and fibroblasts cells at the tested concentrations, as well as no sign of toxicity and mortality or significant alterations on the hematological and biochemical parameters in the acute toxicity model. The extract was also able to neutralize chemical free radicals, with copper and iron chelating effect. For the NO dosage, the extract evidenced the reduction of expression of NO after the administration of the extract (500 and 2000 μg/mL). The edematogenic model revealed a decrease in paw edema and MPO level, while the zymosan-air-pouch model evidenced a reduction of leukocyte number (especially of polymorphornuclears), MPO production, and total protein and cytokine levels, and demonstrated the antioxidant effect through a decrease in MDA and increase in GSH parameters. CONCLUSION This approach demonstrates for the first time that Hb is not cytotoxic, has low acute toxicity, and possesses antioxidant and anti-inflammatory properties in preclinical analyses, corroborating its popular use.
Collapse
Affiliation(s)
- Manoela Torres-Rêgo
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil; Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Ana Karoline Silva de Aquino-Vital
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Felipe França Cavalcanti
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Enos Emanuel Azevedo Rocha
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Alessandra Daniele-Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Allanny Alves Furtado
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Diana Pontes da Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Marcela Abbott Galvão Ururahy
- Department of Clinical Analysis and Toxicology, College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Edilberto Rocha Silveira
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza, Humberto Monte Street, S/N, Campus Pici, Pici, Fortaleza, 60021-970, Brazil.
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Renata Mendonça Araújo
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| |
Collapse
|
18
|
Lisovska N. Multilevel mechanism of immune checkpoint inhibitor action in solid tumors: History, present issues and future development (Review). Oncol Lett 2022; 23:190. [PMID: 35527781 PMCID: PMC9073577 DOI: 10.3892/ol.2022.13310] [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: 01/19/2022] [Accepted: 03/31/2022] [Indexed: 11/12/2022] Open
Abstract
Immunotherapy with checkpoint inhibitors (antibodies that target and block immune checkpoints in the tumor microenvironment) is included in the standard of care for patients with different types of malignancy, such as melanoma, renal cell and urothelial carcinoma, lung cancer etc. The introduction of this new immunotherapy has altered the view on potential targets for treatment of solid tumors from tumor cells themselves to their immune microenvironment; this has led to a reconsideration of the mechanisms of tumor-associated immunity. However, only a subset of patients benefit from immunotherapy and patient response is often unpredictable, even with known initial levels of prognostic markers; the biomarkers for favorable response are still being investigated. Mechanisms of immune checkpoint inhibitors efficiency, as well as the origins of treatment failure, require further investigation. From a clinical standpoint, discrepancies between the theoretical explanation of inhibitors of immune checkpoint actions at the cellular level and their deployment at a tissue/organ level impede the effective clinical implementation of novel immune therapy. The present review assessed existing experimental and clinical data on functional activity of inhibitors of immune checkpoints to provide a more comprehensive picture of their mechanisms of action on a cellular and higher levels of biological organization.
Collapse
Affiliation(s)
- Natalya Lisovska
- Chemotherapy Department, Center of Oncology, ‘Cyber Clinic of Spizhenko’, Kapitanovka, Kyiv 08112, Ukraine
| |
Collapse
|
19
|
Ling J, Chan BCL, Tsang MSM, Gao X, Leung PC, Lam CWK, Hu JM, Wong CK. Current Advances in Mechanisms and Treatment of Dry Eye Disease: Toward Anti-inflammatory and Immunomodulatory Therapy and Traditional Chinese Medicine. Front Med (Lausanne) 2022; 8:815075. [PMID: 35111787 PMCID: PMC8801439 DOI: 10.3389/fmed.2021.815075] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Dry eye is currently one of the most common ocular surface disease. It can lead to ocular discomfort and even cause visual impairment, which greatly affects the work and quality of life of patients. With the increasing incidence of dry eye disease (DED) in recent years, the disease is receiving more and more attention, and has become one of the hot research fields in ophthalmology research. Recently, with the in-depth research on the etiology, pathogenesis and treatment of DED, it has been shown that defects in immune regulation is one of the main pathological mechanisms of DED. Since the non-specific and specific immune response of the ocular surface are jointly regulated, a variety of immune cells and inflammatory factors are involved in the development of DED. The conventional treatment of DED is the application of artificial tears for lubricating the ocular surface. However, for moderate-to-severe DED, treatment with anti-inflammatory drugs is necessary. In this review, the immunomodulatory mechanisms of DED and the latest research progress of its related treatments including Chinese medicine will be discussed.
Collapse
Affiliation(s)
- Jiawei Ling
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung-Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Miranda Sin-Man Tsang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xun Gao
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ping Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher Wai-Kei Lam
- Faculty of Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Jiang-Miao Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Chun Kwok Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Endothelial ACKR3 drives atherosclerosis by promoting immune cell adhesion to vascular endothelium. Basic Res Cardiol 2022; 117:30. [PMID: 35674847 PMCID: PMC9177477 DOI: 10.1007/s00395-022-00937-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 01/31/2023]
Abstract
Atherosclerosis is the foundation of potentially fatal cardiovascular diseases and it is characterized by plaque formation in large arteries. Current treatments aimed at reducing atherosclerotic risk factors still allow room for a large residual risk; therefore, novel therapeutic candidates targeting inflammation are needed. The endothelium is the starting point of vascular inflammation underlying atherosclerosis and we could previously demonstrate that the chemokine axis CXCL12-CXCR4 plays an important role in disease development. However, the role of ACKR3, the alternative and higher affinity receptor for CXCL12 remained to be elucidated. We studied the role of arterial ACKR3 in atherosclerosis using western diet-fed Apoe-/- mice lacking Ackr3 in arterial endothelial as well as smooth muscle cells. We show for the first time that arterial endothelial deficiency of ACKR3 attenuates atherosclerosis as a result of diminished arterial adhesion as well as invasion of immune cells. ACKR3 silencing in inflamed human coronary artery endothelial cells decreased adhesion molecule expression, establishing an initial human validation of ACKR3's role in endothelial adhesion. Concomitantly, ACKR3 silencing downregulated key mediators in the MAPK pathway, such as ERK1/2, as well as the phosphorylation of the NF-kB p65 subunit. Endothelial cells in atherosclerotic lesions also revealed decreased phospho-NF-kB p65 expression in ACKR3-deficient mice. Lack of smooth muscle cell-specific as well as hematopoietic ACKR3 did not impact atherosclerosis in mice. Collectively, our findings indicate that arterial endothelial ACKR3 fuels atherosclerosis by mediating endothelium-immune cell adhesion, most likely through inflammatory MAPK and NF-kB pathways.
Collapse
|
21
|
Nandi SS, Gohil T, Sawant SA, Lambe UP, Ghosh S, Jana S. CD155: A Key Receptor Playing Diversified Roles. Curr Mol Med 2021; 22:594-607. [PMID: 34514998 DOI: 10.2174/1566524021666210910112906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 11/22/2022]
Abstract
Cluster of differentiation (CD155), formerly identified as poliovirus receptor (PVR) and later as immunoglobulin molecule involved in cell adhesion, proliferation, invasion and migration. It is a surface protein expressed mostly on normal and transformed malignant cells. The expression of the receptor varies based on the origin of tissue. The expression of the protein is determined by factors involved in sonic hedgehog pathway, Ras-MEK-ERK pathway and during stress conditions like DNA damage response. The protein uses alternate splicing mechanism, producing four isoforms - two being soluble (CD155β and CD155γ) and two being transmembrane protein (CD155α and CD155δ). Apart from being a viral receptor, researchers have identified CD155 having important roles in cancer research and cell signaling field. The receptor is recognized as biomarker for identifying cancerous tissue. The receptor interacts with molecules involved in cells defense mechanism. The immune-surveillance role of CD155 is being deciphered to understand the mechanistic approach it utilizes as onco-immunologic molecule. CD155 is a non-MHC-I ligand which helps in identifying non-self to NK cells via an inhibitory TIGIT ligand. The TIGIT-CD155 pathway is a novel MHC-I-independent education mechanism for cell tolerance and activation of NK cell. The receptor also has a role in metastasis of cancer and trans endothelial mechanism. In this review, authors discuss the virus-host interaction that occurs via single transmembrane receptor, the poliovirus infection pathway, which is being exploited as therapeutic pathway. The oncolytic virotherapy is now promising way for curing cancer.
Collapse
Affiliation(s)
- Shyam Sundar Nandi
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Trupti Gohil
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Sonali Ankush Sawant
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Upendra Pradeep Lambe
- National Institute of Virology, (Mumbai unit), (Formerly Enterovirus Research Centre). Haffkine Institute Compound, Indian Council of Medical Research, A. D. Marg, Parel. Mumbai-12. India
| | - Sudip Ghosh
- Molecular Biology Division, ICMR-National Institute of Nutrition, Jamai-Osmania PO, Hyderabad. India
| | - Snehasis Jana
- Trivedi Science Research Laboratory Pvt Ltd., Thane-West, Maharashtra-400604. India
| |
Collapse
|
22
|
Angom RS, Zhu J, Wu ATH, Sumitra MR, Pham V, Dutta S, Wang E, Madamsetty VS, Perez-Cordero GD, Huang HS, Mukhopadhyay D, Wang Y. LCC-09, a Novel Salicylanilide Derivative, Exerts Anti-Inflammatory Effect in Vascular Endothelial Cells. J Inflamm Res 2021; 14:4551-4565. [PMID: 34526801 PMCID: PMC8436973 DOI: 10.2147/jir.s305168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/07/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Endothelial cell (EC) activation facilitates leukocyte adhesion to vascular walls, which is implicated in a variety of cardiovascular diseases and is a target for prevention and treatment. Despite the development of anti-inflammatory medications, cost-effective therapies with significant anti-inflammatory effects and lower organ toxicity remain elusive. The goal of this study is to identify novel synthetic compounds that inhibit EC inflammatory response with minimal organ toxicity. METHODS AND RESULTS In this study, we discovered LCC-09, a salicylanilide derivative consisting of the functional fragment of magnolol, 2,4-difluorophenyl, and paeonol moiety of salicylate, as a novel anti-inflammatory compound in cultured ECs and zebrafish model. LCC-09 was shown to inhibit pro-inflammatory cytokine tumor necrosis factor-α (TNFα)-induced expression of adhesion molecules and inflammatory cytokines, leading to reduced leukocyte adhesion to ECs. Mechanistically, LCC-09 inhibits the phosphorylation of signal transducer and activator of transcription 1 (STAT1), TNFα-induced degradation of NF-κ-B Inhibitor-α (IκBα) and phosphorylation of NFκB p65, resulting in reduced NFκB transactivation activity and binding to E-selectin promoter. Additionally, LCC-09 attenuated TNFα-induced generation of reactive oxygen species in ECs. Molecular docking models suggest the binding of LCC-09 to NFκB essential modulator (NEMO) and Janus tyrosine kinase (JAK) may lead to dual inhibition of NFκB and STAT1. Furthermore, the anti-inflammatory effect of LCC-09 was validated in the lipopolysaccharides (LPS)-induced inflammation model in zebrafish. Our results demonstrated that LCC-09 significantly reduced the LPS-induced leukocyte recruitment and mortality of zebrafish embryos. Finally, LCC-09 was administered to cultured ECs and zebrafish embryos and showed minimal toxicities. CONCLUSION Our results support that LCC-09 inhibits EC inflammatory response but does not elicit significant toxicity.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Jian Zhu
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
| | - Alexander T H Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Maryam Rachmawati Sumitra
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
| | - Victoria Pham
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Gabriel D Perez-Cordero
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Hsu-Shan Huang
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ying Wang
- Department of Cardiovascular Medicine, College of Medicine and Science, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
23
|
Wu YC, Sonninen TM, Peltonen S, Koistinaho J, Lehtonen Š. Blood-Brain Barrier and Neurodegenerative Diseases-Modeling with iPSC-Derived Brain Cells. Int J Mol Sci 2021; 22:7710. [PMID: 34299328 PMCID: PMC8307585 DOI: 10.3390/ijms22147710] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) regulates the delivery of oxygen and important nutrients to the brain through active and passive transport and prevents neurotoxins from entering the brain. It also has a clearance function and removes carbon dioxide and toxic metabolites from the central nervous system (CNS). Several drugs are unable to cross the BBB and enter the CNS, adding complexity to drug screens targeting brain disorders. A well-functioning BBB is essential for maintaining healthy brain tissue, and a malfunction of the BBB, linked to its permeability, results in toxins and immune cells entering the CNS. This impairment is associated with a variety of neurological diseases, including Alzheimer's disease and Parkinson's disease. Here, we summarize current knowledge about the BBB in neurodegenerative diseases. Furthermore, we focus on recent progress of using human-induced pluripotent stem cell (iPSC)-derived models to study the BBB. We review the potential of novel stem cell-based platforms in modeling the BBB and address advances and key challenges of using stem cell technology in modeling the human BBB. Finally, we highlight future directions in this area.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Tuuli-Maria Sonninen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Sanni Peltonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
24
|
De Fazio L, Beghetti I, Bertuccio SN, Marsico C, Martini S, Masetti R, Pession A, Corvaglia L, Aceti A. Necrotizing Enterocolitis: Overview on In Vitro Models. Int J Mol Sci 2021; 22:6761. [PMID: 34201786 PMCID: PMC8268427 DOI: 10.3390/ijms22136761] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a gut inflammatory disorder which constitutes one of the leading causes of morbidity and mortality for preterm infants. The pathophysiology of NEC is yet to be fully understood; several observational studies have led to the identification of multiple factors involved in the pathophysiology of the disease, including gut immaturity and dysbiosis of the intestinal microbiome. Given the complex interactions between microbiota, enterocytes, and immune cells, and the limited access to fetal human tissues for experimental studies, animal models have long been essential to describe NEC mechanisms. However, at present there is no animal model perfectly mimicking human NEC; furthermore, the disease mechanisms appear too complex to be studied in single-cell cultures. Thus, researchers have developed new approaches in which intestinal epithelial cells are exposed to a combination of environmental and microbial factors which can potentially trigger NEC. In addition, organoids have gained increasing attention as promising models for studying NEC development. Currently, several in vitro models have been proposed and have contributed to describe the disease in deeper detail. In this paper, we will provide an updated review of available in vitro models of NEC and an overview of current knowledge regarding its molecular underpinnings.
Collapse
Affiliation(s)
- Luigia De Fazio
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Isadora Beghetti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Salvatore Nicola Bertuccio
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Concetta Marsico
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Silvia Martini
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Riccardo Masetti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Andrea Pession
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Pediatric Oncology and Hematology “Lalla Seragnoli”, Pediatric Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| | - Arianna Aceti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (L.D.F.); (S.N.B.); (C.M.); (S.M.); (R.M.); (A.P.); (L.C.); (A.A.)
- Neonatal Intensive Care Unit-IRCCS Azienda Ospedaliero-Universitaria, 40138 Bologna, Italy
| |
Collapse
|
25
|
Chaerkady R, Zhou Y, Delmar JA, Weng SHS, Wang J, Awasthi S, Sims D, Bowen MA, Yu W, Cazares LH, Sims GP, Hess S. Characterization of Citrullination Sites in Neutrophils and Mast Cells Activated by Ionomycin via Integration of Mass Spectrometry and Machine Learning. J Proteome Res 2021; 20:3150-3164. [PMID: 34008986 DOI: 10.1021/acs.jproteome.1c00028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Citrullination is an important post-translational modification implicated in many diseases including rheumatoid arthritis (RA), Alzheimer's disease, and cancer. Neutrophil and mast cells have different expression profiles for protein-arginine deiminases (PADs), and ionomycin-induced activation makes them an ideal cellular model to study proteins susceptible to citrullination. We performed high-resolution mass spectrometry and stringent data filtration to identify citrullination sites in neutrophil and mast cells treated with and without ionomycin. We identified a total of 833 validated citrullination sites on 395 proteins. Several of these citrullinated proteins are important components of pathways involved in innate immune responses. Using this benchmark primary sequence data set, we developed machine learning models to predict citrullination in neutrophil and mast cell proteins. We show that our models predict citrullination likelihood with 0.735 and 0.766 AUCs (area under the receiver operating characteristic curves), respectively, on independent validation sets. In summary, this study provides the largest number of validated citrullination sites in neutrophil and mast cell proteins. The use of our novel motif analysis approach to predict citrullination sites will facilitate the discovery of novel protein substrates of protein-arginine deiminases (PADs), which may be key to understanding immunopathologies of various diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michael A Bowen
- Antibody Discovery and Protein Engineering (ADPE), R&D AstraZeneca, Gaithersburg, Maryland 20878, United States
| | | | | | | | | |
Collapse
|
26
|
Aoun L, Nègre P, Gonsales C, Seveau de Noray V, Brustlein S, Biarnes-Pelicot M, Valignat MP, Theodoly O. Leukocyte transmigration and longitudinal forward-thrusting force in a microfluidic Transwell device. Biophys J 2021; 120:2205-2221. [PMID: 33838136 DOI: 10.1016/j.bpj.2021.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 01/21/2023] Open
Abstract
Transmigration of leukocytes across blood vessels walls is a critical step of the immune response. Transwell assays examine transmigration properties in vitro by counting cells passages through a membrane; however, the difficulty of in situ imaging hampers a clear disentanglement of the roles of adhesion, chemokinesis, and chemotaxis. We used here microfluidic Transwells to image the cells' transition from 2D migration on a surface to 3D migration in a confining microchannel and measure cells longitudinal forward-thrusting force in microchannels. Primary human effector T lymphocytes adhering with integrins LFA-1 (αLβ2) had a marked propensity to transmigrate in Transwells without chemotactic cue. Both adhesion and contractility were important to overcome the critical step of nucleus penetration but were remarkably dispensable for 3D migration in smooth microchannels deprived of topographic features. Transmigration in smooth channels was qualitatively consistent with a propulsion by treadmilling of cell envelope and squeezing of cell trailing edge. Stalling conditions of 3D migration were then assessed by imposing pressure drops across microchannels. Without specific adhesion, the cells slid backward with subnanonewton forces, showing that 3D migration under stress is strongly limited by a lack of adhesion and friction with channels. With specific LFA-1 mediated adhesion, stalling occurred at around 3 and 6 nN in 2 × 4 and 4 × 4 μm2 channels, respectively, supporting that stalling of adherent cells was under pressure control rather than force control. The stall pressure of 4 mbar is consistent with the pressure of actin filament polymerization that mediates lamellipod growth. The arrest of adherent cells under stress therefore seems controlled by the compression of the cell leading edge, which perturbs cells front-rear polarization and triggers adhesion failure or polarization reversal. Although stalling assays in microfluidic Transwells do not mimic in vivo transmigration, they provide a powerful tool to scrutinize 2D and 3D migration, barotaxis, and chemotaxis.
Collapse
Affiliation(s)
- Laurene Aoun
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Paulin Nègre
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Cristina Gonsales
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Sophie Brustlein
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Marie-Pierre Valignat
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Olivier Theodoly
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
27
|
Zhao Y, Ting KK, Coleman P, Qi Y, Chen J, Vadas M, Gamble J. The Tumour Vasculature as a Target to Modulate Leucocyte Trafficking. Cancers (Basel) 2021; 13:cancers13071724. [PMID: 33917287 PMCID: PMC8038724 DOI: 10.3390/cancers13071724] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/29/2021] [Accepted: 04/03/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Tumour blood vessels, characterised by abnormal morphology and function, create an immunosuppressive tumour microenvironment via restricting the appropriate leucocyte subsets trafficking. Strategies to trigger phenotypic alteration in tumour vascular system to resemble normal vascular system, named vascular normalisation, promote effective trafficking of leucocytes into tumours through enhancing the interactions between leucocytes and endothelial cells. This review specifically demonstrates how targeting tumour blood vessels modulates the critical steps of leucocyte trafficking. Furthermore, selective regulation of leucocyte subsets trafficking in tumours can be achieved by vasculature-targeting strategies, contributing to improved immunotherapy and thereby delayed tumour progression. Abstract The effectiveness of immunotherapy against solid tumours is dependent on the appropriate leucocyte subsets trafficking and accumulating in the tumour microenvironment (TME) with recruitment occurring at the endothelium. Such recruitment involves interactions between the leucocytes and the endothelial cells (ECs) of the vessel and occurs through a series of steps including leucocyte capture, their rolling, adhesion, and intraluminal crawling, and finally leucocyte transendothelial migration across the endothelium. The tumour vasculature can curb the trafficking of leucocytes through influencing each step of the leucocyte recruitment process, ultimately producing an immunoresistant microenvironment. Modulation of the tumour vasculature by strategies such as vascular normalisation have proven to be efficient in facilitating leucocyte trafficking into tumours and enhancing immunotherapy. In this review, we discuss the underlying mechanisms of abnormal tumour vasculature and its impact on leucocyte trafficking, and potential strategies for overcoming the tumour vascular abnormalities to boost immunotherapy via increasing leucocyte recruitment.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Correspondence: (Y.Z.); (J.G.); Tel.: +86-025-85811237 (Y.Z.); +61-02-95656225 (J.G.)
| | - Ka Ka Ting
- Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney 2050, Australia; (K.K.T.); (P.C.); (Y.Q.); (M.V.)
| | - Paul Coleman
- Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney 2050, Australia; (K.K.T.); (P.C.); (Y.Q.); (M.V.)
| | - Yanfei Qi
- Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney 2050, Australia; (K.K.T.); (P.C.); (Y.Q.); (M.V.)
| | - Jinbiao Chen
- Liver Injury and Cancer Program, Centenary Institute, The University of Sydney, Sydney 2050, Australia;
| | - Mathew Vadas
- Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney 2050, Australia; (K.K.T.); (P.C.); (Y.Q.); (M.V.)
| | - Jennifer Gamble
- Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney 2050, Australia; (K.K.T.); (P.C.); (Y.Q.); (M.V.)
- Correspondence: (Y.Z.); (J.G.); Tel.: +86-025-85811237 (Y.Z.); +61-02-95656225 (J.G.)
| |
Collapse
|
28
|
Schwartz AB, Campos OA, Criado-Hidalgo E, Chien S, del Álamo JC, Lasheras JC, Yeh YT. Elucidating the Biomechanics of Leukocyte Transendothelial Migration by Quantitative Imaging. Front Cell Dev Biol 2021; 9:635263. [PMID: 33855018 PMCID: PMC8039384 DOI: 10.3389/fcell.2021.635263] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
Leukocyte transendothelial migration is crucial for innate immunity and inflammation. Upon tissue damage or infection, leukocytes exit blood vessels by adhering to and probing vascular endothelial cells (VECs), breaching endothelial cell-cell junctions, and transmigrating across the endothelium. Transendothelial migration is a critical rate-limiting step in this process. Thus, leukocytes must quickly identify the most efficient route through VEC monolayers to facilitate a prompt innate immune response. Biomechanics play a decisive role in transendothelial migration, which involves intimate physical contact and force transmission between the leukocytes and the VECs. While quantifying these forces is still challenging, recent advances in imaging, microfabrication, and computation now make it possible to study how cellular forces regulate VEC monolayer integrity, enable efficient pathfinding, and drive leukocyte transmigration. Here we review these recent advances, paying particular attention to leukocyte adhesion to the VEC monolayer, leukocyte probing of endothelial barrier gaps, and transmigration itself. To offer a practical perspective, we will discuss the current views on how biomechanics govern these processes and the force microscopy technologies that have enabled their quantitative analysis, thus contributing to an improved understanding of leukocyte migration in inflammatory diseases.
Collapse
Affiliation(s)
- Amy B. Schwartz
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Obed A. Campos
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
29
|
Ivan DC, Walthert S, Berve K, Steudler J, Locatelli G. Dwellers and Trespassers: Mononuclear Phagocytes at the Borders of the Central Nervous System. Front Immunol 2021; 11:609921. [PMID: 33746939 PMCID: PMC7973121 DOI: 10.3389/fimmu.2020.609921] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/29/2020] [Indexed: 01/02/2023] Open
Abstract
The central nervous system (CNS) parenchyma is enclosed and protected by a multilayered system of cellular and acellular barriers, functionally separating glia and neurons from peripheral circulation and blood-borne immune cells. Populating these borders as dynamic observers, CNS-resident macrophages contribute to organ homeostasis. Upon autoimmune, traumatic or neurodegenerative inflammation, these phagocytes start playing additional roles as immune regulators contributing to disease evolution. At the same time, pathological CNS conditions drive the migration and recruitment of blood-borne monocyte-derived cells across distinct local gateways. This invasion process drastically increases border complexity and can lead to parenchymal infiltration of blood-borne phagocytes playing a direct role both in damage and in tissue repair. While recent studies and technical advancements have highlighted the extreme heterogeneity of these resident and CNS-invading cells, both the compartment-specific mechanism of invasion and the functional specification of intruding and resident cells remain unclear. This review illustrates the complexity of mononuclear phagocytes at CNS interfaces, indicating how further studies of CNS border dynamics are crucially needed to shed light on local and systemic regulation of CNS functions and dysfunctions.
Collapse
|
30
|
Peelen DM, Hoogduijn MJ, Hesselink DA, Baan CC. Advanced in vitro Research Models to Study the Role of Endothelial Cells in Solid Organ Transplantation. Front Immunol 2021; 12:607953. [PMID: 33664744 PMCID: PMC7921837 DOI: 10.3389/fimmu.2021.607953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
The endothelium plays a key role in acute and chronic rejection of solid organ transplants. During both processes the endothelium is damaged often with major consequences for organ function. Also, endothelial cells (EC) have antigen-presenting properties and can in this manner initiate and enhance alloreactive immune responses. For decades, knowledge about these roles of EC have been obtained by studying both in vitro and in vivo models. These experimental models poorly imitate the immune response in patients and might explain why the discovery and development of agents that control EC responses is hampered. In recent years, various innovative human 3D in vitro models mimicking in vivo organ structure and function have been developed. These models will extend the knowledge about the diverse roles of EC in allograft rejection and will hopefully lead to discoveries of new targets that are involved in the interactions between the donor organ EC and the recipient's immune system. Moreover, these models can be used to gain a better insight in the mode of action of the currently prescribed immunosuppression and will enhance the development of novel therapeutics aiming to reduce allograft rejection and prolong graft survival.
Collapse
Affiliation(s)
- Daphne M Peelen
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
31
|
Segura-Collar B, Mata-Martínez P, Hernández-Laín A, Sánchez-Gómez P, Gargini R. Blood-Brain Barrier Disruption: A Common Driver of Central Nervous System Diseases. Neuroscientist 2021; 28:222-237. [PMID: 33446074 DOI: 10.1177/1073858420985838] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The brain is endowed with a unique cellular composition and organization, embedded within a vascular network and isolated from the circulating blood by a specialized frontier, the so-called blood-brain barrier (BBB), which is necessary for its proper function. Recent reports have shown that increments in the permeability of the blood vessels facilitates the entry of toxic components and immune cells to the brain parenchyma and alters the phenotype of the supporting astrocytes. All of these might contribute to the progression of different pathologies such as brain cancers or neurodegenerative diseases. Although it is well known that BBB breakdown occurs due to pericyte malfunctioning or to the lack of stability of the blood vessels, its participation in the diverse neural diseases needs further elucidation. This review summarizes what it is known about BBB structure and function and how its instability might trigger or promote neuronal degeneration and glioma progression, with a special focus on the role of pericytes as key modulators of the vasculature. Moreover, we will discuss some recent reports that highlights the participation of the BBB alterations in glioma growth. This pan-disease analysis might shed some light into these otherwise untreatable diseases and help to design better therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - Ricardo Gargini
- Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| |
Collapse
|
32
|
Abstract
Aortic dissection (AD) is a serious condition and a health issue on a global scale. β-Aminopropionitrile–induced AD in mice is similar to the pathogenesis of AD in humans. Resveratrol (RSV) is a natural polyphenolic substance that provides anti-inflammatory and cardiovascular effects, but the role of RSV in AD is unclear. In this study, we investigated the effects and mechanisms of RSV on β-aminopropionitrile–induced AD in mice. Our results indicate that RSV can prevent the occurrence of AD. More meaningfully, we found that the protective effect comprises an increase in sirtuin 1 (SIRT1) expression in endothelial cells for the reconstruction of their structure, reducing the recruitment of inflammatory cells by endothelial cells and inhibiting the inflammation response, thereby suppressing the occurrence of AD.
Collapse
|
33
|
Li C, Wang Y, Yan XL, Guo ZN, Yang Y. Pathological changes in neurovascular units: Lessons from cases of vascular dementia. CNS Neurosci Ther 2021; 27:17-25. [PMID: 33423390 PMCID: PMC7804924 DOI: 10.1111/cns.13572] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VD) is the second leading cause of dementia after Alzheimer's disease (AD). The decrease of cerebral blood flow (CBF) to different degrees is one of the main causes of VD. Neurovascular unit (NVU) is a vessel‐centered concept, emphasizing all the cellular components play an integrated role in maintaining the normal physiological functions of the brain. More and more evidence shows that reduced CBF causes a series of changes in NVU, such as impaired neuronal function, abnormal activation of glial cells, and changes in vascular permeability, all of which collectively play a role in the pathogenesis of VD. In this paper, we review NVU changes as CBF decreases, focusing on each cellular component of NVU. We also highlight remote ischemic preconditioning as a promising approach for VD prevention and treatment from the NVU perspective of view.
Collapse
Affiliation(s)
- Chao Li
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yan Wang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, the First Hospital of Jilin University, Changchun, China.,China National Comprehensive Stroke Center, Changchun, China.,Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
34
|
Vassiliou AG, Kotanidou A, Dimopoulou I, Orfanos SE. Endothelial Damage in Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:ijms21228793. [PMID: 33233715 PMCID: PMC7699909 DOI: 10.3390/ijms21228793] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 01/01/2023] Open
Abstract
The pulmonary endothelium is a metabolically active continuous monolayer of squamous endothelial cells that internally lines blood vessels and mediates key processes involved in lung homoeostasis. Many of these processes are disrupted in acute respiratory distress syndrome (ARDS), which is marked among others by diffuse endothelial injury, intense activation of the coagulation system and increased capillary permeability. Most commonly occurring in the setting of sepsis, ARDS is a devastating illness, associated with increased morbidity and mortality and no effective pharmacological treatment. Endothelial cell damage has an important role in the pathogenesis of ARDS and several biomarkers of endothelial damage have been tested in determining prognosis. By further understanding the endothelial pathobiology, development of endothelial-specific therapeutics might arise. In this review, we will discuss the underlying pathology of endothelial dysfunction leading to ARDS and emerging therapies. Furthermore, we will present a brief overview demonstrating that endotheliopathy is an important feature of hospitalised patients with coronavirus disease-19 (COVID-19).
Collapse
Affiliation(s)
- Alice G. Vassiliou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Stylianos E. Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
- 2nd Department of Critical Care, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, 124 62 Athens, Greece
- Correspondence: or ; Tel.: +30-2107-235-521
| |
Collapse
|
35
|
Pujari A, Smith AF, Hall JD, Mei P, Chau K, Nguyen DT, Sweet DT, Jiménez JM. Lymphatic Valves Separate Lymph Flow Into a Central Stream and a Slow-Moving Peri-Valvular Milieu. J Biomech Eng 2020; 142:100805. [PMID: 32766737 PMCID: PMC7477708 DOI: 10.1115/1.4048028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 07/28/2020] [Indexed: 01/09/2023]
Abstract
The lymphatic system plays a pivotal role in the transport of fats, waste, and immune cells, while also serving as a metastatic route for select cancers. Using live imaging and particle tracking, we experimentally characterized the lymph flow field distal from the inguinal lymph node in the vicinity of normal bileaflet and malformed unileaflet intraluminal valves. Particle tracking experiments demonstrated that intraluminal lymphatic valves concentrate higher velocity lymph flow in the center of the vessel, while generating adjacent perivalvular recirculation zones. The recirculation zones are characterized by extended particle residence times and low wall shear stress (WSS) magnitudes in comparison to the rest of the lymphangion. A malformed unileaflet valve skewed lymph flow toward the endothelium on the vessel wall, generating a stagnation point and a much larger recirculation zone on the opposite wall. These studies define physical consequences of bileaflet and unileaflet intraluminal lymphatic valves that affect lymph transport and the generation of a heterogeneous flow field that affects the lymphatic endothelium nonuniformly. The characterized flow fields were recreated in vitro connecting different flow environments present in the lymphangion to a lymphatic endothelial cell (LEC) pro-inflammatory phenotype. Unique and detailed insight into lymphatic flow is provided, with potential applications to a variety of diseases that affect lymph transport and drug delivery.
Collapse
Affiliation(s)
- Akshay Pujari
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Alexander F. Smith
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Joshua D. Hall
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Patrick Mei
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003
| | - Kin Chau
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Duy T. Nguyen
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA 01003
| | - Daniel T. Sweet
- Department of Medicine and Division of Cardiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Juan M. Jiménez
- Department of Mechanical and Industrial Engineering, University of Massachusetts, N575 Life Sciences Laboratory,240 Thatcher Way Amherst Amherst, MA 01003; Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003
| |
Collapse
|
36
|
Morsink MAJ, Willemen NGA, Leijten J, Bansal R, Shin SR. Immune Organs and Immune Cells on a Chip: An Overview of Biomedical Applications. MICROMACHINES 2020; 11:mi11090849. [PMID: 32932680 PMCID: PMC7570325 DOI: 10.3390/mi11090849] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/27/2022]
Abstract
Understanding the immune system is of great importance for the development of drugs and the design of medical implants. Traditionally, two-dimensional static cultures have been used to investigate the immune system in vitro, while animal models have been used to study the immune system’s function and behavior in vivo. However, these conventional models do not fully emulate the complexity of the human immune system or the human in vivo microenvironment. Consequently, many promising preclinical findings have not been reproduced in human clinical trials. Organ-on-a-chip platforms can provide a solution to bridge this gap by offering human micro-(patho)physiological systems in which the immune system can be studied. This review provides an overview of the existing immune-organs-on-a-chip platforms, with a special emphasis on interorgan communication. In addition, future challenges to develop a comprehensive immune system-on-chip model are discussed.
Collapse
Affiliation(s)
- Margaretha A. J. Morsink
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
- Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Niels G. A. Willemen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Jeroen Leijten
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Ruchi Bansal
- Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Correspondence: ; Tel.: +1-617-768-8320
| |
Collapse
|
37
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
38
|
D’Addio M, Frey J, Otto VI. The manifold roles of sialic acid for the biological functions of endothelial glycoproteins. Glycobiology 2020; 30:490-499. [PMID: 32039454 PMCID: PMC7372927 DOI: 10.1093/glycob/cwaa008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Vascular endothelia are covered with a dense glycocalix that is heavily sialylated. Sialylation of vascular glycoconjugates is involved in the regulation of cell-cell interactions, be it among endothelial cells at cell junctions or between endothelial and blood-borne cells. It also plays important roles in modulating the binding of soluble ligands and the signaling by vascular receptors. Here, we provide an overview over the sialylation-function relationships of glycoproteins expressed in the blood and lymphatic vasculature. We first describe cellular interactions in which sialic acid contributes in a stereospecific manner to glycan epitopes recognized by glycan-binding proteins. Our major focus is however on the rarely discussed examples of vascular glycoproteins whose biological functions are modulated by sialylation through other mechanisms.
Collapse
Affiliation(s)
- Marco D’Addio
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Jasmin Frey
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Vivianne I Otto
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
39
|
Zhang J, Li W, Ying Z, Zhao D, Yi G, Li H, Liu X. Soybean protein-derived peptide nutriment increases negative nitrogen balance in burn injury-induced inflammatory stress response in aged rats through the modulation of white blood cells and immune factors. Food Nutr Res 2020; 64:3677. [PMID: 32694965 PMCID: PMC7346896 DOI: 10.29219/fnr.v64.3677] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND As an important nutrient, soybean protein-derived peptides (SPP) affect the immune function of animals. OBJECTIVE This study describes the effects of nutrient supplementation with SPP on the negative nitrogen balance in the burn injury-induced inflammatory response of aged rats. DESIGN Soybean protein isolate (SPI) was hydrolyzed to obtain SPP. A negative nitrogen-balance aged rat model and a major full-thickness 30% total body surface area (TBSA) burn-injury rat model were utilized. RESULTS The results show that SPP can increase the speed and ability of inflammatory stress by adjusting white blood cell counts. Soybean protein-derived peptides significantly increased serum immunoglobulin M (IgM), immunoglobulin G (IgG) and immunoglobulin A (IgA) levels; significantly decreased serum interleukin-1 beta (IL-β), tumor necrosis factor-alpha (TNF-α) and regulated upon activation normal T-cell expressed and secreted (RANTES) levels. These results give conclusive evidence that SPP has a significantly positive effect in improving the immune function on the condition of negative nitrogen balance with burn-injury, and reducing excessive inflammation. CONCLUSIONS Nutrient supplementation of SPP can, therefore, be used as an adjuvant treatment to inhibit the development and severity of inflammatory reactions caused by burns, providing a novel therapy for the treatment and positive prognosis of burn patients.
Collapse
Affiliation(s)
- Jian Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Wenhui Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Zhiwei Ying
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Di Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Guofu Yi
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - He Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Xinqi Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
40
|
Stubbs EB. Targeting the blood-nerve barrier for the management of immune-mediated peripheral neuropathies. Exp Neurol 2020; 331:113385. [PMID: 32562668 DOI: 10.1016/j.expneurol.2020.113385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
Healthy peripheral nerves encounter, with increased frequency, numerous chemical, biological, and biomechanical forces. Over time and with increasing age, these forces collectively contribute to the pathophysiology of a spectrum of traumatic, metabolic, and/or immune-mediated peripheral nerve disorders. The blood-nerve barrier (BNB) serves as a critical first-line defense against chemical and biologic insults while biomechanical forces are continuously buffered by a dense array of longitudinally orientated epineural collagen fibers exhibiting high-tensile strength. As emphasized throughout this Experimental Neurology Special Issue, the BNB is best characterized as a functionally dynamic multicellular vascular unit comprised of not only highly specialized endoneurial endothelial cells, but also associated perineurial cells, pericytes, Schwann cells, basement membrane, and invested axons. The composition of the BNB, while anatomically distinct, is not functionally dissimilar to that of the well characterized neurovascular unit of the central nervous system. While the BNB lacks a glial limitans and an astrocytic endfoot layer, the primary function of both vascular units is to establish, maintain, and protect an optimal endoneurial (PNS) or interstitial (CNS) fluid microenvironment that is vital for proper neuronal function. Altered endoneurial homeostasis as a secondary consequence of BNB dysregulation is considered an early pathological event in the course of a variety of traumatic, immune-mediated, or metabolically acquired peripheral neuropathies. In this review, emerging experimental advancements targeting the endoneurial microvasculature for the therapeutic management of immune-mediated inflammatory peripheral neuropathies, including the AIDP variant of Guillain-Barré syndrome, are discussed.
Collapse
Affiliation(s)
- Evan B Stubbs
- Research Service (151), Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL 60141, USA; Department of Ophthalmology, Loyola University Health Science Division, Maywood, IL 60153, USA.
| |
Collapse
|
41
|
Morikis VA, Masadeh E, Simon SI. Tensile force transmitted through LFA-1 bonds mechanoregulate neutrophil inflammatory response. J Leukoc Biol 2020; 108:1815-1828. [PMID: 32531836 DOI: 10.1002/jlb.3a0520-100rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
Recruitment of leukocytes to sites of acute inflammation is guided by spatial and temporal cues that ensure appropriate cell numbers infiltrate the tissue at precise locations to protect it from infection and initiate repair. On inflamed endothelium, neutrophil rolling via selectins elicits cytosolic calcium release from endoplasmic reticulum (ER)-stores that are synergistic with chemokine signaling to activate formation of high affinity (HA) LFA-1 bonds to ICAM-1, which is necessary to anchor cells against the drag force of blood flow. Bond tension on LFA-1 within the area of adhesive contact with endothelium elicits calcium entry through calcium release-activated calcium channel protein 1 (Orai-1) membrane channels that in turn activate neutrophil shape change and migration. We hypothesized that mechanotransduction via LFA-1 is mediated by assembly of a cytosolic molecular complex consisting of Kindlin-3, receptor for activated C kinase 1 (RACK1), and Orai1. Initiation of Ca2+ flux at sites of adhesive contact required a threshold level of shear stress and increased with the magnitude of bond tension transduced across as few as 200 HA LFA-1. A sequential mechanism triggered by force acting on LFA-1/Kindlin-3 precipitated dissociation of RACK1, which formed a concentration gradient above LFA-1 bond clusters. This directed translocation of ER proximal to Orai1, where binding of inositol 1,4,5-triphosphate receptor type 1 and activation via stromal interaction molecule 1 elicited Ca flux and subsequent neutrophil shape change and motility. We conclude that neutrophils sense adhesive traction on LFA-1 bonds on a submicron scale to direct calcium influx, thereby ensuring sufficient shear stress of blood flow is present to trigger cell arrest and initiate transmigration at precise regions of vascular inflammation.
Collapse
Affiliation(s)
- Vasilios A Morikis
- Department of Biomedical Engineering, University of California-Davis, California, USA
| | - Eman Masadeh
- Department of Biomedical Engineering, University of California-Davis, California, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California-Davis, California, USA
| |
Collapse
|
42
|
Silva T, Jäger W, Neuss-Radu M, Sequeira A. Modeling of the early stage of atherosclerosis with emphasis on the regulation of the endothelial permeability. J Theor Biol 2020; 496:110229. [PMID: 32259543 DOI: 10.1016/j.jtbi.2020.110229] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 02/25/2020] [Accepted: 02/29/2020] [Indexed: 02/03/2023]
Abstract
In this paper, we develop a mathematical model for the early stage of atherosclerosis, as a chronic inflammatory disease. It includes also processes that are relevant for the "thickening" of the vessel walls, and prepares a more complete model including also the later stages of atherosclerosis. The model consists of partial differential equations: Navier-Stokes equations modeling blood flow, Biot equations modeling the fluid flow inside the poroelastic vessel wall, and convection/chemotaxis-reaction-diffusion equations modeling transport, signaling and interaction processes initiating inflammation and atherosclerosis. The main innovations of this model are: a) quantifying the endothelial permeability to low-density-lipoproteins (LDL) and to the monocytes as a function of WSS, cytokines and LDL on the endothelial surface; b) transport of monocytes on the endothelial surface, mimicking the monocytes adhesion and rolling; c) the monocytes influx in the lumen, as a function of factor increasing monocytopoiesis; d) coupling between Navier-Stokes system, Biot system and convection/chemotaxis-reaction-diffusion equations. Numerical simulations of a simplified model were performed in an idealized two-dimensional geometry in order to investigate the dynamics of endothelial permeability, and the growth and spread of immune cells populations and their dependence in particular on low-density-lipoprotein and wall-shear stress.
Collapse
Affiliation(s)
- Telma Silva
- Mathematics Department and CEMAT, IST, University of Lisbon, Portugal.
| | - Willi Jäger
- IWR, University of Heidelberg, Heidelberg, Germany.
| | - Maria Neuss-Radu
- Mathematics Department, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Adélia Sequeira
- Mathematics Department and CEMAT, IST, University of Lisbon, Portugal.
| |
Collapse
|
43
|
Kuter KZ, Cenci MA, Carta AR. The role of glia in Parkinson's disease: Emerging concepts and therapeutic applications. PROGRESS IN BRAIN RESEARCH 2020; 252:131-168. [PMID: 32247363 DOI: 10.1016/bs.pbr.2020.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Originally believed to primarily affect neurons, Parkinson's disease (PD) has recently been recognized to also affect the functions and integrity of microglia and astroglia, two cell categories of fundamental importance to brain tissue homeostasis, defense, and repair. Both a loss of glial supportive-defensive functions and a toxic gain of glial functions are implicated in the neurodegenerative process. Moreover, the chronic treatment with L-DOPA may cause maladaptive glial plasticity favoring a development of therapy complications. This chapter focuses on the pathophysiology of PD from a glial point of view, presenting this rapidly growing field from the first discoveries made to the most recent developments. We report and compare histopathological and molecular findings from experimental models of PD and human studies. We moreover discuss the important role played by astrocytes in compensatory adaptations taking place during presymptomatic disease stages. We finally describe examples of potential therapeutic applications stemming from an increased understanding of the important roles of glia in PD.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy.
| |
Collapse
|
44
|
Biology of the human blood-nerve barrier in health and disease. Exp Neurol 2020; 328:113272. [PMID: 32142802 DOI: 10.1016/j.expneurol.2020.113272] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
A highly regulated endoneurial microenvironment is required for normal axonal function in peripheral nerves and nerve roots, which structurally consist of an outer collagenous epineurium, inner perineurium consisting of multiple concentric layers of specialized epithelioid myofibroblasts that surround the innermost endoneurium, which consists of myelinated and unmyelinated axons embedded in a looser mesh of collagen fibers. Endoneurial homeostasis is achieved by tight junction-forming endoneurial microvessels that control ion, solute, water, nutrient, macromolecule and leukocyte influx and efflux between the bloodstream and endoneurium, and the innermost layers of the perineurium that control interstitial fluid component flux between the freely permeable epineurium and endoneurium. Strictly speaking, endoneurial microvascular endothelium should be considered the blood-nerve barrier (BNB) due to direct communication with circulating blood. The mammalian BNB is considered the second most restrictive vascular system after the blood-brain barrier (BBB) based on classic in situ permeability studies. Structural alterations in endoneurial microvessels or interactions with hematogenous leukocytes have been described in several human peripheral neuropathies; however major advances in BNB biology in health and disease have been limited over the past 50 years. Guided by transcriptome and proteome studies of normal and pathologic human peripheral nerves, purified primary and immortalized human endoneurial endothelial cells that form the BNB and leukocytes from patients with well-characterized peripheral neuropathies, validated by in situ or ex vivo protein expression studies, data are emerging on the molecular and functional characteristics of the human BNB in health and in specific peripheral neuropathies, as well as chronic neuropathic pain. These early advancements have the potential to not only increase our understanding of how the BNB works and adapts or fails to adapt to varying insult, but provide insights relevant to pathogenic leukocyte trafficking, with translational potential and specific therapeutic application for chronic peripheral neuropathies and neuropathic pain.
Collapse
|
45
|
Delgado-Bellido D, Bueno-Galera C, López-Jiménez L, Garcia-Diaz A, Oliver FJ. Endothelial Phosphatase VE-PTP Participates in Vasculogenic Mimicry by Preventing Autophagic Degradation of VE-Cadherin. Front Oncol 2020; 10:18. [PMID: 32117706 PMCID: PMC7025541 DOI: 10.3389/fonc.2020.00018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/07/2020] [Indexed: 11/13/2022] Open
Abstract
Aberrant extra-vascular expression of VE-cadherin has been observed in metastasis associated with Vasculogenic Mimicry (VM); we have recently shown that in VM prone cells VE-cadherin is mainly in the form of phospho-VE-cadherin in Y658 allowing increased plasticity that potentiates VM development in malignant cells. In the current study, we present results to show that human malignant melanoma cells VM+, express the VE-cadherin phosphatase VE-PTP. VE-PTP forms a complex with VE-Cadherin and p120-catenin and the presence of this complex act as a safeguard to prevent VE-Cadherin protein degradation by autophagy. Indeed, VE-PTP silencing results in complete degradation of VE-cadherin with the features of autophagy. In summary, this study shows that VE-PTP is involved in VM formation and disruption of VE-PTP/VE-Cadherin/p120 complex results in enhanced autophagy in aggressive VM+ cells. Thus, we identify VE-PTP as a key player in VM development by regulating VE-cadherin protein degradation through autophagy.
Collapse
Affiliation(s)
| | | | - Laura López-Jiménez
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, CIBERONC, Granada, Spain
| | - Angel Garcia-Diaz
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, CIBERONC, Granada, Spain
| | - F Javier Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, CIBERONC, Granada, Spain
| |
Collapse
|
46
|
Gui F, You Z, Fu S, Wu H, Zhang Y. Endothelial Dysfunction in Diabetic Retinopathy. Front Endocrinol (Lausanne) 2020; 11:591. [PMID: 33013692 PMCID: PMC7499433 DOI: 10.3389/fendo.2020.00591] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
Diabetic retinopathy (DR) is a diabetic complication which affects retinal function and results in severe loss of vision and relevant retinal diseases. Retinal vascular dysfunction caused by multifactors, such as advanced glycosylation end products and receptors, pro-inflammatory cytokines and chemokines, proliferator-activated receptor-γ disruption, growth factors, oxidative stress, and microRNA. These factors promote retinal endothelial dysfunction, which results in the development of DR. In this review, we summarize the contributors in the pathophysiology of DR for a better understanding of the molecular and cellular mechanism in the development of DR with a special emphasis on retinal endothelial dysfunction.
Collapse
|
47
|
Intermittent rolling is a defect of the extravasation cascade caused by Myosin1e-deficiency in neutrophils. Proc Natl Acad Sci U S A 2019; 116:26752-26758. [PMID: 31811025 DOI: 10.1073/pnas.1902502116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neutrophil extravasation is a migratory event in response to inflammation that depends on cytoskeletal dynamics regulated by myosins. Myosin-1e (Myo1e) is a long-tailed class-I myosin that has not yet been studied in the context of neutrophil-endothelial interactions and neutrophil extravasation. Intravital microscopy of TNFα-inflamed cremaster muscles in Myo1e-deficient mice revealed that Myo1e is required for efficient neutrophil extravasation. Specifically, Myo1e deficiency caused increased rolling velocity, decreased firm adhesion, aberrant crawling, and strongly reduced transmigration. Interestingly, we observed a striking discontinuous rolling behavior termed "intermittent rolling," during which Myo1e-deficient neutrophils showed alternating rolling and jumping movements. Surprisingly, chimeric mice revealed that these effects were due to Myo1e deficiency in leukocytes. Vascular permeability was not significantly altered in Myo1e KO mice. Myo1e-deficient neutrophils showed diminished arrest, spreading, uropod formation, and chemotaxis due to defective actin polymerization and integrin activation. In conclusion, Myo1e critically regulates adhesive interactions of neutrophils with the vascular endothelium and neutrophil extravasation. Myo1e may therefore be an interesting target in chronic inflammatory diseases characterized by excessive neutrophil recruitment.
Collapse
|
48
|
Simula L, Pacella I, Colamatteo A, Procaccini C, Cancila V, Bordi M, Tregnago C, Corrado M, Pigazzi M, Barnaba V, Tripodo C, Matarese G, Piconese S, Campello S. Drp1 Controls Effective T Cell Immune-Surveillance by Regulating T Cell Migration, Proliferation, and cMyc-Dependent Metabolic Reprogramming. Cell Rep 2019; 25:3059-3073.e10. [PMID: 30540939 PMCID: PMC6302735 DOI: 10.1016/j.celrep.2018.11.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 08/01/2018] [Accepted: 11/01/2018] [Indexed: 01/09/2023] Open
Abstract
Mitochondria are key players in the regulation of T cell biology by dynamically responding to cell needs, but how these dynamics integrate in T cells is still poorly understood. We show here that the mitochondrial pro-fission protein Drp1 fosters migration and expansion of developing thymocytes both in vitro and in vivo. In addition, we find that Drp1 sustains in vitro clonal expansion and cMyc-dependent metabolic reprogramming upon activation, also regulating effector T cell numbers in vivo. Migration and extravasation defects are also exhibited in Drp1-deficient mature T cells, unveiling its crucial role in controlling both T cell recirculation in secondary lymphoid organs and accumulation at tumor sites. Moreover, the observed Drp1-dependent imbalance toward a memory-like phenotype favors T cell exhaustion in the tumor microenvironment. All of these findings support a crucial role for Drp1 in several processes during T cell development and in anti-tumor immune-surveillance. The pro-fission protein Drp1 sustains correct thymocyte maturation Drp1 promotes T cell metabolic reprogramming and expansion upon activation Drp1 allows efficient T cell extravasation from blood and infiltration into tumors An optimal T cell anti-tumor response requires Drp1
Collapse
Affiliation(s)
- Luca Simula
- Department of Biology, University of Rome Tor Vergata, Rome, Italy; IRCCS, Fondazione Santa Lucia, Rome, Italy
| | - Ilenia Pacella
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessandra Colamatteo
- Department of Molecular Medicine and Biotechnologies, University of Naples "Federico II," Naples, Italy
| | - Claudio Procaccini
- IRCCS, Fondazione Santa Lucia, Rome, Italy; Institute of Experimental Oncology and Endocrinology, National Research Council (IEOS-CNR), Naples, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo School of Medicine, Italy
| | - Matteo Bordi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Tregnago
- Department of Women and Child Health, Haematology-Oncology Clinic and Lab, University of Padova, Padova, Italy
| | - Mauro Corrado
- Max Planck Institute of Immunology and Epigenetics, Freiburg im Breisgau, Germany
| | - Martina Pigazzi
- Department of Women and Child Health, Haematology-Oncology Clinic and Lab, University of Padova, Padova, Italy
| | - Vincenzo Barnaba
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo School of Medicine, Italy
| | - Giuseppe Matarese
- Department of Molecular Medicine and Biotechnologies, University of Naples "Federico II," Naples, Italy; Institute of Experimental Oncology and Endocrinology, National Research Council (IEOS-CNR), Naples, Italy
| | - Silvia Piconese
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Silvia Campello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy; IRCCS, Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
49
|
Chiang HY, Chu PH, Lee TH. MFG-E8 mediates arterial aging by promoting the proinflammatory phenotype of vascular smooth muscle cells. J Biomed Sci 2019; 26:61. [PMID: 31470852 PMCID: PMC6716880 DOI: 10.1186/s12929-019-0559-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 08/22/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Among older adults, arterial aging is the major factor contributing to increased risk for cardiovascular disease-related morbidity and mortality. The chronic vascular inflammation that accompanies aging causes diffuse intimal-medial thickening of the arterial wall, thus increasing the vulnerability of aged vessels to vascular insults. Milk fat globule-epidermal growth factor 8 (MFG-E8) is a biomarker for aging arteries. This integrin-binding glycoprotein, induced by angiotensin II, facilitates vascular smooth muscle cell (VSMC) proliferation and invasion in aging vasculatures. This study investigated whether MFG-E8 directly mediates the initial inflammatory responses in aged arteries or VSMCs. METHODS A model of neointimal hyperplasia was induced in the common carotid artery (CCA) of aged mice to exacerbate age-associated vascular remodeling. Recombinant MFG-E8 (rMFG-E8) was administered to the injured artery using Pluronic gel to accentuate the effect on age-related vascular pathophysiology. The MFG-E8 level, leukocyte infiltration, and proinflammatory cell adhesion molecule (CAM) expression in the arterial wall were evaluated through immunohistochemistry. By using immunofluorescence and immunoblotting, the activation of the critical proinflammatory transcription factor nuclear factor (NF)-κB in the injured CCAs was analyzed. Immunofluorescence, immunoblotting, and quantitative real-time polymerase chain reaction were conducted using VSMCs isolated from the aortas of young and aged mice to assess NF-κB nuclear translocation, NF-κB-dependent gene expression, and cell proliferation. The extent of intimal-medial thickening in the injured vessels was analyzed morphometrically. Finally, Transwell migration assay was used to examine VSMC migration. RESULTS Endogenous MFG-E8 expression in aged CCAs was significantly induced by ligation injury. Aged CCAs treated with rMFG-E8 exhibited increased leukocyte extravasation, CAM expression, and considerably increased NF-κB activation induced by rMFG-E8 in the ligated vessels. Exposure of early passage VSMCs from aged aortas to rMFG-E8 substantially increased NF-κB activation, proinflammatory gene expression, and cell proliferation. However, rMFG-E8 attenuated VSMC migration. CONCLUSIONS MFG-E8 promoted the proinflammatory phenotypic shift of aged VSMCs and arteries, rendering the vasculature prone to vascular diseases. MFG-E8 may constitute a novel therapeutic target for retarding the aging processes in such vessels.
Collapse
Affiliation(s)
- Hou-Yu Chiang
- Department of Anatomy, College of Medicine, Chang Gung University, 259 Wenhua 1st Rd., Guishan Dist, Taoyuan City, 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Hein Lee
- Department of Anatomy, College of Medicine, Chang Gung University, 259 Wenhua 1st Rd., Guishan Dist, Taoyuan City, 33302, Taiwan. .,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
50
|
Singh M, Benencia F. Inflammatory processes in obesity: focus on endothelial dysfunction and the role of adipokines as inflammatory mediators. Int Rev Immunol 2019; 38:157-171. [DOI: 10.1080/08830185.2019.1638921] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Manindra Singh
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Fabian Benencia
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University, Athens, OH, USA
| |
Collapse
|