1
|
Alnafisah KH, Ranjan A, Sahu SP, Chen J, Alhejji SM, Noël A, Gartia MR, Mukhopadhyay S. Machine learning for automated classification of lung collagen in a urethane-induced lung injury mouse model. BIOMEDICAL OPTICS EXPRESS 2024; 15:5980-5998. [PMID: 39421774 PMCID: PMC11482176 DOI: 10.1364/boe.527972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 10/19/2024]
Abstract
Dysregulation of lung tissue collagen level plays a vital role in understanding how lung diseases progress. However, traditional scoring methods rely on manual histopathological examination introducing subjectivity and inconsistency into the assessment process. These methods are further hampered by inter-observer variability, lack of quantification, and their time-consuming nature. To mitigate these drawbacks, we propose a machine learning-driven framework for automated scoring of lung collagen content. Our study begins with the collection of a lung slide image dataset from adult female mice using second harmonic generation (SHG) microscopy. In our proposed approach, first, we manually extracted features based on the 46 statistical parameters of fibrillar collagen. Subsequently, we pre-processed the images and utilized a pre-trained VGG16 model to uncover hidden features from pre-processed images. We then combined both image and statistical features to train various machine learning and deep neural network models for classification tasks. We employed advanced unsupervised techniques like K-means, principal component analysis (PCA), t-distributed stochastic neighbour embedding (t-SNE), and uniform manifold approximation and projection (UMAP) to conduct thorough image analysis for lung collagen content. Also, the evaluation of the trained models using the collagen data includes both binary and multi-label classification to predict lung cancer in a urethane-induced mouse model. Experimental validation of our proposed approach demonstrates promising results. We obtained an average accuracy of 83% and an area under the receiver operating characteristic curve (ROC AUC) values of 0.96 through the use of a support vector machine (SVM) model for binary categorization tasks. For multi-label classification tasks, to quantify the structural alteration of collagen, we attained an average accuracy of 73% and ROC AUC values of 1.0, 0.38, 0.95, and 0.86 for control, baseline, treatment_1, and treatment_2 groups, respectively. Our findings provide significant potential for enhancing diagnostic accuracy, understanding disease mechanisms, and improving clinical practice using machine learning and deep learning models.
Collapse
Affiliation(s)
| | - Amit Ranjan
- Center for Computation & Technology and Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Sushant P Sahu
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
- Amity Institute of Biotechnology and Applied Sciences, Amity University, Mumbai, Maharashtra-410206, India
| | - Jianhua Chen
- Department of Computer Science, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | - Alexandra Noël
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Supratik Mukhopadhyay
- Center for Computation & Technology and Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
2
|
Cleary SJ, Qiu L, Seo Y, Baluk P, Liu D, Serwas NK, Cyster JG, McDonald DM, Krummel MF, Looney MR. Intravital imaging of pulmonary lymphatics in inflammation and metastatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612619. [PMID: 39345499 PMCID: PMC11430110 DOI: 10.1101/2024.09.12.612619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Intravital microscopy has enabled the study of immune dynamics in the pulmonary microvasculature, but many key events remain unseen because they occur in deeper lung regions. We therefore developed a technique for stabilized intravital imaging of bronchovascular cuffs and collecting lymphatics surrounding pulmonary veins in mice. Intravital imaging of pulmonary lymphatics revealed ventilation-dependence of steady-state lung lymph flow and ventilation-independent lymph flow during inflammation. We imaged the rapid exodus of migratory dendritic cells through lung lymphatics following inflammation and measured effects of pharmacologic and genetic interventions targeting chemokine signaling. Intravital imaging also captured lymphatic immune surveillance of lung-metastatic cancers and lymphatic metastasis of cancer cells. To our knowledge, this is the first imaging of lymph flow and leukocyte migration through intact pulmonary lymphatics. This approach will enable studies of protective and maladaptive processes unfolding within the lungs and in other previously inaccessible locations.
Collapse
Affiliation(s)
- Simon J. Cleary
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
- Institute of Pharmaceutical Science, King’s College London, London, UK
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Yurim Seo
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Peter Baluk
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, CA, USA
| | - Dan Liu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, UCSF, CA, USA
- Westlake Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou, Zhejiang, China
| | | | - Jason G. Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, UCSF, CA, USA
- Bakar ImmunoX Initiative, UCSF, CA, USA
| | - Donald M. McDonald
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, CA, USA
| | - Matthew F. Krummel
- Department of Pathology, UCSF, CA, USA
- Bakar ImmunoX Initiative, UCSF, CA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
- Bakar ImmunoX Initiative, UCSF, CA, USA
| |
Collapse
|
3
|
Xu C, Nedergaard M, Fowell DJ, Friedl P, Ji N. Multiphoton fluorescence microscopy for in vivo imaging. Cell 2024; 187:4458-4487. [PMID: 39178829 PMCID: PMC11373887 DOI: 10.1016/j.cell.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/26/2024]
Abstract
Multiphoton fluorescence microscopy (MPFM) has been a game-changer for optical imaging, particularly for studying biological tissues deep within living organisms. MPFM overcomes the strong scattering of light in heterogeneous tissue by utilizing nonlinear excitation that confines fluorescence emission mostly to the microscope focal volume. This enables high-resolution imaging deep within intact tissue and has opened new avenues for structural and functional studies. MPFM has found widespread applications and has led to numerous scientific discoveries and insights into complex biological processes. Today, MPFM is an indispensable tool in many research communities. Its versatility and effectiveness make it a go-to technique for researchers investigating biological phenomena at the cellular and subcellular levels in their native environments. In this Review, the principles, implementations, capabilities, and limitations of MPFM are presented. Three application areas of MPFM, neuroscience, cancer biology, and immunology, are reviewed in detail and serve as examples for applying MPFM to biological research.
Collapse
Affiliation(s)
- Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 3B, 2200 Copenhagen, Denmark; University of Rochester Medical School, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Deborah J Fowell
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Peter Friedl
- Department of Medical BioSciences, Radboud University Medical Centre, Geert Grooteplein 26-28, Nijmegen HB 6500, the Netherlands
| | - Na Ji
- Department of Neuroscience, Department of Physics, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
4
|
van de Wall S, Anthony SM, Hancox LS, Pewe LL, Langlois RA, Zehn D, Badovinac VP, Harty JT. Dynamic landscapes and protective immunity coordinated by influenza-specific lung-resident memory CD8 + T cells revealed by intravital imaging. Immunity 2024; 57:1878-1892.e5. [PMID: 39043185 DOI: 10.1016/j.immuni.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 04/09/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
Lung-tissue-resident memory (TRM) CD8+ T cells are critical for heterosubtypic immunity against influenza virus (IAV) reinfection. How TRM cells surveil the lung, respond to infection, and interact with other cells remains unresolved. Here, we used IAV infection of mice in combination with intravital and static imaging to define the spatiotemporal dynamics of lung TRM cells before and after recall infection. CD69+CD103+ TRM cells preferentially localized to lung sites of prior IAV infection, where they exhibited patrolling behavior. After rechallenge, lung TRM cells formed tight clusters in an antigen-dependent manner. Transcriptomic analysis of IAV-specific TRM cells revealed the expression of several factors that regulate myeloid cell biology. In vivo rechallenge experiments demonstrated that protection elicited by TRM cells is orchestrated in part by interferon (IFN)-γ-mediated recruitment of inflammatory monocytes into the lungs. Overall, these data illustrate the dynamic landscapes of CD103+ lung TRM cells that mediate early protective immunity against IAV infection.
Collapse
Affiliation(s)
- Stephanie van de Wall
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Scott M Anthony
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lisa S Hancox
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lecia L Pewe
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ryan A Langlois
- University of Minnesota, Department of Microbiology and Immunology and the Center for Immunology, Minneapolis, MN, USA
| | - Dietmar Zehn
- TUM Center for Infection Prevention (ZIP) and Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Vladimir P Badovinac
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - John T Harty
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
5
|
LeBourdais R, Grifno GN, Banerji R, Regan K, Suki B, Nia HT. Mapping the strain-stiffening behavior of the lung and lung cancer at microscale resolution using the crystal ribcage. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1396593. [PMID: 39050550 PMCID: PMC11266057 DOI: 10.3389/fnetp.2024.1396593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/10/2024] [Indexed: 07/27/2024]
Abstract
Lung diseases such as cancer substantially alter the mechanical properties of the organ with direct impact on the development, progression, diagnosis, and treatment response of diseases. Despite significant interest in the lung's material properties, measuring the stiffness of intact lungs at sub-alveolar resolution has not been possible. Recently, we developed the crystal ribcage to image functioning lungs at optical resolution while controlling physiological parameters such as air pressure. Here, we introduce a data-driven, multiscale network model that takes images of the lung at different distending pressures, acquired via the crystal ribcage, and produces corresponding absolute stiffness maps. Following validation, we report absolute stiffness maps of the functioning lung at microscale resolution in health and disease. For representative images of a healthy lung and a lung with primary cancer, we find that while the lung exhibits significant stiffness heterogeneity at the microscale, primary tumors introduce even greater heterogeneity into the lung's microenvironment. Additionally, we observe that while the healthy alveoli exhibit strain-stiffening of ∼1.75 times, the tumor's stiffness increases by a factor of six across the range of measured transpulmonary pressures. While the tumor stiffness is 1.4 times the lung stiffness at a transpulmonary pressure of three cmH2O, the tumor's mean stiffness is nearly five times greater than that of the surrounding tissue at a transpulmonary pressure of 18 cmH2O. Finally, we report that the variance in both strain and stiffness increases with transpulmonary pressure in both the healthy and cancerous lungs. Our new method allows quantitative assessment of disease-induced stiffness changes in the alveoli with implications for mechanotransduction.
Collapse
Affiliation(s)
| | | | | | | | | | - Hadi T. Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
6
|
Schmitt P, Duval A, Camus M, Lefrançais E, Roga S, Dedieu C, Ortega N, Bellard E, Mirey E, Mouton-Barbosa E, Burlet-Schiltz O, Gonzalez-de-Peredo A, Cayrol C, Girard JP. TL1A is an epithelial alarmin that cooperates with IL-33 for initiation of allergic airway inflammation. J Exp Med 2024; 221:e20231236. [PMID: 38597952 PMCID: PMC11010340 DOI: 10.1084/jem.20231236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Epithelium-derived cytokines or alarmins, such as interleukin-33 (IL-33) and thymic stromal lymphopoietin (TSLP), are major players in type 2 immunity and asthma. Here, we demonstrate that TNF-like ligand 1A (TL1A) is an epithelial alarmin, constitutively expressed in alveolar epithelium at steady state in both mice and humans, which cooperates with IL-33 for early induction of IL-9high ILC2s during the initiation of allergic airway inflammation. Upon synergistic activation by IL-33 and TL1A, lung ILC2s acquire a transient IL-9highGATA3low "ILC9" phenotype and produce prodigious amounts of IL-9. A combination of large-scale proteomic analyses, lung intravital microscopy, and adoptive transfer of ILC9 cells revealed that high IL-9 expression distinguishes a multicytokine-producing state-of-activated ILC2s with an increased capacity to initiate IL-5-dependent allergic airway inflammation. Similar to IL-33 and TSLP, TL1A is expressed in airway basal cells in healthy and asthmatic human lungs. Together, these results indicate that TL1A is an epithelium-derived cytokine and an important cofactor of IL-33 in the airways.
Collapse
Affiliation(s)
- Pauline Schmitt
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Anais Duval
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Mylène Camus
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emma Lefrançais
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Stéphane Roga
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Cécile Dedieu
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Nathalie Ortega
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Elisabeth Bellard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emilie Mirey
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Anne Gonzalez-de-Peredo
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Corinne Cayrol
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
7
|
Wang C, Hezam K, Fu E, Pan K, Liu Y, Li Z. In vivo tracking of mesenchymal stem cell dynamics and therapeutics in LPS-induced acute lung injury models. Exp Cell Res 2024; 437:114013. [PMID: 38555014 DOI: 10.1016/j.yexcr.2024.114013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Mesenchymal stem cells (MSCs) have been widely used to treat various inflammatory and immune-related diseases in preclinical and clinical settings. Intravital microscopy (IVM) is considered the gold standard for investigating pathophysiological conditions in living animals. However, the potential for real-time monitoring of MSCs in the pulmonary microenvironment remains underexplored. In this study, we first constructed a lung window and captured changes in the lung at the cellular level under both inflammatory and noninflammatory conditions with a microscope. We further investigated the dynamics and effects of MSCs under two different conditions. Meanwhile, we assessed the alterations in the adhesive capacity of vascular endothelial cells in vitro to investigate the underlying mechanisms of MSC retention in an inflammatory environment. This study emphasizes the importance of the "lung window" for live imaging of the cellular behavior of MSCs by vein injection. Moreover, our results revealed that the upregulation of vascular cell adhesion molecule 1 (VCAM1) in endothelial cells post-inflammatory injury could enhance MSC retention in the lung, further ameliorating acute lung injury. In summary, intravital microscopy imaging provides a practical method to investigate the therapeutic effects of MSCs in acute lung injury.
Collapse
Affiliation(s)
- Chen Wang
- Nankai University School of Medicine, Tianjin 300071, China; Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin 300052, China; The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, College of Life Sciences, Tianjin 300071, China; Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou 450016, China
| | - Kamal Hezam
- Nankai University School of Medicine, Tianjin 300071, China
| | - Enze Fu
- Nankai University School of Medicine, Tianjin 300071, China
| | - Kai Pan
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Yue Liu
- Nankai University School of Medicine, Tianjin 300071, China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin 300071, China; Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin 300052, China; The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, College of Life Sciences, Tianjin 300071, China; Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou 450016, China.
| |
Collapse
|
8
|
Chioccioli M, Liu S, Magruder S, Tata A, Borriello L, McDonough JE, Konkimalla A, Kim SH, Nouws J, Gonzalez DG, Traub B, Ye X, Yang T, Entenberg DR, Krishnaswamy S, Hendry CE, Kaminski N, Tata PR, Sauler M. Stem cell migration drives lung repair in living mice. Dev Cell 2024; 59:830-840.e4. [PMID: 38377991 PMCID: PMC11003834 DOI: 10.1016/j.devcel.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/12/2023] [Accepted: 02/06/2024] [Indexed: 02/22/2024]
Abstract
Tissue repair requires a highly coordinated cellular response to injury. In the lung, alveolar type 2 cells (AT2s) act as stem cells to replenish both themselves and alveolar type 1 cells (AT1s); however, the complex orchestration of stem cell activity after injury is poorly understood. Here, we establish longitudinal imaging of AT2s in murine intact tissues ex vivo and in vivo in order to track their dynamic behavior over time. We discover that a large fraction of AT2s become motile following injury and provide direct evidence for their migration between alveolar units. High-resolution morphokinetic mapping of AT2s further uncovers the emergence of distinct motile phenotypes. Inhibition of AT2 migration via genetic depletion of ArpC3 leads to impaired regeneration of AT2s and AT1s in vivo. Together, our results establish a requirement for stem cell migration between alveolar units and identify properties of stem cell motility at high cellular resolution.
Collapse
Affiliation(s)
- Maurizio Chioccioli
- Department of Genetics and Comparative Medicine, Yale University, New Haven, CT 06519, USA; Department of Comparative Medicine, Yale University, New Haven, CT 06519, USA.
| | - Shuyu Liu
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sumner Magruder
- Department of Computer Science, Yale University, New Haven, CT 06511, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lucia Borriello
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Fox Chase Cancer, Philadelphia, PA 19140, USA
| | - John E McDonough
- Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Arvind Konkimalla
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA; Medical Scientist Training Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sang-Hun Kim
- Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jessica Nouws
- Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - David G Gonzalez
- Department of Genetics and Comparative Medicine, Yale University, New Haven, CT 06519, USA
| | - Brian Traub
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - Xianjun Ye
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - Tao Yang
- Section of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - David R Entenberg
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - Smita Krishnaswamy
- Department of Genetics and Comparative Medicine, Yale University, New Haven, CT 06519, USA; Department of Computer Science, Yale University, New Haven, CT 06511, USA
| | - Caroline E Hendry
- Department of Genetics and Comparative Medicine, Yale University, New Haven, CT 06519, USA
| | - Naftali Kaminski
- Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Maor Sauler
- Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
9
|
Gu Y, Bartolomé-Casado R, Xu C, Bertocchi A, Janney A, Heuberger C, Pearson CF, Teichmann SA, Thornton EE, Powrie F. Immune microniches shape intestinal T reg function. Nature 2024; 628:854-862. [PMID: 38570678 PMCID: PMC11041794 DOI: 10.1038/s41586-024-07251-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
The intestinal immune system is highly adapted to maintaining tolerance to the commensal microbiota and self-antigens while defending against invading pathogens1,2. Recognizing how the diverse network of local cells establish homeostasis and maintains it in the complex immune environment of the gut is critical to understanding how tolerance can be re-established following dysfunction, such as in inflammatory disorders. Although cell and molecular interactions that control T regulatory (Treg) cell development and function have been identified3,4, less is known about the cellular neighbourhoods and spatial compartmentalization that shapes microorganism-reactive Treg cell function. Here we used in vivo live imaging, photo-activation-guided single-cell RNA sequencing5-7 and spatial transcriptomics to follow the natural history of T cells that are reactive towards Helicobacter hepaticus through space and time in the settings of tolerance and inflammation. Although antigen stimulation can occur anywhere in the tissue, the lamina propria-but not embedded lymphoid aggregates-is the key microniche that supports effector Treg (eTreg) cell function. eTreg cells are stable once their niche is established; however, unleashing inflammation breaks down compartmentalization, leading to dominance of CD103+SIRPα+ dendritic cells in the lamina propria. We identify and validate the putative tolerogenic interaction between CD206+ macrophages and eTreg cells in the lamina propria and identify receptor-ligand pairs that are likely to govern the interaction. Our results reveal a spatial mechanism of tolerance in the lamina propria and demonstrate how knowledge of local interactions may contribute to the next generation of tolerance-inducing therapies.
Collapse
Affiliation(s)
- Yisu Gu
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Raquel Bartolomé-Casado
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Alice Bertocchi
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Alina Janney
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Cornelia Heuberger
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
- Roche Innovation Center Zurich, Pharma Research and Early Development, F. Hoffmann-La Roche, Schlieren, Switzerland
| | - Claire F Pearson
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Theory of Condensed Matter, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Emily E Thornton
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK.
- MRC Translational Immune Discovery Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Rizo-Téllez SA, Filep JG. Beyond host defense and tissue injury: the emerging role of neutrophils in tissue repair. Am J Physiol Cell Physiol 2024; 326:C661-C683. [PMID: 38189129 PMCID: PMC11193466 DOI: 10.1152/ajpcell.00652.2023] [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: 11/29/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
Neutrophils, the most abundant immune cells in human blood, play a fundamental role in host defense against invading pathogens and tissue injury. Neutrophils carry potentially lethal weaponry to the affected site. Inadvertent and perpetual neutrophil activation could lead to nonresolving inflammation and tissue damage, a unifying mechanism of many common diseases. The prevailing view emphasizes the dichotomy of their function, host defense versus tissue damage. However, tissue injury may also persist during neutropenia, which is associated with disease severity and poor outcome. Numerous studies highlight neutrophil phenotypic heterogeneity and functional versatility, indicating that neutrophils play more complex roles than previously thought. Emerging evidence indicates that neutrophils actively orchestrate resolution of inflammation and tissue repair and facilitate return to homeostasis. Thus, neutrophils mobilize multiple mechanisms to limit the inflammatory reaction, assure debris removal, matrix remodeling, cytokine scavenging, macrophage reprogramming, and angiogenesis. In this review, we will summarize the homeostatic and tissue-reparative functions and mechanisms of neutrophils across organs. We will also discuss how the healing power of neutrophils might be harnessed to develop novel resolution and repair-promoting therapies while maintaining their defense functions.
Collapse
Affiliation(s)
- Salma A Rizo-Téllez
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Uderhardt S, Neag G, Germain RN. Dynamic Multiplex Tissue Imaging in Inflammation Research. ANNUAL REVIEW OF PATHOLOGY 2024; 19:43-67. [PMID: 37722698 DOI: 10.1146/annurev-pathmechdis-070323-124158] [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: 09/20/2023]
Abstract
Inflammation is a highly dynamic process with immune cells that continuously interact with each other and parenchymal components as they migrate through tissue. The dynamic cellular responses and interaction patterns are a function of the complex tissue environment that cannot be fully reconstructed ex vivo, making it necessary to assess cell dynamics and changing spatial patterning in vivo. These dynamics often play out deep within tissues, requiring the optical focus to be placed far below the surface of an opaque organ. With the emergence of commercially available two-photon excitation lasers that can be combined with existing imaging systems, new avenues for imaging deep tissues over long periods of time have become available. We discuss a selected subset of studies illustrating how two-photon microscopy (2PM) has helped to relate the dynamics of immune cells to their in situ function and to understand the molecular patterns that govern their behavior in vivo. We also review some key practical aspects of 2PM methods and point out issues that can confound the results, so that readers can better evaluate the reliability of conclusions drawn using this technology.
Collapse
Affiliation(s)
- Stefan Uderhardt
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Competence Centre, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Georgiana Neag
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Competence Centre, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Center for Advanced Tissue Imaging (CAT-I), National Institute of Allergy and Infectious Diseases and National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
12
|
Mukhopadhyay A, Tsukasaki Y, Chan WC, Le JP, Kwok ML, Zhou J, Natarajan V, Mostafazadeh N, Maienschein-Cline M, Papautsky I, Tiruppathi C, Peng Z, Rehman J, Ganesh B, Komarova Y, Malik AB. trans-Endothelial neutrophil migration activates bactericidal function via Piezo1 mechanosensing. Immunity 2024; 57:52-67.e10. [PMID: 38091995 PMCID: PMC10872880 DOI: 10.1016/j.immuni.2023.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/02/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The regulation of polymorphonuclear leukocyte (PMN) function by mechanical forces encountered during their migration across restrictive endothelial cell junctions is not well understood. Using genetic, imaging, microfluidic, and in vivo approaches, we demonstrated that the mechanosensor Piezo1 in PMN plasmalemma induced spike-like Ca2+ signals during trans-endothelial migration. Mechanosensing increased the bactericidal function of PMN entering tissue. Mice in which Piezo1 in PMNs was genetically deleted were defective in clearing bacteria, and their lungs were predisposed to severe infection. Adoptive transfer of Piezo1-activated PMNs into the lungs of Pseudomonas aeruginosa-infected mice or exposing PMNs to defined mechanical forces in microfluidic systems improved bacterial clearance phenotype of PMNs. Piezo1 transduced the mechanical signals activated during transmigration to upregulate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4, crucial for the increased PMN bactericidal activity. Thus, Piezo1 mechanosensing of increased PMN tension, while traversing the narrow endothelial adherens junctions, is a central mechanism activating the host-defense function of transmigrating PMNs.
Collapse
Affiliation(s)
- Amitabha Mukhopadhyay
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Wan Ching Chan
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jonathan P Le
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Man Long Kwok
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jian Zhou
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA; Department of Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Nima Mostafazadeh
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Ian Papautsky
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Chinnaswamy Tiruppathi
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Zhangli Peng
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Balaji Ganesh
- Flow Cytometry Core, Research Resources Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yulia Komarova
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
13
|
Kwak A, Thanabalasuriar A. Intravital Microscopy for Imaging and Live Cell Tracking of Alveolar Macrophages in Real Time. Methods Mol Biol 2024; 2813:189-204. [PMID: 38888779 DOI: 10.1007/978-1-0716-3890-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Classic in vitro coculture assays of pathogens with host cells have contributed significantly to our understanding of the intracellular lifestyle of several pathogens. Coculture assays with pathogens and eukaryotic cells can be analyzed through various techniques including plating for colony-forming units (CFU), confocal microscopy, and flow cytometry. However, findings from in vitro assays require validation in an in vivo model. Several physiological conditions can influence host-pathogen interactions, which cannot easily be mimicked in vitro. Intravital microscopy (IVM) is emerging as a powerful tool for studying host-pathogen interactions by enabling in vivo imaging of living organisms. As a result, IVM has significantly enhanced the understanding of infection mediated by diverse pathogens. The versatility of IVM has also allowed for the imaging of various organs as sites of local infection. This chapter specifically focuses on IVM conducted on the lung for elucidating pulmonary immune response, primarily involving alveolar macrophages, to pathogens. Additionally, in this chapter we outline the protocol for lung IVM that utilizes a thoracic suction window to stabilize the lung for acquiring stable images.
Collapse
Affiliation(s)
- Ashley Kwak
- School of Biomedical Sciences Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Ajitha Thanabalasuriar
- School of Biomedical Sciences Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
14
|
Ahn S, Yoon JY, Kim P. Intravital imaging of cardiac tissue utilizing tissue-stabilized heart window chamber in live animal model. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae062. [PMID: 39224098 PMCID: PMC11367956 DOI: 10.1093/ehjimp/qyae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/07/2024] [Indexed: 09/04/2024]
Abstract
Aims To develop and validate an optimized intravital heart microimaging protocol using a suction-based tissue motion-stabilizing cardiac imaging window to facilitate real-time observation of dynamic cellular behaviours within cardiac tissue in live mouse models. Methods and results Intravital heart imaging was conducted using dual-mode confocal and two-photon microscopy. Mice were anesthetized, intubated, and maintained at a stable body temperature during the procedure. LysM-eGFP transgenic mice were utilized to visualize immune cell dynamics with vascular labelling by intravenous injection of anti-CD31 antibody and DiD-labelled red blood cells (RBCs). A heart imaging window chamber with a vacuum-based tissue motion stabilizer with 890-920 mbar was applied following a chest incision to expose the cardiac tissue. The suction-based heart imaging window chamber system and artificial intelligence-based motion compensation function significantly reduced motion artefacts and facilitated real-time in vivo cell analysis of immune cell and RBC trafficking, revealing a mean neutrophil movement velocity of 1.66 mm/s, which was slower compared to the RBC flow velocity of 9.22 mm/s. Intravital two-photon microscopic heart imaging enabled label-free second harmonic generation imaging of cardiac muscle structures with 820-840 nm excitation wavelength, revealing detailed biodistributions and structural variations in sarcomeres and fibrillar organization in the heart. Conclusion The optimized intravital heart imaging protocol successfully demonstrates its capability to provide high-resolution, real-time visualization of dynamic cellular activities within live cardiac tissue.
Collapse
Affiliation(s)
- Soyeon Ahn
- R&D Center, IVIM Technology, 17 Techno 4-ro, Yuseong-gu, Daejeon, 34013, Republic of Korea
| | - Jung-yeon Yoon
- R&D Center, IVIM Technology, 17 Techno 4-ro, Yuseong-gu, Daejeon, 34013, Republic of Korea
| | - Pilhan Kim
- R&D Center, IVIM Technology, 17 Techno 4-ro, Yuseong-gu, Daejeon, 34013, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
15
|
Banerji R, Grifno GN, Shi L, Smolen D, LeBourdais R, Muhvich J, Eberman C, Hiller BE, Lee J, Regan K, Zheng S, Zhang S, Jiang J, Raslan AA, Breda JC, Pihl R, Traber K, Mazzilli S, Ligresti G, Mizgerd JP, Suki B, Nia HT. Crystal ribcage: a platform for probing real-time lung function at cellular resolution. Nat Methods 2023; 20:1790-1801. [PMID: 37710017 PMCID: PMC10860663 DOI: 10.1038/s41592-023-02004-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 08/10/2023] [Indexed: 09/16/2023]
Abstract
Understanding the dynamic pathogenesis and treatment response in pulmonary diseases requires probing the lung at cellular resolution in real time. Despite advances in intravital imaging, optical imaging of the lung during active respiration and circulation has remained challenging. Here, we introduce the crystal ribcage: a transparent ribcage that allows multiscale optical imaging of the functioning lung from whole-organ to single-cell level. It enables the modulation of lung biophysics and immunity through intravascular, intrapulmonary, intraparenchymal and optogenetic interventions, and it preserves the three-dimensional architecture, air-liquid interface, cellular diversity and respiratory-circulatory functions of the lung. Utilizing these capabilities on murine models of pulmonary pathologies we probed remodeling of respiratory-circulatory functions at the single-alveolus and capillary levels during disease progression. The crystal ribcage and its broad applications presented here will facilitate further studies of nearly any pulmonary disease as well as lead to the identification of new targets for treatment strategies.
Collapse
Affiliation(s)
- Rohin Banerji
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Gabrielle N Grifno
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Linzheng Shi
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Dylan Smolen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rob LeBourdais
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Johnathan Muhvich
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cate Eberman
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Bradley E Hiller
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jisu Lee
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kathryn Regan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Siyi Zheng
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - John Jiang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ahmed A Raslan
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Julia C Breda
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Riley Pihl
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Katrina Traber
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sarah Mazzilli
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Giovanni Ligresti
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
16
|
Bousso P, Grandjean CL. Immunomodulation under the lens of real-time in vivo imaging. Eur J Immunol 2023; 53:e2249921. [PMID: 37051691 DOI: 10.1002/eji.202249921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 04/11/2023] [Indexed: 04/14/2023]
Abstract
Modulation of cells and molecules of the immune system not only represents a major opportunity to treat a variety of diseases including infections, cancer, autoimmune, and inflammatory disorders but could also help understand the intricacies of immune responses. A detailed mechanistic understanding of how a specific immune intervention may provide clinical benefit is essential for the rational design of efficient immunomodulators. Visualizing the impact of immunomodulation in real-time and in vivo has emerged as an important approach to achieve this goal. In this review, we aim to illustrate how multiphoton intravital imaging has helped clarify the mode of action of immunomodulatory strategies such as antibodies or cell therapies. We also discuss how optogenetics combined with imaging will further help manipulate and precisely understand immunomodulatory pathways. Combined with other single-cell technologies, in vivo dynamic imaging has therefore a major potential for guiding preclinical development of immunomodulatory drugs.
Collapse
Affiliation(s)
- Philippe Bousso
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| | - Capucine L Grandjean
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| |
Collapse
|
17
|
Burkard P, Schonhart C, Vögtle T, Köhler D, Tang L, Johnson D, Hemmen K, Heinze KG, Zarbock A, Hermanns HM, Rosenberger P, Nieswandt B. A key role for platelet GPVI in neutrophil recruitment, migration, and NETosis in the early stages of acute lung injury. Blood 2023; 142:1463-1477. [PMID: 37441848 DOI: 10.1182/blood.2023019940] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/13/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are associated with high morbidity and mortality. Excessive neutrophil infiltration into the pulmonary airspace is the main cause for the acute inflammation and lung injury. Platelets have been implicated in the pathogenesis of ALI/ARDS, but the underlying mechanisms are not fully understood. Here, we show that the immunoreceptor tyrosine-based activation motif-coupled immunoglobulin-like platelet receptor, glycoprotein VI (GPVI), plays a key role in the early phase of pulmonary thrombo-inflammation in a model of lipopolysaccharide (LPS)-induced ALI in mice. In wild-type (WT) control mice, intranasal LPS application triggered severe pulmonary and blood neutrophilia, hypothermia, and increased blood lactate levels. In contrast, GPVI-deficient mice as well as anti-GPVI-treated WT mice were markedly protected from pulmonary and systemic compromises and showed no increased pulmonary bleeding. High-resolution multicolor microscopy of lung sections and intravital confocal microcopy of the ventilated lung revealed that anti-GPVI treatment resulted in less stable platelet interactions with neutrophils and overall reduced platelet-neutrophil complex (PNC) formation. Anti-GPVI treatment also reduced neutrophil crawling and adhesion on endothelial cells, resulting in reduced neutrophil transmigration and alveolar infiltrates. Remarkably, neutrophil activation was also diminished in anti-GPVI-treated animals, associated with strongly reduced formation of PNC clusters and neutrophil extracellular traps (NETs) compared with that in control mice. These results establish GPVI as a key mediator of neutrophil recruitment, PNC formation, and NET formation (ie, NETosis) in experimental ALI. Thus, GPVI inhibition might be a promising strategy to reduce the acute pulmonary inflammation that causes ALI/ARDS.
Collapse
Affiliation(s)
- Philipp Burkard
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Charlotte Schonhart
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
| | - Timo Vögtle
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - David Köhler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Linyan Tang
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Denise Johnson
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Katherina Hemmen
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Heike M Hermanns
- Medical Clinic II, Division of Hepatology, University Hospital Würzburg, Würzburg, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| |
Collapse
|
18
|
Lin Q, Choyke PL, Sato N. Visualizing vasculature and its response to therapy in the tumor microenvironment. Theranostics 2023; 13:5223-5246. [PMID: 37908739 PMCID: PMC10614675 DOI: 10.7150/thno.84947] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/30/2023] [Indexed: 11/02/2023] Open
Abstract
Tumor vasculature plays a critical role in the progression and metastasis of tumors, antitumor immunity, drug delivery, and resistance to therapies. The morphological and functional changes of tumor vasculature in response to therapy take place in a spatiotemporal-dependent manner, which can be predictive of treatment outcomes. Dynamic monitoring of intratumor vasculature contributes to an improved understanding of the mechanisms of action of specific therapies or reasons for treatment failure, leading to therapy optimization. There is a rich history of methods used to image the vasculature. This review describes recent advances in imaging technologies to visualize the tumor vasculature, with a focus on enhanced intravital imaging techniques and tumor window models. We summarize new insights on spatial-temporal vascular responses to various therapies, including changes in vascular perfusion and permeability and immune-vascular crosstalk, obtained from intravital imaging. Finally, we briefly discuss the clinical applications of intravital imaging techniques.
Collapse
Affiliation(s)
| | | | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
19
|
McArdle S, Seo GY, Kronenberg M, Mikulski Z. Intravital Imaging of Intestinal Intraepithelial Lymphocytes. Bio Protoc 2023; 13:e4720. [PMID: 37497460 PMCID: PMC10366999 DOI: 10.21769/bioprotoc.4720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/21/2023] [Accepted: 05/04/2023] [Indexed: 07/28/2023] Open
Abstract
Intestinal intraepithelial lymphocytes (IEL) are a numerous population of T cells located within the epithelium of the small and large intestines, being more numerous in the small intestine (SI). They surveil this tissue by interacting with epithelial cells. Intravital microscopy is an important tool for visualizing the patrolling activity of IEL in the SI of live mice. Most IEL express CD8α; therefore, here we describe an established protocol of intravital imaging that tracks lymphocytes labeled with a CD8α-specific monoclonal antibody in the SI epithelium of live mice. We also describe data acquisition and quantification of the movement metrics, including mean speed, track length, displacement length, and paths for each CD8α+ IEL using the available software. The intravital imaging technique for measuring IEL movement will provide a better understanding of the role of IEL in homeostasis and protection from injury or infection in vivo.
Collapse
Affiliation(s)
- Sara McArdle
- La Jolla Institute for Immunology; La Jolla, CA, USA
| | - Goo-Young Seo
- La Jolla Institute for Immunology; La Jolla, CA, USA
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology; La Jolla, CA, USA
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
20
|
Hong Y, Walling BL, Kim HR, Serratelli WS, Lozada JR, Sailer CJ, Amitrano AM, Lim K, Mongre RK, Kim KD, Capece T, Lomakina EB, Reilly NS, Vo K, Gerber SA, Fan TC, Yu ALT, Oakes PW, Waugh RE, Jun CD, Reagan PM, Kim M. ST3GAL1 and βII-spectrin pathways control CAR T cell migration to target tumors. Nat Immunol 2023; 24:1007-1019. [PMID: 37069398 PMCID: PMC10515092 DOI: 10.1038/s41590-023-01498-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 03/21/2023] [Indexed: 04/19/2023]
Abstract
Adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells is becoming a promising treatment option for hematological malignancies. However, T cell immunotherapies have mostly failed in individuals with solid tumors. Here, with a CRISPR-Cas9 pooled library, we performed an in vivo targeted loss-of-function screen and identified ST3 β-galactoside α-2,3-sialyltransferase 1 (ST3GAL1) as a negative regulator of the cancer-specific migration of CAR T cells. Analysis of glycosylated proteins revealed that CD18 is a major effector of ST3GAL1 in activated CD8+ T cells. ST3GAL1-mediated glycosylation induces the spontaneous nonspecific tissue sequestration of T cells by altering lymphocyte function-associated antigen-1 (LFA-1) endocytic recycling. Engineered CAR T cells with enhanced expression of βII-spectrin, a central LFA-1-associated cytoskeleton molecule, reversed ST3GAL1-mediated nonspecific T cell migration and reduced tumor growth in mice by improving tumor-specific homing of CAR T cells. These findings identify the ST3GAL1-βII-spectrin axis as a major cell-intrinsic program for cancer-targeting CAR T cell migration and as a promising strategy for effective T cell immunotherapy.
Collapse
Affiliation(s)
- Yeonsun Hong
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Brandon L Walling
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Hye-Ran Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - William S Serratelli
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - John R Lozada
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Cooper J Sailer
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrea M Amitrano
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Raj Kumar Mongre
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Kyun-Do Kim
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Tara Capece
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Elena B Lomakina
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Nicholas S Reilly
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, USA
| | - Kevin Vo
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Scott A Gerber
- Department of Surgery, University of Rochester, Rochester, NY, USA
| | - Tan-Chi Fan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Alice Lin-Tsing Yu
- Department of Pediatrics/Hematology Oncology, University of California in San Diego, San Diego, CA, USA
| | - Patrick W Oakes
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, USA
| | - Richard E Waugh
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Patrick M Reagan
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
21
|
Chakraborty S, Tabrizi Z, Bhatt NN, Franciosa SA, Bracko O. A Brief Overview of Neutrophils in Neurological Diseases. Biomolecules 2023; 13:biom13050743. [PMID: 37238612 DOI: 10.3390/biom13050743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Neutrophils are the most abundant leukocyte in circulation and are the first line of defense after an infection or injury. Neutrophils have a broad spectrum of functions, including phagocytosis of microorganisms, the release of pro-inflammatory cytokines and chemokines, oxidative burst, and the formation of neutrophil extracellular traps. Traditionally, neutrophils were thought to be most important for acute inflammatory responses, with a short half-life and a more static response to infections and injury. However, this view has changed in recent years showing neutrophil heterogeneity and dynamics, indicating a much more regulated and flexible response. Here we will discuss the role of neutrophils in aging and neurological disorders; specifically, we focus on recent data indicating the impact of neutrophils in chronic inflammatory processes and their contribution to neurological diseases. Lastly, we aim to conclude that reactive neutrophils directly contribute to increased vascular inflammation and age-related diseases.
Collapse
Affiliation(s)
| | - Zeynab Tabrizi
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | | | | | - Oliver Bracko
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
- Department of Neurology, University of Miami-Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
22
|
Babes L, Yipp BG, Senger DL. Intravital Microscopy of the Metastatic Pulmonary Environment. Methods Mol Biol 2023; 2614:383-396. [PMID: 36587137 DOI: 10.1007/978-1-0716-2914-7_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Real-time in vivo imaging has become an integral tool for the investigation and understanding of cellular processes in health and disease at single-cell resolution. This includes the dynamic and complex cellular interactions that occur during cancer progression and the subsequent metastatic dissemination of tumor cells to sites distant from the primary tumor. Herein we outline the methodology for the establishment and intravital imaging of the pulmonary metastatic niche, a preferred site of metastasis for many cancers, and describe the implementation of a lung window to visualize and dissect the intricate behaviour of multiple cell types within this environment. We also address the advantages and limitations of this high-resolution technology.
Collapse
Affiliation(s)
- Liane Babes
- Lady Davis Institute for Medical Research, Montreal, QC, Canada
| | - Bryan George Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Donna Lorraine Senger
- Lady Davis Institute for Medical Research, Montreal, QC, Canada. .,Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
23
|
Review on the Biogenesis of Platelets in Lungs and Its Alterations in SARS-CoV-2 Infection Patients. J Renin Angiotensin Aldosterone Syst 2023; 2023:7550197. [PMID: 36891250 PMCID: PMC9988383 DOI: 10.1155/2023/7550197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 01/28/2023] [Accepted: 02/08/2023] [Indexed: 03/01/2023] Open
Abstract
Thrombocytes (platelets) are the type of blood cells that are involved in hemostasis, thrombosis, etc. For the conversion of megakaryocytes into thrombocytes, the thrombopoietin (TPO) protein is essential which is encoded by the TPO gene. TPO gene is present in the long arm of chromosome number 3 (3q26). This TPO protein interacts with the c-Mpl receptor, which is present on the outer surface of megakaryocytes. As a result, megakaryocyte breaks into the production of functional thrombocytes. Some of the evidence shows that the megakaryocytes, the precursor of thrombocytes, are seen in the lung's interstitium. This review focuses on the involvement of the lungs in the production of thrombocytes and their mechanism. A lot of findings show that viral diseases, which affect the lungs, cause thrombocytopenia in human beings. One of the notable viral diseases is COVID-19 or severe acute respiratory syndrome caused by SARS-associated coronavirus 2 (SARS-CoV-2). SARS-CoV-2 caused a worldwide alarm in 2019 and a lot of people suffered because of this disease. It mainly targets the lung cells for its replication. To enter the cells, these virus targets the angiotensin-converting enzyme-2 (ACE-2) receptors that are abundantly seen on the surface of the lung cells. Recent reports of COVID-19-affected patients reveal the important fact that these peoples develop thrombocytopenia as a post-COVID condition. This review elaborates on the biogenesis of platelets in the lungs and the alterations of thrombocytes during the COVID-19 infection.
Collapse
|
24
|
Dean CH, Cheong SS. Simple Models of Lung Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:17-28. [PMID: 37195524 DOI: 10.1007/978-3-031-26625-6_2] [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: 05/18/2023]
Abstract
Models are essential to further our understanding of lung development and regeneration and to facilitate identification and testing of potential treatments for lung diseases. A wide variety of rodent and human models are available that recapitulate one or more stages of lung development. This chapter describes the existing 'simple' in vitro, in silico and ex vivo models of lung development. We define which stage(s) of development each model recapitulates and highlight their pros and cons.
Collapse
Affiliation(s)
- Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Sek-Shir Cheong
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
25
|
Paulson B, Darian SB, Kim Y, Oh J, Ghasemi M, Lee K, Kim JK. Spectral Multiplexing of Fluorescent Endoscopy for Simultaneous Imaging with Multiple Fluorophores and Multiple Fields of View. BIOSENSORS 2022; 13:33. [PMID: 36671868 PMCID: PMC9855833 DOI: 10.3390/bios13010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Complex clinical procedures and small-animal research procedures can benefit from dual-site imaging provided by multiple endoscopic devices. Here, an endoscopic system is proposed which enables multiple fluorescence microendoscopes to be spectrally multiplexed on a single microscope base, enabling light sources and optical relays to be shared between endoscopes. The presented system is characterized for resolution using USAF-1951 resolution test charts and for modulation transfer function using the slanted edge method. Imaging is demonstrated both directly and with microendoscopes attached. Imaging of phantoms was demonstrated by targeting USAF charts and fiber tissues dyed for FITC and Texas Red fluorescence. Afterwards, simultaneous liver and kidney imaging was demonstrated in mice expressing mitochondrial Dendra2 and injected with Texas Red-dextran. The results indicate that the system achieves high channel isolation and submicron and subcellular resolution, with resolution limited by the endoscopic probe and by physiological movement during endoscopic imaging. Multi-channel microendoscopy provides a potentially low-cost means of simultaneous multiple endoscopic imaging during biological experiments, resulting in reduced animal harm and potentially increasing insight into temporal connections between connected biological systems.
Collapse
Affiliation(s)
- Bjorn Paulson
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Saeed Bohlooli Darian
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Youngkyu Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jeongmin Oh
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Marjan Ghasemi
- Department of Physics, Yonsei University, Seoul 03722, Republic of Korea
| | - Kwanhee Lee
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jun Ki Kim
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| |
Collapse
|
26
|
Politowicz AL, Burks AT, Dong Y, Htwe YM, Dudek SM, Elisabeta Marai G, Belvitch P. Alveolus analysis: a web browser-based tool to analyze lung intravital microscopy. BMC Pulm Med 2022; 22:480. [PMID: 36528564 PMCID: PMC9759058 DOI: 10.1186/s12890-022-02274-7] [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: 10/20/2021] [Accepted: 12/03/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Acute lung injury and the acute respiratory distress syndrome are characterized by pulmonary inflammation, reduced endothelial barrier integrity and filling of the alveolar space with protein rich edema fluid and infiltrating leukocytes. Animal models are critical to uncovering the pathologic mechanisms of this devastating syndrome. Intravital imaging of the intact lung via two-photon intravital microscopy has proven a valuable method to investigate lung injury in small rodent models through characterization of inflammatory cells and vascular changes in real time. However, respiratory motion complicates the analysis of these time series images and requires selective data extraction to stabilize the image. Consequently, analysis of individual alveoli may not provide a complete picture of the integrated mechanical, vascular and inflammatory processes occurring simultaneously in the intact lung. To address these challenges, we developed a web browser-based visualization application named Alveolus Analysis to process, analyze and graphically display intravital lung microscopy data. RESULTS The designed tool takes raw temporal image data as input, performs image preprocessing and feature extraction offline, and visualizes the extracted information in a web browser-based interface. The interface allows users to explore multiple experiments in three panels corresponding to different levels of detail: summary statistics of alveolar/neutrophil behavior, characterization of alveolar dynamics including lung edema and inflammatory cells at specific time points, and cross-experiment analysis. We performed a case study on the utility of the visualization with two members or our research team and they found the tool useful because of its ability to preprocess data consistently and visualize information in a digestible and informative format. CONCLUSIONS Application of our software tool, Alveolus Analysis, to intravital lung microscopy data has the potential to enhance the information gained from these experiments and provide new insights into the pathologic mechanisms of inflammatory lung injury.
Collapse
Affiliation(s)
- Alexander L Politowicz
- Department of Computer Science, College of Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Andrew T Burks
- Department of Computer Science, College of Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Yushen Dong
- Department of Mathematics, Statistics, and Computer Science, University of Illinois Chicago, Chicago, IL, USA
| | - Yu Maw Htwe
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois Chicago, CSB 915, 840 S. Wood St., Chicago, IL, 60612, USA
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois Chicago, CSB 915, 840 S. Wood St., Chicago, IL, 60612, USA
| | - G Elisabeta Marai
- Department of Computer Science, College of Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Patrick Belvitch
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois Chicago, CSB 915, 840 S. Wood St., Chicago, IL, 60612, USA.
| |
Collapse
|
27
|
Sarden N, Sinha S, Potts KG, Pernet E, Hiroki CH, Hassanabad MF, Nguyen AP, Lou Y, Farias R, Winston BW, Bromley A, Snarr BD, Zucoloto AZ, Andonegui G, Muruve DA, McDonald B, Sheppard DC, Mahoney DJ, Divangahi M, Rosin N, Biernaskie J, Yipp BG. A B1a-natural IgG-neutrophil axis is impaired in viral- and steroid-associated aspergillosis. Sci Transl Med 2022; 14:eabq6682. [PMID: 36475902 DOI: 10.1126/scitranslmed.abq6682] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The lung naturally resists Aspergillus fumigatus (Af) in healthy individuals, but multiple conditions can disrupt this resistance, leading to lethal invasive infections. Core processes of natural resistance and its breakdown are undefined. We investigated three distinct conditions predisposing to lethal aspergillosis-severe SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection, influenza A viral pneumonia, and systemic corticosteroid use-in human patients and murine models. We found a conserved and essential coupling of innate B1a lymphocytes, Af-binding natural immunoglobulin G antibodies, and lung neutrophils. Failure of this axis concealed Af from neutrophils, allowing rapid fungal invasion and disease. Reconstituting the axis with immunoglobulin therapy reestablished resistance, thus representing a realistic pathway to repurpose currently available therapies. Together, we report a vital host resistance pathway that is responsible for protecting against life-threatening aspergillosis in the context of distinct susceptibilities.
Collapse
Affiliation(s)
- Nicole Sarden
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Kyle G Potts
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Erwan Pernet
- Meakins-Christie Laboratories, Departments of Medicine and Pathology, McGill International TB Centre, McGill University, Montreal, QC H4A 3JI, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Carlos H Hiroki
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mortaza F Hassanabad
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Angela P Nguyen
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yuefei Lou
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Raquel Farias
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Brent W Winston
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Amy Bromley
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Brendan D Snarr
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Amanda Z Zucoloto
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Graciela Andonegui
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Daniel A Muruve
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Braedon McDonald
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Donald C Sheppard
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada.,Division of Infectious Diseases and Department of Medical Microbiology, McGill University Health Centre, Montreal, QC H4A 3JI, Canada
| | - Douglas J Mahoney
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Departments of Medicine and Pathology, McGill International TB Centre, McGill University, Montreal, QC H4A 3JI, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Nicole Rosin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Bryan G Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
28
|
Gelon L, Fromont L, Lefrançais E. Occurrence and role of lung megakaryocytes in infection and inflammation. Front Immunol 2022; 13:1029223. [PMID: 36524131 PMCID: PMC9745136 DOI: 10.3389/fimmu.2022.1029223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/09/2022] [Indexed: 12/03/2022] Open
Abstract
Megakaryocytes (MKs) are large cells giving rise to platelets. It is well established that in adults, MKs develop from hematopoietic stem cells and reside in the bone marrow. MKs are also rare but normal constituents of the venous blood returning to the lungs, and MKs are found in the lung vasculature (MKcirc), suggesting that these cells are migrants from the bone marrow and get trapped in lung capillaries where the final steps of platelet production can occur. An unprecedented increase in the number of lung and circulating MKs was described in coronavirus disease 2019 (COVID-19) patients, suggesting that lung thrombopoiesis may be increased during lung infection and/or thromboinflammation. In addition to the population of platelet-producing intravascular MKs in the lung, a population of lung-resident megakaryocytes (MKL) has been identified and presents a specific immune signature compared to its bone marrow counterparts. Recent single-cell analysis and intravital imaging have helped us gain a better understanding of these populations in mouse and human. This review aims at summarizing the recent data on increased occurrence of lung MKs and discusses their origin, specificities, and potential role in homeostasis and inflammatory and infectious lung diseases. Here, we address remaining questions, controversies, and methodologic challenges for further studies of both MKcirc and MKL.
Collapse
|
29
|
Seo GY, Takahashi D, Wang Q, Mikulski Z, Chen A, Chou TF, Marcovecchio P, McArdle S, Sethi A, Shui JW, Takahashi M, Surh CD, Cheroutre H, Kronenberg M. Epithelial HVEM maintains intraepithelial T cell survival and contributes to host protection. Sci Immunol 2022; 7:eabm6931. [PMID: 35905286 PMCID: PMC9422995 DOI: 10.1126/sciimmunol.abm6931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intraepithelial T cells (IETs) are in close contact with intestinal epithelial cells and the underlying basement membrane, and they detect invasive pathogens. How intestinal epithelial cells and basement membrane influence IET survival and function, at steady state or after infection, is unclear. The herpes virus entry mediator (HVEM), a member of the TNF receptor superfamily, is constitutively expressed by intestinal epithelial cells and is important for protection from pathogenic bacteria. Here, we showed that at steady-state LIGHT, an HVEM ligand, binding to epithelial HVEM promoted the survival of small intestine IETs. RNA-seq and addition of HVEM ligands to epithelial organoids indicated that HVEM increased epithelial synthesis of basement membrane proteins, including collagen IV, which bound to β1 integrins expressed by IETs. Therefore, we proposed that IET survival depended on β1 integrin binding to collagen IV and showed that β1 integrin-collagen IV interactions supported IET survival in vitro. Moreover, the absence of β1 integrin expression by T lymphocytes decreased TCR αβ+ IETs in vivo. Intravital microscopy showed that the patrolling movement of IETs was reduced without epithelial HVEM. As likely consequences of decreased number and movement, protective responses to Salmonella enterica were reduced in mice lacking either epithelial HVEM, HVEM ligands, or β1 integrins. Therefore, IETs, at steady state and after infection, depended on HVEM expressed by epithelial cells for the synthesis of collagen IV by epithelial cells. Collagen IV engaged β1 integrins on IETs that were important for their maintenance and for their protective function in mucosal immunity.
Collapse
Affiliation(s)
- Goo-Young Seo
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Qingyang Wang
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Angeline Chen
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Sara McArdle
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Ashu Sethi
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jr-Wen Shui
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Charles D Surh
- La Jolla Institute for Immunology, La Jolla, CA, USA.,Institute for Basic Science (IBS), Academy of Immunology and Microbiology, Pohang, South Korea
| | | | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, USA.,Division of Biology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
30
|
Caspase-1-driven neutrophil pyroptosis and its role in host susceptibility to Pseudomonas aeruginosa. PLoS Pathog 2022; 18:e1010305. [PMID: 35849616 PMCID: PMC9345480 DOI: 10.1371/journal.ppat.1010305] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 08/02/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
Multiple regulated neutrophil cell death programs contribute to host defense against infections. However, despite expressing all necessary inflammasome components, neutrophils are thought to be generally defective in Caspase-1-dependent pyroptosis. By screening different bacterial species, we found that several Pseudomonas aeruginosa (P. aeruginosa) strains trigger Caspase-1-dependent pyroptosis in human and murine neutrophils. Notably, deletion of Exotoxins U or S in P. aeruginosa enhanced neutrophil death to Caspase-1-dependent pyroptosis, suggesting that these exotoxins interfere with this pathway. Mechanistically, P. aeruginosa Flagellin activates the NLRC4 inflammasome, which supports Caspase-1-driven interleukin (IL)-1β secretion and Gasdermin D (GSDMD)-dependent neutrophil pyroptosis. Furthermore, P. aeruginosa-induced GSDMD activation triggers Calcium-dependent and Peptidyl Arginine Deaminase-4-driven histone citrullination and translocation of neutrophil DNA into the cell cytosol without inducing extracellular Neutrophil Extracellular Traps. Finally, we show that neutrophil Caspase-1 contributes to IL-1β production and susceptibility to pyroptosis-inducing P. aeruginosa strains in vivo. Overall, we demonstrate that neutrophils are not universally resistant for Caspase-1-dependent pyroptosis. Neutrophils play an essential role against infections. Although multiple neutrophil death programs contribute to host defense against infections, neutrophils are thought to be defective in Caspase-1-dependent pyroptosis. We screened several microbial species for the capacity to overcome neutrophil resistance to Caspase-1-driven pyroptosis, and show that the bacterium Pseudomonas aeruginosa specifically engages the NLRC4 inflammasome to promote Caspase-1-dependent Gasdermin D activation and subsequent neutrophil pyroptosis. Furthermore, NLRC4 inflammasome-driven pyroptosis leads to histone citrullination, nuclear DNA decondensation and expansion into the host cell cytosol. However, Neutrophil Extracellular Trap (NET) are not formed because DNA is kept in the intracellular space despite plasma membrane permeabilization and extracellular release of soluble and insoluble alarmins. Finally, in vivo P. aeruginosa infections highlight that Caspase-1-driven neutrophil pyroptosis is detrimental to the host upon P. aeruginosa infection. Altogether, our results demonstrate Caspase-1-dependent pyroptosis in neutrophils as a process that contributes to host susceptibility to P. aeruginosa infection.
Collapse
|
31
|
Herminghaus A, Kozlov AV, Szabó A, Hantos Z, Gylstorff S, Kuebart A, Aghapour M, Wissuwa B, Walles T, Walles H, Coldewey SM, Relja B. A Barrier to Defend - Models of Pulmonary Barrier to Study Acute Inflammatory Diseases. Front Immunol 2022; 13:895100. [PMID: 35874776 PMCID: PMC9300899 DOI: 10.3389/fimmu.2022.895100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Pulmonary diseases represent four out of ten most common causes for worldwide mortality. Thus, pulmonary infections with subsequent inflammatory responses represent a major public health concern. The pulmonary barrier is a vulnerable entry site for several stress factors, including pathogens such as viruses, and bacteria, but also environmental factors e.g. toxins, air pollutants, as well as allergens. These pathogens or pathogen-associated molecular pattern and inflammatory agents e.g. damage-associated molecular pattern cause significant disturbances in the pulmonary barrier. The physiological and biological functions, as well as the architecture and homeostatic maintenance of the pulmonary barrier are highly complex. The airway epithelium, denoting the first pulmonary barrier, encompasses cells releasing a plethora of chemokines and cytokines, and is further covered with a mucus layer containing antimicrobial peptides, which are responsible for the pathogen clearance. Submucosal antigen-presenting cells and neutrophilic granulocytes are also involved in the defense mechanisms and counterregulation of pulmonary infections, and thus may directly affect the pulmonary barrier function. The detailed understanding of the pulmonary barrier including its architecture and functions is crucial for the diagnosis, prognosis, and therapeutic treatment strategies of pulmonary diseases. Thus, considering multiple side effects and limited efficacy of current therapeutic treatment strategies in patients with inflammatory diseases make experimental in vitro and in vivo models necessary to improving clinical therapy options. This review describes existing models for studyying the pulmonary barrier function under acute inflammatory conditions, which are meant to improve the translational approaches for outcome predictions, patient monitoring, and treatment decision-making.
Collapse
Affiliation(s)
- Anna Herminghaus
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Andrey V. Kozlov
- L Boltzmann Institute for Traumatology in Cooperation with AUVA and Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department of Human Pathology , IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Zoltán Hantos
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Severin Gylstorff
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Kuebart
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Mahyar Aghapour
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Bianka Wissuwa
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, Magdeburg University Medicine, Magdeburg, Germany
| | - Heike Walles
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- Core Facility Tissue Engineering, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sina M. Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- *Correspondence: Borna Relja,
| |
Collapse
|
32
|
Jeong S, Kim B, Byun DJ, Jin S, Seo BS, Shin MH, Leem AY, Choung JJ, Park MS, Hyun YM. Lysophosphatidylcholine Alleviates Acute Lung Injury by Regulating Neutrophil Motility and Neutrophil Extracellular Trap Formation. Front Cell Dev Biol 2022; 10:941914. [PMID: 35859904 PMCID: PMC9289271 DOI: 10.3389/fcell.2022.941914] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/14/2022] [Indexed: 11/28/2022] Open
Abstract
Sepsis is predominantly initiated by bacterial infection and can cause systemic inflammation, which frequently leads to rapid death of the patient. However, this acute systemic inflammatory response requires further investigation from the perspectives of clinical judgment criteria and early treatment strategies for the relief of symptoms. Lysophosphatidylcholine (LPC) 18:0 may relieve septic symptoms, but the relevant mechanism is not clearly understood. Therefore, we aimed to assess the effectiveness of LPC as a therapeutic treatment for acute inflammation in the lung induced by lipopolysaccharide in mice. Systemic inflammation of mice was induced by lipopolysaccharide (LPS) inoculation to investigate the role of LPC in the migration and the immune response of neutrophils during acute lung injury. By employing two-photon intravital imaging of the LPS-stimulated LysM-GFP mice and other in vitro and in vivo assays, we examined whether LPC alleviates the inflammatory effect of sepsis. We also tested the effect of LPC to human neutrophils from healthy control and sepsis patients. Our data showed that LPC treatment reduced the infiltration of innate immune cells into the lung. Specifically, LPC altered neutrophil migratory patterns and enhanced phagocytic efficacy in the damaged lung. Moreover, LPC treatment reduced the release of neutrophil extracellular trap (NET), which can damage tissue in the inflamed organ and exacerbate disease. It also reduced human neutrophil migration under inflammatory environment. Our results suggest that LPC can alleviate sepsis-induced lung inflammation by regulating the function of neutrophils. These findings provide evidence for the beneficial application of LPC treatment as a potential therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Soi Jeong
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Bora Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Da Jeong Byun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sunmin Jin
- R&D Center, AriBio Co., Ltd., Sengnam, South Korea
| | - Bo Seung Seo
- R&D Center, AriBio Co., Ltd., Sengnam, South Korea
| | - Mi Hwa Shin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ah Young Leem
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Moo Suk Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Moo Suk Park, ; Young-Min Hyun,
| | - Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Moo Suk Park, ; Young-Min Hyun,
| |
Collapse
|
33
|
Shim HB, Deniset JF, Kubes P. Neutrophils in homeostasis and tissue repair. Int Immunol 2022; 34:399-407. [PMID: 35752158 DOI: 10.1093/intimm/dxac029] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/25/2022] [Indexed: 11/13/2022] Open
Abstract
Neutrophils are the most abundant innate immune cell and are equipped with highly destructive molecular cargo. As such, these cells were long thought to be short-lived killer cells that unleash their full cytotoxic programs on pathogens following infection and on host bystander cells after sterile injury. However, this view of neutrophils is overly simplistic and as a result is outdated. Numerous studies now collectively highlight neutrophils as far more complex and having a host of homeostatic and tissue-reparative functions. In this review, we summarize these underappreciated roles across organs and injury models.
Collapse
Affiliation(s)
- Hanjoo Brian Shim
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Justin F Deniset
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
34
|
Shaw PA, Forsyth E, Haseeb F, Yang S, Bradley M, Klausen M. Two-Photon Absorption: An Open Door to the NIR-II Biological Window? Front Chem 2022; 10:921354. [PMID: 35815206 PMCID: PMC9263132 DOI: 10.3389/fchem.2022.921354] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
The way in which photons travel through biological tissues and subsequently become scattered or absorbed is a key limitation for traditional optical medical imaging techniques using visible light. In contrast, near-infrared wavelengths, in particular those above 1000 nm, penetrate deeper in tissues and undergo less scattering and cause less photo-damage, which describes the so-called "second biological transparency window". Unfortunately, current dyes and imaging probes have severely limited absorption profiles at such long wavelengths, and molecular engineering of novel NIR-II dyes can be a tedious and unpredictable process, which limits access to this optical window and impedes further developments. Two-photon (2P) absorption not only provides convenient access to this window by doubling the absorption wavelength of dyes, but also increases the possible resolution. This review aims to provide an update on the available 2P instrumentation and 2P luminescent materials available for optical imaging in the NIR-II window.
Collapse
Affiliation(s)
| | | | | | | | | | - Maxime Klausen
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
35
|
Gawne P, Man F, Blower PJ, T. M. de Rosales R. Direct Cell Radiolabeling for in Vivo Cell Tracking with PET and SPECT Imaging. Chem Rev 2022; 122:10266-10318. [PMID: 35549242 PMCID: PMC9185691 DOI: 10.1021/acs.chemrev.1c00767] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 02/07/2023]
Abstract
The arrival of cell-based therapies is a revolution in medicine. However, its safe clinical application in a rational manner depends on reliable, clinically applicable methods for determining the fate and trafficking of therapeutic cells in vivo using medical imaging techniques─known as in vivo cell tracking. Radionuclide imaging using single photon emission computed tomography (SPECT) or positron emission tomography (PET) has several advantages over other imaging modalities for cell tracking because of its high sensitivity (requiring low amounts of probe per cell for imaging) and whole-body quantitative imaging capability using clinically available scanners. For cell tracking with radionuclides, ex vivo direct cell radiolabeling, that is, radiolabeling cells before their administration, is the simplest and most robust method, allowing labeling of any cell type without the need for genetic modification. This Review covers the development and application of direct cell radiolabeling probes utilizing a variety of chemical approaches: organic and inorganic/coordination (radio)chemistry, nanomaterials, and biochemistry. We describe the key early developments and the most recent advances in the field, identifying advantages and disadvantages of the different approaches and informing future development and choice of methods for clinical and preclinical application.
Collapse
Affiliation(s)
- Peter
J. Gawne
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Francis Man
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
- Institute
of Pharmaceutical Science, School of Cancer
and Pharmaceutical Sciences, King’s College London, London, SE1 9NH, U.K.
| | - Philip J. Blower
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| |
Collapse
|
36
|
De Niz M, Figueiredo LM. Surgical and intravital microscopy protocol to image Trypanosoma brucei–host interactions in live rodent models. STAR Protoc 2022; 3:101450. [PMID: 35719266 PMCID: PMC9201065 DOI: 10.1016/j.xpro.2022.101450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
37
|
Wen SW, Shim R, Hall P, Bedo J, Wilson JL, Nicholls AJ, Hickey MJ, Wong CHY. Lung Imaging Reveals Stroke-Induced Impairment in Pulmonary Intravascular Neutrophil Function, a Response Exacerbated with Aging. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2019-2028. [PMID: 35365565 DOI: 10.4049/jimmunol.2100997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/05/2022] [Indexed: 06/14/2023]
Abstract
In stroke patients, infection is a significant contributor to morbidity and mortality. Moreover, older stroke patients show an increased risk of developing stroke-associated infection, although the mechanisms underlying this increased susceptibility to infection are unknown. In this study, using an experimental mouse model of ischemic stroke, we showed that older (12-15 mo of age) mice had elevated lung bacterial infection and inflammatory damage after stroke when compared with young (8-10 wk of age) counterparts, despite undergoing the same degree of brain injury. Intravital microscopy of the lung microvasculature revealed that in younger mice, stroke promoted neutrophil arrest in pulmonary microvessels, but this response was not seen in older poststroke mice. In addition, bacterial phagocytosis by neutrophils in the lung microvasculature was reduced by both aging and stroke, such that neutrophils in aged poststroke mice showed the greatest impairment in this function. Analysis of neutrophil migration in vitro and in the cremaster muscle demonstrated that stroke alone did not negatively impact neutrophil migration, but that the combination of increased age and stroke led to reduced effectiveness of neutrophil chemotaxis. Transcriptomic analysis of pulmonary neutrophils using RNA sequencing identified 79 genes that were selectively altered in the context of combined aging and stroke, and they were associated with pathways that control neutrophil chemotaxis. Taken together, the findings of this study show that stroke in older animals results in worsening of neutrophil antibacterial responses and changes in neutrophil gene expression that have the potential to underpin elevated risk of stroke-associated infection in the context of increased age.
Collapse
Affiliation(s)
- Shu Wen Wen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Raymond Shim
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Pam Hall
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Justin Bedo
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; and
- School of Computing and Information Systems, The University of Melbourne, Parkville, Victoria, Australia
| | - Jenny L Wilson
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Alyce J Nicholls
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Connie H Y Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia;
| |
Collapse
|
38
|
Valet C, Magnen M, Qiu L, Cleary SJ, Wang KM, Ranucci S, Grockowiak E, Boudra R, Conrad C, Seo Y, Calabrese DR, Greenland JR, Leavitt AD, Passegué E, Méndez-Ferrer S, Swirski FK, Looney MR. Sepsis promotes splenic production of a protective platelet pool with high CD40 ligand expression. J Clin Invest 2022; 132:e153920. [PMID: 35192546 PMCID: PMC8970674 DOI: 10.1172/jci153920] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Platelets have a wide range of functions including critical roles in hemostasis, thrombosis, and immunity. We hypothesized that during acute inflammation, such as in life-threatening sepsis, there are fundamental changes in the sites of platelet production and phenotypes of resultant platelets. Here, we showed during sepsis that the spleen was a major site of megakaryopoiesis and platelet production. Sepsis provoked an adrenergic-dependent mobilization of megakaryocyte-erythrocyte progenitors (MEPs) from the bone marrow to the spleen, where IL-3 induced their differentiation into megakaryocytes (MKs). In the spleen, immune-skewed MKs produced a CD40 ligandhi platelet population with potent immunomodulatory functions. Transfusions of post-sepsis platelets enriched from splenic production enhanced immune responses and reduced overall mortality in sepsis-challenged animals. These findings identify a spleen-derived protective platelet population that may be broadly immunomodulatory in acute inflammatory states such as sepsis.
Collapse
Affiliation(s)
- Colin Valet
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Mélia Magnen
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Longhui Qiu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Simon J. Cleary
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Kristin M. Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Serena Ranucci
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Elodie Grockowiak
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Rafik Boudra
- Department of Dermatology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Yurim Seo
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | | | | | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York, USA
| | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge, United Kingdom
| | - Filip K. Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mark R. Looney
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
39
|
An JH, Qi FR, Cheng XY, Liu XQ, Luo P, Chen Q, Qian S, Zhang YHZ, Lian L, Guo Z, Liu L, Tan XH. Dynamic Characteristics of Blood Platelet Count in COVID-19 Patients. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background and purpose: Coronavirus disease 2019 (COVID-19) was spreading all over the world. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) primarily invades and infects the lungs of humans leading to COVID-19. Mild to severe clinical symptoms such as fever, cough,
and shortness of breath were existed in those patients. One of the most common changes in these patients was abnormal blood routine. However, uncertainty remains regarding the dynamic characteristics of platelet in COVID-19 patients due to limited data. Therefore, we aimed to analyze the association
between dynamic characteristics of blood platelet and disease severity, and to identify new monitoring indicators to treat the COVID-19 patients. Methods: In this cohort study, 398 COVID-19 patients treated in the Shenzhen Third People’s hospital from December 16, 2019 to March
26, 2020 were collected and participated. All data of participants including the clinical characteristics, imaging and laboratory information were collected. All patients included in our study were classified as four groups (mild, common, severe, and critical types) regarding clinical symptoms
and relevant severe failures based on the Diagnosis Criteria. Platelet count was examined at the baseline and every 3–5 days during hospitalization. Results: The platelet count varied with clinical classifications. The platelet count in mild type was normal without significant
fluctuation. While the blood platelet count of most common and severe patients had obvious fluctuations, showing as a dynamic change that first rose and then fell to the level at admission, which was consistent with the trend of lung inflammation. Bone marrow smears further showed that bone
marrow hyperplasia was normal in mild, common and severe type patients, and megakaryocytes and their platelet-producing functions were not abnormal. Conclusions: Our results suggested that the dynamic changes of platelet count might be a predictor of lung inflammation alteration for
COVID-19 patients. The changes in platelet count might be a responsive pattern secondary to lung inflammation. The function of bone marrow may be slightly affected by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jiang-Hong An
- Department of Oncology and Hematology, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Fu-Rong Qi
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Xiao-Ya Cheng
- Department of Oncology and Hematology, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Xun-Qi Liu
- Department of Oncology and Hematology, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Pu Luo
- Department of Oncology and Hematology, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Qiong Chen
- Department of Oncology and Hematology, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Shen Qian
- Institute of Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| | - Yi-Hui-Zhi Zhang
- Department of Hematology & Oncology, National Cancer Center/National Clinical Research Cancer for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116,
China
| | - Li Lian
- Department of Hematology & Oncology, National Cancer Center/National Clinical Research Cancer for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Zhi Guo
- Department of Hematology & Oncology, National Cancer Center/National Clinical Research Cancer for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Lei Liu
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518112, Guangdong Province, China
| | - Xiao-Hua Tan
- Department of Oncology and Hematology, Shenzhen Third People’s Hospital, Shenzhen, 518112, Guangdong Province, China
| |
Collapse
|
40
|
Bachmaier K, Stuart A, Singh A, Mukhopadhyay A, Chakraborty S, Hong Z, Wang L, Tsukasaki Y, Maienschein-Cline M, Ganesh BB, Kanteti P, Rehman J, Malik AB. Albumin Nanoparticle Endocytosing Subset of Neutrophils for Precision Therapeutic Targeting of Inflammatory Tissue Injury. ACS NANO 2022; 16:4084-4101. [PMID: 35230826 PMCID: PMC8945372 DOI: 10.1021/acsnano.1c09762] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/23/2022] [Indexed: 05/30/2023]
Abstract
The complex involvement of neutrophils in inflammatory diseases makes them intriguing but challenging targets for therapeutic intervention. Here, we tested the hypothesis that varying endocytosis capacities would delineate functionally distinct neutrophil subpopulations that could be specifically targeted for therapeutic purposes. By using uniformly sized (∼120 nm in diameter) albumin nanoparticles (ANP) to characterize mouse neutrophils in vivo, we found two subsets of neutrophils, one that readily endocytosed ANP (ANPhigh neutrophils) and another that failed to endocytose ANP (ANPlow population). These ANPhigh and ANPlow subsets existed side by side simultaneously in bone marrow, peripheral blood, spleen, and lungs, both under basal conditions and after inflammatory challenge. Human peripheral blood neutrophils showed a similar duality. ANPhigh and ANPlow neutrophils had distinct cell surface marker expression and transcriptomic profiles, both in naive mice and in mice after endotoxemic challenge. ANPhigh and ANPlow neutrophils were functionally distinct in their capacities to kill bacteria and to produce inflammatory mediators. ANPhigh neutrophils produced inordinate amounts of reactive oxygen species and inflammatory chemokines and cytokines. Targeting this subset with ANP loaded with the drug piceatannol, a spleen tyrosine kinase (Syk) inhibitor, mitigated the effects of polymicrobial sepsis by reducing tissue inflammation while fully preserving neutrophilic host-defense function.
Collapse
Affiliation(s)
- Kurt Bachmaier
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
- Nano
Biotherapeutics, Inc., 2201 West Campbell Park Drive, Chicago, Illinois 60612, United States
| | - Andrew Stuart
- Nano
Biotherapeutics, Inc., 2201 West Campbell Park Drive, Chicago, Illinois 60612, United States
| | - Abhalaxmi Singh
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
- Nano
Biotherapeutics, Inc., 2201 West Campbell Park Drive, Chicago, Illinois 60612, United States
| | - Amitabha Mukhopadhyay
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
| | - Sreeparna Chakraborty
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
| | - Zhigang Hong
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
| | - Li Wang
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
- Division
of Cardiology, Department of Medicine, The
University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Yoshikazu Tsukasaki
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
| | - Mark Maienschein-Cline
- Research
Resources Center, University of Illinois
at Chicago, Chicago, Illinois 60612, United States
| | - Balaji B. Ganesh
- Research
Resources Center, University of Illinois
at Chicago, Chicago, Illinois 60612, United States
| | - Prasad Kanteti
- Nano
Biotherapeutics, Inc., 2201 West Campbell Park Drive, Chicago, Illinois 60612, United States
| | - Jalees Rehman
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
- Division
of Cardiology, Department of Medicine, The
University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Asrar B. Malik
- Department
of Pharmacology and Regenerative Medicine and the Center for Lung
and Vascular Biology, The University of
Illinois College of Medicine, E403, 835 South Wolcott Avenue, Chicago, Illinois 60612, United States
- Nano
Biotherapeutics, Inc., 2201 West Campbell Park Drive, Chicago, Illinois 60612, United States
| |
Collapse
|
41
|
Necroptosis triggers spatially restricted neutrophil-mediated vascular damage during lung ischemia reperfusion injury. Proc Natl Acad Sci U S A 2022; 119:e2111537119. [PMID: 35238643 PMCID: PMC8917381 DOI: 10.1073/pnas.2111537119] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Intravital imaging, oxidative lipidomics, and a transplant model were used to define mechanisms that regulate neutrophil recruitment into lungs during ischemia reperfusion injury, a clinically relevant form of sterile inflammation. We found that early neutrophil-mediated damage is largely confined to the subpleural vasculature, a process that is orchestrated by a spatially restricted distribution of nonclassical monocytes that produce chemokines following necroptosis of pulmonary cells. Neutrophils disrupt the integrity of subpleural capillaries, which is associated with impaired lung function. Neutrophil-mediated vascular leakage is dependent on TLR4 expression on vascular endothelium, NOX4 signaling, and formation of neutrophil extracellular traps. Our research provides insights into mechanisms that regulate neutrophil recruitment during sterile lung inflammation and lays the foundation for developing new therapies. Ischemia reperfusion injury represents a common pathological condition that is triggered by the release of endogenous ligands. While neutrophils are known to play a critical role in its pathogenesis, the tissue-specific spatiotemporal regulation of ischemia-reperfusion injury is not understood. Here, using oxidative lipidomics and intravital imaging of transplanted mouse lungs that are subjected to severe ischemia reperfusion injury, we discovered that necroptosis, a nonapoptotic form of cell death, triggers the recruitment of neutrophils. During the initial stages of inflammation, neutrophils traffic predominantly to subpleural vessels, where their aggregation is directed by chemoattractants produced by nonclassical monocytes that are spatially restricted in this vascular compartment. Subsequent neutrophilic disruption of capillaries resulting in vascular leakage is associated with impaired graft function. We found that TLR4 signaling in vascular endothelial cells and downstream NADPH oxidase 4 expression mediate the arrest of neutrophils, a step upstream of their extravasation. Neutrophil extracellular traps formed in injured lungs and their disruption with DNase prevented vascular leakage and ameliorated primary graft dysfunction. Thus, we have uncovered mechanisms that regulate the initial recruitment of neutrophils to injured lungs, which result in selective damage to subpleural pulmonary vessels and primary graft dysfunction. Our findings could lead to the development of new therapeutics that protect lungs from ischemia reperfusion injury.
Collapse
|
42
|
Jacobelli J, Buser AE, Heiden DL, Friedman RS. Autoimmunity in motion: Mechanisms of immune regulation and destruction revealed by in vivo imaging. Immunol Rev 2022; 306:181-199. [PMID: 34825390 PMCID: PMC9135487 DOI: 10.1111/imr.13043] [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: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 11/30/2022]
Abstract
Autoimmunity arises when mechanisms of immune tolerance fail. Here we discuss mechanisms of T cell activation and tolerance and the dynamics of the autoimmune response at the site of disease. Live imaging of autoimmunity provides the ability to analyze immune cell dynamics at the single-cell level within the complex intact environment where disease occurs. These analyses have revealed mechanisms of T cell activation and tolerance in the lymph nodes, mechanisms of T cell entry into sites of autoimmune disease, and mechanisms leading to pathogenesis or protection in the autoimmune lesions. The overarching conclusions point to stable versus transient T cell antigen presenting cell interactions dictating the balance between T cell activation and tolerance, and T cell restimulation as a driver of pathogenesis at the site of autoimmunity. Findings from models of multiple sclerosis and type 1 diabetes are highlighted, however, the results have implications for basic mechanisms of T cell regulation during immune responses, tumor immunity, and autoimmunity.
Collapse
Affiliation(s)
- Jordan Jacobelli
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alan E. Buser
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Dustin L. Heiden
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Rachel S. Friedman
- Barbara Davis Center for Diabetes, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
43
|
Conrad C, Yildiz D, Cleary SJ, Margraf A, Cook L, Schlomann U, Panaretou B, Bowser JL, Karmouty-Quintana H, Li J, Berg NK, Martin SC, Aljohmani A, Moussavi-Harami SF, Wang KM, Tian JJ, Magnen M, Valet C, Qiu L, Singer JP, Eltzschig HK, Bertrams W, Herold S, Suttorp N, Schmeck B, Ball ZT, Zarbock A, Looney MR, Bartsch JW. ADAM8 signaling drives neutrophil migration and ARDS severity. JCI Insight 2022; 7:e149870. [PMID: 35132956 PMCID: PMC8855804 DOI: 10.1172/jci.insight.149870] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/21/2021] [Indexed: 01/27/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) results in catastrophic lung failure and has an urgent, unmet need for improved early recognition and therapeutic development. Neutrophil influx is a hallmark of ARDS and is associated with the release of tissue-destructive immune effectors, such as matrix metalloproteinases (MMPs) and membrane-anchored metalloproteinase disintegrins (ADAMs). Here, we observed using intravital microscopy that Adam8-/- mice had impaired neutrophil transmigration. In mouse pneumonia models, both genetic deletion and pharmacologic inhibition of ADAM8 attenuated neutrophil infiltration and lung injury while improving bacterial containment. Unexpectedly, the alterations of neutrophil function were not attributable to impaired proteolysis but resulted from reduced intracellular interactions of ADAM8 with the actin-based motor molecule Myosin1f that suppressed neutrophil motility. In 2 ARDS cohorts, we analyzed lung fluid proteolytic signatures and identified that ADAM8 activity was positively correlated with disease severity. We propose that in acute inflammatory lung diseases such as pneumonia and ARDS, ADAM8 inhibition might allow fine-tuning of neutrophil responses for therapeutic gain.
Collapse
Affiliation(s)
- Catharina Conrad
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Daniela Yildiz
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Simon J. Cleary
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Lena Cook
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Uwe Schlomann
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Barry Panaretou
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jessica L. Bowser
- Department of Pathology & Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Jiwen Li
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nathaniel K. Berg
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Ahmad Aljohmani
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - S. Farshid Moussavi-Harami
- Department of Pediatrics, Division of Pediatric Critical Care, University of California, San Francisco, San Francisco, California, USA
| | - Kristin M. Wang
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jennifer J. Tian
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Mélia Magnen
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Colin Valet
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Longhui Qiu
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jonathan P. Singer
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Wilhelm Bertrams
- Institute for Lung Research (iLung), Philipps-University, Marburg, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University Medical Center Giessen and Marburg, Giessen, Germany
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Schmeck
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
- Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Marburg, Germany
- German Center for Infectious Disease Research (DZIF), Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Zachary T. Ball
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Mark R. Looney
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jörg W. Bartsch
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| |
Collapse
|
44
|
Aroca-Crevillén A, Hidalgo A, Adrover JM. In Vivo Imaging of Circadian NET Formation During Lung Injury by Four-Dimensional Intravital Microscopy. Methods Mol Biol 2022; 2482:285-300. [PMID: 35610434 DOI: 10.1007/978-1-0716-2249-0_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Neutrophil extracellular traps (NETs) are toxic extracellular structures deployed by neutrophils in response to pathogens and sterile danger signals. NETs are circadian in nature as mouse and human neutrophils preferentially deploy them at night or early morning. Traditionally, NETs have been quantified using a plethora of methods including immunofluorescence and ELISA-based assays; however few options are available to visualize them in vivo. Here we describe a method to directly visualize and quantify NET formation and release in the microvasculature of the lung using intravital imaging in a model of acute lung injury. The method allows four-dimensional capture and quantification of NET formation dynamics over time and should be a useful resource for those interested in visualizing neutrophil responses in vivo.
Collapse
Affiliation(s)
- Alejandra Aroca-Crevillén
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Andres Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jose M Adrover
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
45
|
Kozlovski S, Regev O, Sapoznikov A, Kizner M, Achdout H, Petrovich-Kopitman E, Elkahal J, Addadi Y, Silva Castanheira FVE, Feigelson SW, Kubes P, Erez N, Garbi N, Alon R. ICAMs are dispensable for influenza clearance and anti-viral humoral and cellular immunity. Front Immunol 2022; 13:1041552. [PMID: 36895258 PMCID: PMC9988921 DOI: 10.3389/fimmu.2022.1041552] [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: 09/11/2022] [Accepted: 12/12/2022] [Indexed: 02/25/2023] Open
Abstract
αLβ2 (LFA-1) mediated interactions with ICAM-1 and ICAM-2 predominate leukocyte-vascular interactions, but their functions in extravascular cell-cell communications is still debated. The roles of these two ligands in leukocyte trafficking, lymphocyte differentiation, and immunity to influenza infections were dissected in the present study. Surprisingly, double ICAM-1 and ICAM-2 knock out mice (herein ICAM-1/2-/- mice) infected with a lab adapted H1N1 influenza A virus fully recovered from infection, elicited potent humoral immunity, and generated normal long lasting anti-viral CD8+ T cell memory. Furthermore, lung capillary ICAMs were dispensable for both NK and neutrophil entry to virus infected lungs. Mediastinal lymph nodes (MedLNs) of ICAM-1/2-/- mice poorly recruited naïve T cells and B lymphocytes but elicited normal humoral immunity critical for viral clearance and effective CD8+ differentiation into IFN-γ producing T cells. Furthermore, whereas reduced numbers of virus specific effector CD8+ T cells accumulated inside infected ICAM-1/2-/- lungs, normal virus-specific TRM CD8+ cells were generated inside these lungs and fully protected ICAM-1/2-/- mice from secondary heterosubtypic infections. B lymphocyte entry to the MedLNs and differentiation into extrafollicular plasmablasts, producing high affinity anti-influenza IgG2a antibodies, were also ICAM-1 and ICAM-2 independent. A potent antiviral humoral response was associated with accumulation of hyper-stimulated cDC2s in ICAM null MedLNs and higher numbers of virus-specific T follicular helper (Tfh) cells generated following lung infection. Mice selectively depleted of cDC ICAM-1 expression supported, however, normal CTL and Tfh differentiation following influenza infection, ruling out essential co-stimulatory functions of DC ICAM-1 in CD8+ and CD4+ T cell differentiation. Collectively our findings suggest that lung ICAMs are dispensable for innate leukocyte trafficking to influenza infected lungs, for the generation of peri-epithelial TRM CD8+ cells, and long term anti-viral cellular immunity. In lung draining LNs, although ICAMs promote lymphocyte homing, these key integrin ligands are not required for influenza-specific humoral immunity or generation of IFN-γ effector CD8+ T cells. In conclusion, our findings suggest unexpected compensatory mechanisms that orchestrate protective anti-influenza immunity in the absence of vascular and extravascular ICAMs.
Collapse
Affiliation(s)
- Stav Kozlovski
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofer Regev
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Marina Kizner
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Achdout
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | | | - Jacob Elkahal
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sara W Feigelson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Paul Kubes
- Department of Pharmacology and Physiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Noam Erez
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Natalio Garbi
- Department of Cellular Immunology, Institute of Experimental Immunology Medical Faculty, University of Bonn, Bonn, Germany
| | - Ronen Alon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
46
|
Tsukasaki Y, Toth PT, Davoodi-Bojd E, Rehman J, Malik AB. Quantitative Pulmonary Neutrophil Dynamics Using Computer-Vision Stabilized Intravital Imaging. Am J Respir Cell Mol Biol 2022; 66:12-22. [PMID: 34555309 PMCID: PMC8803365 DOI: 10.1165/rcmb.2021-0318ma] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/22/2021] [Indexed: 11/24/2022] Open
Abstract
In vivo intravital imaging in animal models in the lung remains challenging owing to respiratory motion artifacts. Here we describe a novel intravital imaging approach based on the computer-vision stabilization algorithm, Computer-Vision Stabilized Intravital Imaging. This method corrects lung movements and deformations at submicron precision in respiring mouse lungs. The precision enables high-throughput quantitative analysis of intravital pulmonary polymorphonuclear neutrophil (PMN) dynamics in lungs. We quantified real-time PMN patrolling dynamics of microvessels in the basal state and PMN recruitment resulting from sequestration in a model of endotoxemia in mice. We focused on determining the marginated pool of PMNs in the lung. Direct visualization of marginated PMNs revealed that they are not static but highly dynamic and undergo repeated cycles of "catch and release." PMNs briefly arrest in larger diameter capillary junction (∼10 μm) and then squeeze into narrower, approximately 5-μm diameter vessels through PMN deformation. We also observed that the sequestered PMNs in lung microvessels lost their migratory capabilities in association with cell morphological change following prolonged endotoxemia. These observations underscore the value of direct visualization and quantitative analysis of PMN dynamics in lungs to study PMN physiology and pathophysiology and role in inflammatory lung injury.
Collapse
Affiliation(s)
- Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology
| | - Peter T. Toth
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology
- Research Resources Center Fluorescence Imaging Core, and
| | - Esmaeil Davoodi-Bojd
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology
- Division of Cardiology, Department of Medicine, College of Medicine, the University of Illinois, Chicago, Illinois
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology
| |
Collapse
|
47
|
Moses ME, Hofmeyr S, Cannon JL, Andrews A, Gridley R, Hinga M, Leyba K, Pribisova A, Surjadidjaja V, Tasnim H, Forrest S. Spatially distributed infection increases viral load in a computational model of SARS-CoV-2 lung infection. PLoS Comput Biol 2021; 17:e1009735. [PMID: 34941862 PMCID: PMC8740970 DOI: 10.1371/journal.pcbi.1009735] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/07/2022] [Accepted: 12/09/2021] [Indexed: 01/03/2023] Open
Abstract
A key question in SARS-CoV-2 infection is why viral loads and patient outcomes vary dramatically across individuals. Because spatial-temporal dynamics of viral spread and immune response are challenging to study in vivo, we developed Spatial Immune Model of Coronavirus (SIMCoV), a scalable computational model that simulates hundreds of millions of lung cells, including respiratory epithelial cells and T cells. SIMCoV replicates viral growth dynamics observed in patients and shows how spatially dispersed infections can lead to increased viral loads. The model also shows how the timing and strength of the T cell response can affect viral persistence, oscillations, and control. By incorporating spatial interactions, SIMCoV provides a parsimonious explanation for the dramatically different viral load trajectories among patients by varying only the number of initial sites of infection and the magnitude and timing of the T cell immune response. When the branching airway structure of the lung is explicitly represented, we find that virus spreads faster than in a 2D layer of epithelial cells, but much more slowly than in an undifferentiated 3D grid or in a well-mixed differential equation model. These results illustrate how realistic, spatially explicit computational models can improve understanding of within-host dynamics of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Melanie E. Moses
- Department of Computer Science, University of New Mexico, Albuquerque, New Mexico, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | - Steven Hofmeyr
- Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Judy L. Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Akil Andrews
- Department of Computer Science, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Rebekah Gridley
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Monica Hinga
- Department of Computer Science, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Kirtus Leyba
- Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Abigail Pribisova
- Department of Computer Science, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Vanessa Surjadidjaja
- Department of Computer Science, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Humayra Tasnim
- Department of Computer Science, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Stephanie Forrest
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
- Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
48
|
Neupane AS, Kubes P. Imaging reveals novel innate immune responses in lung, liver, and beyond. Immunol Rev 2021; 306:244-257. [PMID: 34816440 DOI: 10.1111/imr.13040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022]
Abstract
Highly dynamic immune responses are generated toward pathogens or injuries, in vivo. Multiple immune cell types participate in various facets of the response which leads to a concerted effort in the removal and clearance of pathogens or injured tissue and a return to homeostasis. Intravital microscopy (IVM) has been extensively utilized to unravel the dynamics of immune responses, visualizing immune cell behavior in intact living tissues, within a living organism. For instance, the phenomenon of leukocyte recruitment cascade. Importantly, IVM has led to a deep appreciation that immune cell behavior and responses in individual organs are distinct, but also can influence one another. In this review, we discuss how IVM as a tool has been used to study the innate immune responses in various tissues during homeostasis, injury, and infection.
Collapse
Affiliation(s)
- Arpan Sharma Neupane
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.,Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.,Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, California, USA
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.,Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
49
|
Kim SJ, Carestia A, McDonald B, Zucoloto AZ, Grosjean H, Davis RP, Turk M, Naumenko V, Antoniak S, Mackman N, Abdul-Cader MS, Abdul-Careem MF, Hollenberg MD, Jenne CN. Platelet-Mediated NET Release Amplifies Coagulopathy and Drives Lung Pathology During Severe Influenza Infection. Front Immunol 2021; 12:772859. [PMID: 34858432 PMCID: PMC8632260 DOI: 10.3389/fimmu.2021.772859] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 01/30/2023] Open
Abstract
The influenza A virus (IAV) causes a respiratory tract infection with approximately 10% of the population infected by the virus each year. Severe IAV infection is characterized by excessive inflammation and tissue pathology in the lungs. Platelet and neutrophil recruitment to the lung are involved in the pathogenesis of IAV, but the specific mechanisms involved have not been clarified. Using confocal intravital microscopy in a mouse model of IAV infection, we observed profound neutrophil recruitment, platelet aggregation, neutrophil extracellular trap (NET) production and thrombin activation within the lung microvasculature in vivo. Importantly, deficiency or antagonism of the protease-activated receptor 4 (PAR4) reduced platelet aggregation, NET production, and neutrophil recruitment. Critically, inhibition of thrombin or PAR4 protected mice from virus-induced lung tissue damage and edema. Together, these data imply thrombin-stimulated platelets play a critical role in the activation/recruitment of neutrophils, NET release and directly contribute to IAV pathogenesis in the lung.
Collapse
MESH Headings
- Animals
- Blood Coagulation Disorders/immunology
- Blood Coagulation Disorders/metabolism
- Blood Coagulation Disorders/virology
- Blood Platelets/immunology
- Blood Platelets/metabolism
- Blood Platelets/virology
- Disease Models, Animal
- Extracellular Traps/immunology
- Extracellular Traps/metabolism
- Extracellular Traps/virology
- Female
- Humans
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza, Human/immunology
- Influenza, Human/metabolism
- Influenza, Human/virology
- Lung/immunology
- Lung/metabolism
- Lung/virology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Neutrophil Infiltration/immunology
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/virology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/metabolism
- Orthomyxoviridae Infections/virology
- Platelet Aggregation/immunology
- Mice
Collapse
Affiliation(s)
- Seok-Joo Kim
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Agostina Carestia
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, University of Calgary, Calgary, AB, Canada
| | - Amanda Z. Zucoloto
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Heidi Grosjean
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Rachelle P. Davis
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Madison Turk
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Victor Naumenko
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Silvio Antoniak
- UNC Blood Research Center, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nigel Mackman
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - Morley D. Hollenberg
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Craig N. Jenne
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
50
|
Endothelial Heparan Sulfate Mediates Hepatic Neutrophil Trafficking and Injury during Staphylococcus aureus Sepsis. mBio 2021; 12:e0118121. [PMID: 34544271 PMCID: PMC8546592 DOI: 10.1128/mbio.01181-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatic failure is an important risk factor for poor outcome in septic patients. Using a chemical tagging workflow and high-resolution mass spectrometry, we demonstrate that rapid proteome remodeling of the vascular surfaces precedes hepatic damage in a murine model of Staphylococcus aureus sepsis. These early changes include vascular deposition of neutrophil-derived proteins, shedding of vascular receptors, and altered levels of heparin/heparan sulfate-binding factors. Modification of endothelial heparan sulfate, a major component of the vascular glycocalyx, diminishes neutrophil trafficking to the liver and reduces hepatic coagulopathy and organ damage during the systemic inflammatory response to infection. Modifying endothelial heparan sulfate likewise reduces neutrophil trafficking in sterile hepatic injury, reflecting a more general role of heparan sulfate contribution to the modulation of leukocyte behavior during inflammation. IMPORTANCE Vascular glycocalyx remodeling is critical to sepsis pathology, but the glycocalyx components that contribute to this process remain poorly characterized. This article shows that during Staphylococcus aureus sepsis, the liver vascular glycocalyx undergoes dramatic changes in protein composition associated with neutrophilic activity and heparin/heparan sulfate binding, all before organ damage is detectable by standard circulating liver damage markers or histology. Targeted manipulation of endothelial heparan sulfate modulates S. aureus sepsis-induced hepatotoxicity by controlling the magnitude of neutrophilic infiltration into the liver in both nonsterile and sterile injury. These data identify an important vascular glycocalyx component that impacts hepatic failure during nonsterile and sterile injury.
Collapse
|