1
|
Nitzsche B, Rong WW, Goede A, Hoffmann B, Scarpa F, Kuebler WM, Secomb TW, Pries AR. Coalescent angiogenesis-evidence for a novel concept of vascular network maturation. Angiogenesis 2021; 25:35-45. [PMID: 34905124 PMCID: PMC8669669 DOI: 10.1007/s10456-021-09824-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/07/2021] [Indexed: 02/06/2023]
Abstract
Angiogenesis describes the formation of new blood vessels from pre-existing vascular structures. While the most studied mode of angiogenesis is vascular sprouting, specific conditions or organs favor intussusception, i.e., the division or splitting of an existing vessel, as preferential mode of new vessel formation. In the present study, sustained (33-h) intravital microscopy of the vasculature in the chick chorioallantoic membrane (CAM) led to the hypothesis of a novel non-sprouting mode for vessel generation, which we termed "coalescent angiogenesis." In this process, preferential flow pathways evolve from isotropic capillary meshes enclosing tissue islands. These preferential flow pathways progressively enlarge by coalescence of capillaries and elimination of internal tissue pillars, in a process that is the reverse of intussusception. Concomitantly, less perfused segments regress. In this way, an initially mesh-like capillary network is remodeled into a tree structure, while conserving vascular wall components and maintaining blood flow. Coalescent angiogenesis, thus, describes the remodeling of an initial, hemodynamically inefficient mesh structure, into a hierarchical tree structure that provides efficient convective transport, allowing for the rapid expansion of the vasculature with maintained blood supply and function during development.
Collapse
Affiliation(s)
- Bianca Nitzsche
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, 10117, Berlin, Germany
| | - Wen Wei Rong
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany
| | - Andrean Goede
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany
| | - Björn Hoffmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany
| | - Fabio Scarpa
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Wolfgang M Kuebler
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, 10117, Berlin, Germany
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, AZ, 85724, USA
| | - Axel R Pries
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany. .,German Center for Cardiovascular Research (DZHK), Partner site Berlin, 10117, Berlin, Germany.
| |
Collapse
|
2
|
Alizadeh-Tabrizi N, Hall S, Lehmann C. Intravital Imaging of Pulmonary Immune Response in Inflammation and Infection. Front Cell Dev Biol 2021; 8:620471. [PMID: 33520993 PMCID: PMC7843704 DOI: 10.3389/fcell.2020.620471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/18/2020] [Indexed: 12/29/2022] Open
Abstract
Intravital microscopy (IVM) is a unique imaging method providing insights in cellular functions and interactions in real-time, without the need for tissue extraction from the body. IVM of the lungs has specific challenges such as restricted organ accessibility, respiratory movements, and limited penetration depth. Various surgical approaches and microscopic setups have been adapted in order to overcome these challenges. Among others, these include the development of suction stabilized lung windows and the use of more advanced optical techniques. Consequently, lung IVM has uncovered mechanisms of leukocyte recruitment and function in several models of pulmonary inflammation and infection. This review focuses on bacterial pneumonia, aspiration pneumonia, sepsis-induced acute lung Injury, and cystic fibrosis, as examples of lung inflammation and infection. In addition, critical details of intravital imaging techniques of the lungs are discussed.
Collapse
Affiliation(s)
| | - Stefan Hall
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Christian Lehmann
- Department of Physiology & Biophysics, Dalhousie University, Halifax, NS, Canada.,Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
3
|
|
4
|
Aizad S, Zubairi SI, Yahaya BH, Lazim AM. Centella asiatica Extract Potentiates Anticancer Activity in an Improved 3-D PHBV-Composite-CMC A549 Lung Cancer Microenvironment Scaffold. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2020. [DOI: 10.1007/s13369-020-05072-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
5
|
Wang N, Cao H, Wang L, Ren F, Zeng Q, Xu X, Liang J, Zhan Y, Chen X. Recent Advances in Spontaneous Raman Spectroscopic Imaging: Instrumentation and Applications. Curr Med Chem 2019; 27:6188-6207. [PMID: 31237196 DOI: 10.2174/0929867326666190619114431] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Spectroscopic imaging based on the spontaneous Raman scattering effects can provide unique fingerprint information in relation to the vibration bands of molecules. Due to its advantages of high chemical specificity, non-invasive detection capability, low sensitivity to water, and no special sample pretreatment, Raman Spectroscopic Imaging (RSI) has become an invaluable tool in the field of biomedicine and medicinal chemistry. METHODS There are three methods to implement RSI, including point scanning, line scanning and wide-field RSI. Point-scanning can achieve two-and three-dimensional imaging of target samples. High spectral resolution, full spectral range and confocal features render this technique highly attractive. However, point scanning based RSI is a time-consuming process that can take several hours to map a small area. Line scanning RSI is an extension of point scanning method, with an imaging speed being 300-600 times faster. In the wide-field RSI, the laser illuminates the entire region of interest directly and all the images then collected for analysis. In general, it enables more accurate chemical imaging at faster speeds. RESULTS This review focuses on the recent advances in RSI, with particular emphasis on the latest developments on instrumentation and the related applications in biomedicine and medicinal chemistry. Finally, we prospect the development trend of RSI as well as its potential to translation from bench to bedside. CONCLUSION RSI is a powerful technique that provides unique chemical information, with a great potential in the fields of biomedicine and medicinal chemistry.
Collapse
Affiliation(s)
- Nan Wang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| | - Honghao Cao
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| | - Lin Wang
- School of Information Sciences and Techonlogy, Northwest University, Xi’an, Shaanxi 710127, China
| | - Feng Ren
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| | - Qi Zeng
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| | - Xinyi Xu
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| | - Jimin Liang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| | - Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of China, Xi’an, Shaanxi 710126, China,School of Life Science and Technology, Xidian University, P.O. Box: 0528, Xi’an, Shaanxi 710126, China
| |
Collapse
|
6
|
Regelin N, Heyder S, Laschke MW, Hadizamani Y, Borgmann M, Moehrlen U, Schramm R, Bals R, Menger MD, Hamacher J. A murine model to study vasoreactivity and intravascular flow in lung isograft microvessels. Sci Rep 2019; 9:5170. [PMID: 30914786 PMCID: PMC6435642 DOI: 10.1038/s41598-019-41590-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/20/2019] [Indexed: 11/09/2022] Open
Abstract
Intravital microscopy of orthotopic lung tissue is technically demanding, especially for repeated investigations. Therefore, we have established a novel approach, which allows non-invasive repetitive in vivo microscopy of ectopic lung tissue in dorsal skinfold chambers. Syngeneic subpleural peripheral lung tissue and autologous endometrium (control) were transplanted onto the striated muscle within dorsal skinfold chambers of C57BL/6 mice. Grafts were analysed by intravital fluorescence microscopy over 14 days. Angiogenesis occurred in the grafts on day 3, as indicated by sinusoidal microvessels on the grafts’ edges with very slow blood flow, perifocal oedema, and haemorrhage. By day 10, lung transplants were completely revascularized, exhibited a dense network of microvessels with irregular diameters, chaotic angioarchitecture, and high blood flow. Compared to lung tissue, endometrial grafts contained a structured, glomerulus-like vessel architecture with lower blood flow. Despite missing ventilation, hypoxic vasoconstriction of the lung tissue arterioles occurred. In contrast, endometrium tissue arterioles dilated during hypoxia and constricted in hyperoxia. This demonstrates that ectopic lung grafts keep their ability for organ-specific hypoxic vasoconstriction. These findings indicate that our approach is suitable for repetitive in vivo pulmonary microcirculation analyses. The high blood flow and hypoxia-induced vasoconstriction in lung grafts suggest a physiological intrinsic vasoregulation independent of the recipient tissue.
Collapse
Affiliation(s)
- Nora Regelin
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, Saarland University Hospital, 66424, Homburg, Germany.,Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany
| | - Susanne Heyder
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, Saarland University Hospital, 66424, Homburg, Germany.,Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany.,Mediclin Albert Schweitzer Clinic, Pneumology, 78126, Königsfeld, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany
| | - Yalda Hadizamani
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012, Bern, Switzerland.,Lungen-und Atmungsstiftung, Bern, 3012, Bern, Switzerland
| | - Michèle Borgmann
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012, Bern, Switzerland.,Lungen-und Atmungsstiftung, Bern, 3012, Bern, Switzerland
| | - Ueli Moehrlen
- Lungen-und Atmungsstiftung, Bern, 3012, Bern, Switzerland.,Pediatric Surgery, University Children's Hospital Zurich, 8032, Zurich, Switzerland
| | - René Schramm
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany.,Heart and Diabetes Centre North Rhine-Westphalia, University Hospital of the Ruhr University of Bochum, 32545, Bad Oeynhausen, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, Saarland University Hospital, 66424, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany
| | - Jürg Hamacher
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, Saarland University Hospital, 66424, Homburg, Germany. .,Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, 66421, Homburg, Germany. .,Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012, Bern, Switzerland. .,Lungen-und Atmungsstiftung, Bern, 3012, Bern, Switzerland.
| |
Collapse
|
7
|
Wagner WW, Jaryszak EM, Peterson AJ, Doerschuk CM, Bohlen HG, King JAC, Tanner JA, Crockett ES, Glenny RW, Presson RG. A perpetual switching system in pulmonary capillaries. J Appl Physiol (1985) 2019; 126:494-501. [PMID: 30571293 PMCID: PMC6397411 DOI: 10.1152/japplphysiol.00507.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022] Open
Abstract
Of the 300 billion capillaries in the human lung, a small fraction meet normal oxygen requirements at rest, with the remainder forming a large reserve. The maximum oxygen demands of the acute stress response require that the reserve capillaries are rapidly recruited. To remain primed for emergencies, the normal cardiac output must be parceled throughout the capillary bed to maintain low opening pressures. The flow-distributing system requires complex switching. Because the pulmonary microcirculation contains contractile machinery, one hypothesis posits an active switching system. The opposing hypothesis is based on passive switching that requires no regulation. Both hypotheses were tested ex vivo in canine lung lobes. The lobes were perfused first with autologous blood, and capillary switching patterns were recorded by videomicroscopy. Next, the vasculature of the lobes was saline flushed, fixed by glutaraldehyde perfusion, flushed again, and then reperfused with the original, unfixed blood. Flow patterns through the same capillaries were recorded again. The 16-min-long videos were divided into 4-s increments. Each capillary segment was recorded as being perfused if at least one red blood cell crossed the entire segment. Otherwise it was recorded as unperfused. These binary measurements were made manually for each segment during every 4 s throughout the 16-min recordings of the fresh and fixed capillaries (>60,000 measurements). Unexpectedly, the switching patterns did not change after fixation. We conclude that the pulmonary capillaries can remain primed for emergencies without requiring regulation: no detectors, no feedback loops, and no effectors-a rare system in biology. NEW & NOTEWORTHY The fluctuating flow patterns of red blood cells within the pulmonary capillary networks have been assumed to be actively controlled within the pulmonary microcirculation. Here we show that the capillary flow switching patterns in the same network are the same whether the lungs are fresh or fixed. This unexpected observation can be successfully explained by a new model of pulmonary capillary flow based on chaos theory and fractal mathematics.
Collapse
Affiliation(s)
- Wiltz W Wagner
- Department of Anesthesiology, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Molecular and Cellular Pharmacology, Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama , Mobile, Alabama
| | - Eric M Jaryszak
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Amanda J Peterson
- Department of Anesthesiology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Claire M Doerschuk
- Center for Airways Disease, Department of Medicine, University of North Carolina , Chapel Hill, North Carolina
| | - H Glenn Bohlen
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Judy A C King
- Department of Molecular and Cellular Pharmacology, Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama , Mobile, Alabama
| | - Judith A Tanner
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Edward S Crockett
- Department of Molecular and Cellular Pharmacology, Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama , Mobile, Alabama
| | - Robb W Glenny
- Departments of Medicine and of Physiology and Biophysics, University of Washington , Seattle, Washington
| | - Robert G Presson
- Department of Anesthesiology, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
8
|
Pinar IP, Jones HD. Novel imaging approaches for small animal models of lung disease (2017 Grover Conference series). Pulm Circ 2018; 8:2045894018762242. [PMID: 29480066 PMCID: PMC5888832 DOI: 10.1177/2045894018762242] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Imaging in small animal models of lung disease is challenging, as existing technologies are limited either by resolution or by the terminal nature of the imaging approach. Here, we describe the current state of small animal lung imaging, the technological advances of laboratory-sourced phase contrast X-ray imaging, and the application of this novel technology and its attendant image analysis techniques to the in vivo imaging of the large airways and pulmonary vasculature in murine models of lung health and disease.
Collapse
Affiliation(s)
- Isaac P Pinar
- 1 Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, VIC, Australia.,2 Division of Biological Engineering, Faculty of Engineering, Monash University, Melbourne, VIC, Australia
| | - Heather D Jones
- 3 Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
9
|
Abstract
While reductionist in vitro approaches have allowed for careful interrogation of cellular pathways that underlie innate immune responses, they often fail to capture the complex multicellular interactions characteristic of acute inflammation. Intravital microscopy, by directly observing alveolar cell-cell interactions, provides unique insight into the complex intercellular mechanisms responsible for alveolar inflammation. This review discusses multiple potential approaches to intravital pulmonary imaging, with specific attention to in vivo microscopy of the freely moving mouse lung.
Collapse
|
10
|
Osgood RS, Upham BL, Bushel PR, Velmurugan K, Xiong KN, Bauer AK. Secondhand Smoke-Prevalent Polycyclic Aromatic Hydrocarbon Binary Mixture-Induced Specific Mitogenic and Pro-inflammatory Cell Signaling Events in Lung Epithelial Cells. Toxicol Sci 2017; 157:156-171. [PMID: 28329830 PMCID: PMC5808746 DOI: 10.1093/toxsci/kfx027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Low molecular weight polycyclic aromatic hydrocarbons (LMW PAHs; < 206.3 g/mol) are prevalent and ubiquitous environmental contaminants, presenting a human health concern, and have not been as thoroughly studied as the high MW PAHs. LMW PAHs exert their pulmonary effects, in part, through P38-dependent and -independent mechanisms involving cell-cell communication and the production of pro-inflammatory mediators known to contribute to lung disease. Specifically, we determined the effects of two representative LMW PAHs, 1-methylanthracene (1-MeA) and fluoranthene (Flthn), individually and as a binary PAH mixture on the dysregulation of gap junctional intercellular communication (GJIC) and connexin 43 (Cx43), activation of mitogen activated protein kinases (MAPK), and induction of inflammatory mediators in a mouse non-tumorigenic alveolar type II cell line (C10). Both 1-MeA, Flthn, and the binary PAH mixture of 1-MeA and Flthn dysregulated GJIC in a dose and time-dependent manner, reduced Cx43 protein, and activated the following MAPKs: P38, ERK1/2, and JNK. Inhibition of P38 MAPK prevented PAH-induced dysregulation of GJIC, whereas inhibiting ERK and JNK did not prevent these PAHs from dysregulating GJIC indicating a P38-dependent mechanism. A toxicogenomic approach revealed significant P38-dependent and -independent pathways involved in inflammation, steroid synthesis, metabolism, and oxidative responses. Genes in these pathways were significantly altered by the binary PAH mixture when compared with 1-MeA and Flthn alone suggesting interactive effects. Exposure to the binary PAH mixture induced the production and release of cytokines and metalloproteinases from the C10 cells. Our findings with a binary mixture of PAHs suggest that combinations of LMW PAHs may elicit synergistic or additive inflammatory responses which warrant further investigation and confirmation.
Collapse
Affiliation(s)
- Ross S. Osgood
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Brad L. Upham
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, Michigan 48824
| | - Pierre R. Bushel
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Kalpana Velmurugan
- Department of Environmental and Occupational Health, University of Colorado Anschutz Medical Center, Aurora, Colorado 80045
| | - Ka-Na Xiong
- Department of Environmental and Occupational Health, University of Colorado Anschutz Medical Center, Aurora, Colorado 80045
| | - Alison K. Bauer
- Department of Environmental and Occupational Health, University of Colorado Anschutz Medical Center, Aurora, Colorado 80045
| |
Collapse
|
11
|
Furuya K, Tan JJ, Boudreault F, Sokabe M, Berthiaume Y, Grygorczyk R. Real-time imaging of inflation-induced ATP release in the ex vivo rat lung. Am J Physiol Lung Cell Mol Physiol 2016; 311:L956-L969. [PMID: 27638905 DOI: 10.1152/ajplung.00425.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 09/13/2016] [Indexed: 12/25/2022] Open
Abstract
Extracellular ATP and other nucleotides are important autocrine/paracrine mediators that regulate diverse processes critical for lung function, including mucociliary clearance, surfactant secretion, and local blood flow. Cellular ATP release is mechanosensitive; however, the impact of physical stimuli on ATP release during breathing has never been tested in intact lungs in real time and remains elusive. In this pilot study, we investigated inflation-induced ATP release in rat lungs ex vivo by real-time luciferin-luciferase (LL) bioluminescence imaging coupled with simultaneous infrared tissue imaging to identify ATP-releasing sites. With LL solution introduced into air spaces, brief inflation of such edematous lung (1 s, ∼20 cmH2O) induced transient (<30 s) ATP release in a limited number of air-inflated alveolar sacs during their recruitment/opening. Released ATP reached concentrations of ∼10-6 M, relevant for autocrine/paracrine signaling, but it remained spatially restricted to single alveolar sacs or their clusters. ATP release was stimulus dependent: prolonged (100 s) inflation evoked long-lasting ATP release that terminated upon alveoli deflation/derecruitment while cyclic inflation/suction produced cyclic ATP release. With LL introduced into blood vessels, inflation induced transient ATP release in many small patchlike areas the size of alveolar sacs. Findings suggest that inflation induces ATP release in both alveoli and the surrounding blood capillary network; the functional units of ATP release presumably consist of alveolar sacs or their clusters. Our study demonstrates the feasibility of real-time ATP release imaging in ex vivo lungs and provides the first direct evidence of inflation-induced ATP release in lung air spaces and in pulmonary blood capillaries, highlighting the importance of purinergic signaling in lung function.
Collapse
Affiliation(s)
- Kishio Furuya
- Mechanobiology Laboratory, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Ju Jing Tan
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Francis Boudreault
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yves Berthiaume
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; and.,Institut de recherches cliniques de Montréal (IRCM), Quebec, Canada
| | - Ryszard Grygorczyk
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; .,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; and
| |
Collapse
|
12
|
Fiole D, Tournier JN. Intravital microscopy of the lung: minimizing invasiveness. JOURNAL OF BIOPHOTONICS 2016; 9:868-878. [PMID: 26846880 DOI: 10.1002/jbio.201500246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/08/2016] [Accepted: 01/09/2016] [Indexed: 06/05/2023]
Abstract
In vivo microscopy has recently become a gold standard in lung immunology studies involving small animals, largely benefiting from the democratization of multiphoton microscopy allowing for deep tissue imaging. This technology represents currently our only way of exploring the lungs and inferring what happens in human respiratory medicine. The interest of lung in vivo microscopy essentially relies upon its relevance as a study model, fulfilling physiological requirements in comparison with in vitro and ex vivo experiments. However, strategies developed in order to overcome movements of the thorax caused by breathing and heartbeats remain the chief drawback of the technique and a major source of invasiveness. In this context, minimizing invasiveness is an unavoidable prerequisite for any improvement of lung in vivo microscopy. This review puts into perspective the main techniques enabling lung in vivo microscopy, providing pros and cons regarding invasiveness.
Collapse
Affiliation(s)
- Daniel Fiole
- Unité Interactions Hôte-Agents pathogènes, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge cedex, 91223, France.
- Human Histopathology and Animal Models, Institut Pasteur, 28 rue du docteur Roux, Paris, 75725, France.
| | - Jean-Nicolas Tournier
- Unité Interactions Hôte-Agents pathogènes, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge cedex, 91223, France
- Laboratoire Pathogénie des Toxi-Infections Bactériennes, Institut Pasteur, 28 rue du docteur Roux, Paris, 75725, France
- Ecole du Val-de-Grâce, 1 place Alphonse Laveran, Paris, 75230, France
| |
Collapse
|
13
|
Kuebler WM, Wittenberg C, Lee WL, Reppien E, Goldenberg NM, Lindner K, Gao Y, Winoto-Morbach S, Drab M, Mühlfeld C, Dombrowsky H, Ochs M, Schütze S, Uhlig S. Thrombin stimulates albumin transcytosis in lung microvascular endothelial cells via activation of acid sphingomyelinase. Am J Physiol Lung Cell Mol Physiol 2016; 310:L720-32. [PMID: 26851257 DOI: 10.1152/ajplung.00157.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 01/22/2016] [Indexed: 01/12/2023] Open
Abstract
Transcellular albumin transport occurs via caveolae that are abundant in lung microvascular endothelial cells. Stimulation of albumin transcytosis by proinflammatory mediators may contribute to alveolar protein leak in lung injury, yet the regulation of albumin transport and its underlying molecular mechanisms are so far incompletely understood. Here we tested the hypothesis that thrombin may stimulate transcellular albumin transport across lung microvascular endothelial cells in an acid-sphingomyelinase dependent manner. Thrombin increased the transport of fluorescently labeled albumin across confluent human lung microvascular endothelial cell (HMVEC-L) monolayers to an extent that markedly exceeds the rate of passive diffusion. Thrombin activated acid sphingomyelinase (ASM) and increased ceramide production in HMVEC-L, but not in bovine pulmonary artery cells, which showed little albumin transport in response to thrombin. Thrombin increased total caveolin-1 (cav-1) content in both whole cell lysates and lipid rafts from HMVEC-L, and this effect was blocked by inhibition of ASM or de novo protein biosynthesis. Thrombin-induced uptake of albumin into lung microvascular endothelial cells was confirmed in isolated-perfused lungs by real-time fluorescence imaging and electron microscopy of gold-labeled albumin. Inhibition of ASM attenuated thrombin-induced albumin transport both in confluent HMVEC-L and in intact lungs, whereas HMVEC-L treatment with exogenous ASM increased albumin transport and enriched lipid rafts in cav-1. Our findings indicate that thrombin stimulates transcellular albumin transport in an acid sphingomyelinase-dependent manner by inducing de novo synthesis of cav-1 and its recruitment to membrane lipid rafts.
Collapse
Affiliation(s)
- Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Heart Institute Berlin, Berlin, Germany; The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Claudia Wittenberg
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Warren L Lee
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care, Departments of Medicine and Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Eike Reppien
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Neil M Goldenberg
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada
| | - Karsten Lindner
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Yizhuo Gao
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada
| | | | - Marek Drab
- Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DLZ), Hannover, Germany; Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany; and
| | - Heike Dombrowsky
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DLZ), Hannover, Germany; Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany; and
| | - Stefan Schütze
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Stefan Uhlig
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany; Institute of Pharmacology and Toxicology, RWTH Aachen, Aachen, Germany
| |
Collapse
|
14
|
Patel BV, Tatham KC, Wilson MR, O'Dea KP, Takata M. In vivo compartmental analysis of leukocytes in mouse lungs. Am J Physiol Lung Cell Mol Physiol 2015; 309:L639-52. [PMID: 26254421 PMCID: PMC4593833 DOI: 10.1152/ajplung.00140.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/04/2015] [Indexed: 12/12/2022] Open
Abstract
The lung has a unique structure consisting of three functionally different compartments (alveolar, interstitial, and vascular) situated in an extreme proximity. Current methods to localize lung leukocytes using bronchoalveolar lavage and/or lung perfusion have significant limitations for determination of location and phenotype of leukocytes. Here we present a novel method using in vivo antibody labeling to enable accurate compartmental localization/quantification and phenotyping of mouse lung leukocytes. Anesthetized C57BL/6 mice received combined in vivo intravenous and intratracheal labeling with fluorophore-conjugated anti-CD45 antibodies, and lung single-cell suspensions were analyzed by flow cytometry. The combined in vivo intravenous and intratracheal CD45 labeling enabled robust separation of the alveolar, interstitial, and vascular compartments of the lung. In naive mice, the alveolar compartment consisted predominantly of resident alveolar macrophages. The interstitial compartment, gated by events negative for both intratracheal and intravenous CD45 staining, showed two conventional dendritic cell populations, as well as a Ly6Clo monocyte population. Expression levels of MHCII on these interstitial monocytes were much higher than on the vascular Ly6Clo monocyte populations. In mice exposed to acid aspiration-induced lung injury, this protocol also clearly distinguished the three lung compartments showing the dynamic trafficking of neutrophils and exudative monocytes across the lung compartments during inflammation and resolution. This simple in vivo dual-labeling technique substantially increases the accuracy and depth of lung flow cytometric analysis, facilitates a more comprehensive examination of lung leukocyte pools, and enables the investigation of previously poorly defined “interstitial” leukocyte populations during models of inflammatory lung diseases.
Collapse
Affiliation(s)
- Brijesh V Patel
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Kate C Tatham
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Michael R Wilson
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Kieran P O'Dea
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Masao Takata
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| |
Collapse
|
15
|
Burgstaller G, Vierkotten S, Lindner M, Königshoff M, Eickelberg O. Multidimensional immunolabeling and 4D time-lapse imaging of vital ex vivo lung tissue. Am J Physiol Lung Cell Mol Physiol 2015; 309:L323-32. [PMID: 26092995 DOI: 10.1152/ajplung.00061.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/10/2015] [Indexed: 11/22/2022] Open
Abstract
During the last decades, the study of cell behavior was largely accomplished in uncoated or extracellular matrix (ECM)-coated plastic dishes. To date, considerable cell biological efforts have tried to model in vitro the natural microenvironment found in vivo. For the lung, explants cultured ex vivo as lung tissue cultures (LTCs) provide a three-dimensional (3D) tissue model containing all cells in their natural microenvironment. Techniques for assessing the dynamic live interaction between ECM and cellular tissue components, however, are still missing. Here, we describe specific multidimensional immunolabeling of living 3D-LTCs, derived from healthy and fibrotic mouse lungs, as well as patient-derived 3D-LTCs, and concomitant real-time four-dimensional multichannel imaging thereof. This approach allowed the evaluation of dynamic interactions between mesenchymal cells and macrophages with their ECM. Furthermore, fibroblasts transiently expressing focal adhesions markers incorporated into the 3D-LTCs, paving new ways for studying the dynamic interaction between cellular adhesions and their natural-derived ECM. A novel protein transfer technology (FuseIt/Ibidi) shuttled fluorescently labeled α-smooth muscle actin antibodies into the native cells of living 3D-LTCs, enabling live monitoring of α-smooth muscle actin-positive stress fibers in native tissue myofibroblasts residing in fibrotic lesions of 3D-LTCs. Finally, this technique can be applied to healthy and diseased human lung tissue, as well as to adherent cells in conventional two-dimensional cell culture. This novel method will provide valuable new insights into the dynamics of ECM (patho)biology, studying in detail the interaction between ECM and cellular tissue components in their natural microenvironment.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| | - Sarah Vierkotten
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| | - Michael Lindner
- Center for Thoracic Surgery, Asklepios Biobank for Lung Diseases, Comprehensive Pneumology Center, Asklepios Clinic Munich-Gauting, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| |
Collapse
|
16
|
McLaughlin RA, Noble PB, Sampson DD. Optical coherence tomography in respiratory science and medicine: from airways to alveoli. Physiology (Bethesda) 2015; 29:369-80. [PMID: 25180266 DOI: 10.1152/physiol.00002.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Optical coherence tomography is a rapidly maturing optical imaging technology, enabling study of the in vivo structure of lung tissue at a scale of tens of micrometers. It has been used to assess the layered structure of airway walls, quantify both airway lumen caliber and compliance, and image individual alveoli. This article provides an overview of the technology and reviews its capability to provide new insights into respiratory disease.
Collapse
Affiliation(s)
- Robert A McLaughlin
- Optical & Biomedical Engineering Laboratory, School of Electrical, Electronic & Computer Engineering, The University of Western Australia, Perth, Australia;
| | - Peter B Noble
- School of Anatomy, Physiology & Human Biology, and Centre for Neonatal Research & Education, School of Paediatrics and Child Health, The University of Western Australia, Crawley, Australia; and
| | - David D Sampson
- Optical & Biomedical Engineering Laboratory, School of Electrical, Electronic & Computer Engineering, The University of Western Australia, Perth, Australia; Centre for Microscopy, Characterisation & Analysis, The University of Western Australia, Perth, Australia
| |
Collapse
|
17
|
Scolaro L, Lorenser D, Madore WJ, Kirk RW, Kramer AS, Yeoh GC, Godbout N, Sampson DD, Boudoux C, McLaughlin RA. Molecular imaging needles: dual-modality optical coherence tomography and fluorescence imaging of labeled antibodies deep in tissue. BIOMEDICAL OPTICS EXPRESS 2015; 6:1767-81. [PMID: 26137379 PMCID: PMC4467702 DOI: 10.1364/boe.6.001767] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/06/2015] [Accepted: 04/14/2015] [Indexed: 05/04/2023]
Abstract
Molecular imaging using optical techniques provides insight into disease at the cellular level. In this paper, we report on a novel dual-modality probe capable of performing molecular imaging by combining simultaneous three-dimensional optical coherence tomography (OCT) and two-dimensional fluorescence imaging in a hypodermic needle. The probe, referred to as a molecular imaging (MI) needle, may be inserted tens of millimeters into tissue. The MI needle utilizes double-clad fiber to carry both imaging modalities, and is interfaced to a 1310-nm OCT system and a fluorescence imaging subsystem using an asymmetrical double-clad fiber coupler customized to achieve high fluorescence collection efficiency. We present, to the best of our knowledge, the first dual-modality OCT and fluorescence needle probe with sufficient sensitivity to image fluorescently labeled antibodies. Such probes enable high-resolution molecular imaging deep within tissue.
Collapse
Affiliation(s)
- Loretta Scolaro
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic, & Computer Engineering, The University of Western Australia, Crawley, Australia
| | - Dirk Lorenser
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic, & Computer Engineering, The University of Western Australia, Crawley, Australia
| | - Wendy-Julie Madore
- Centre d'optique, photonique et lasers, Department of Engineering Physics, Polytechnique Montréal, Montréal (QC), Canada
| | - Rodney W. Kirk
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic, & Computer Engineering, The University of Western Australia, Crawley, Australia
| | - Anne S. Kramer
- Centre for Medical Research, The Harry Perkins Institute of Medical Research and School of Chemistry & Biochemistry, The University of Western Australia, Crawley, Australia
| | - George C. Yeoh
- Centre for Medical Research, The Harry Perkins Institute of Medical Research and School of Chemistry & Biochemistry, The University of Western Australia, Crawley, Australia
| | - Nicolas Godbout
- Centre d'optique, photonique et lasers, Department of Engineering Physics, Polytechnique Montréal, Montréal (QC), Canada
| | - David D. Sampson
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic, & Computer Engineering, The University of Western Australia, Crawley, Australia
- Centre for Microscopy, Characterisation & Analysis, The University of Western Australia, Crawley, Australia
| | - Caroline Boudoux
- Centre d'optique, photonique et lasers, Department of Engineering Physics, Polytechnique Montréal, Montréal (QC), Canada
| | - Robert A. McLaughlin
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic, & Computer Engineering, The University of Western Australia, Crawley, Australia
| |
Collapse
|
18
|
Teichert-Kuliszewska K, Tsoporis JN, Desjardins JF, Yin J, Wang L, Kuebler WM, Parker TG. Absence of the calcium-binding protein, S100A1, confers pulmonary hypertension in mice associated with endothelial dysfunction and apoptosis. Cardiovasc Res 2014; 105:8-19. [PMID: 25395393 DOI: 10.1093/cvr/cvu241] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS S100A1, a 10-kDa, Ca(2+)-binding protein, is expressed in endothelial cells (ECs) and binds eNOS. Its absence is associated with impaired production of nitric oxide (NO) and mild systemic hypertension. As endothelial dysfunction contributes to clinical and experimental pulmonary hypertension (PH), we investigated the impact of deleting S100A1 in mice, on pulmonary haemodynamics, endothelial function, NO production, associated signalling pathways, and apoptosis. METHODS AND RESULTS Compared with wild-type (WT), S100A1-knock-out mice (KO) exhibited increased right ventricular (RV) weight/body weight ratio and elevated RV pressure in the absence of altered left ventricular filling pressures, accompanied by increase in wall thickness of muscularized pulmonary arteries and a reduction in microvascular perfusion. In isolated lung preparations, KO revealed reduced basal NO, blunted dose-responsiveness to acetylcholine, and augmented basal and angiotensin (AII)-induced pulmonary vascular resistance (R₀) compared with WT. Pre-treatment of KO lungs with S100A1 attenuated the AII-induced increase in pulmonary arterial pressure and R₀. S100A1-induced phosphorylation of eNOS, Akt, and ERK1/2 is attenuated in pulmonary EC of KO compared with WT. Basal and TNF-α-induced EC apoptosis is greater in KO vs. WT, and cell survival is enhanced by S100A1 treatment. CONCLUSION Our data demonstrate that the absence of S100A1 results in PH by disruption of its normal capacity to (i) enhance pulmonary EC function by induction of eNOS activity and NO levels via Akt/ERK1/2 pathways and (ii) promote EC survival. The ability of exogenously administered S100A1 to rescue this phenotype makes it an attractive therapeutic target in the treatment of PH.
Collapse
Affiliation(s)
| | | | | | - Jun Yin
- Department of Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Canada
| | - Liming Wang
- Department of Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Canada
| | - Wolfgang M Kuebler
- Department of Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Canada
| | | |
Collapse
|
19
|
Vinegoni C, Lee S, Gorbatov R, Weissleder R. Motion compensation using a suctioning stabilizer for intravital microscopy. INTRAVITAL 2014; 1:115-121. [PMID: 24086796 DOI: 10.4161/intv.23017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Motion artifacts continue to present a major challenge to single cell imaging in cardiothoracic organs such as the beating heart, blood vessels, or lung. In this study, we present a new water-immersion suctioning stabilizer that enables minimally invasive intravital fluorescence microscopy using water-based stick objectives. The stabilizer works by reducing major motion excursions and can be used in conjunction with both prospective or retrospective gating approaches. We show that the new approach offers cellular resolution in the beating murine heart without perturbing normal physiology. In addition, because this technique allows multiple areas to be easily probed, it offers the opportunity for wide area coverage at high resolution.
Collapse
|
20
|
Becker MO, Kill A, Kutsche M, Guenther J, Rose A, Tabeling C, Witzenrath M, Kühl AA, Heidecke H, Ghofrani HA, Tiede H, Schermuly RT, Nickel N, Hoeper MM, Lukitsch I, Gollasch M, Kuebler WM, Bock S, Burmester GR, Dragun D, Riemekasten G. Vascular Receptor Autoantibodies in Pulmonary Arterial Hypertension Associated with Systemic Sclerosis. Am J Respir Crit Care Med 2014; 190:808-17. [DOI: 10.1164/rccm.201403-0442oc] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
21
|
Benfeitas R, Selvaggio G, Antunes F, Coelho PMBM, Salvador A. Hydrogen peroxide metabolism and sensing in human erythrocytes: a validated kinetic model and reappraisal of the role of peroxiredoxin II. Free Radic Biol Med 2014; 74:35-49. [PMID: 24952139 DOI: 10.1016/j.freeradbiomed.2014.06.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/26/2014] [Accepted: 06/10/2014] [Indexed: 01/09/2023]
Abstract
Hydrogen peroxide (H2O2) metabolism in human erythrocytes has been thoroughly investigated, but unclear points persist. By integrating the available data into a mathematical model that accurately represents the current understanding and comparing computational predictions to observations we sought to (a) identify inconsistencies in present knowledge, (b) propose resolutions, and (c) examine their functional implications. The systematic confrontation of computational predictions with experimental observations of the responses of intact erythrocytes highlighted the following important discrepancy. The high rate constant (10(7)-10(8) M(-1) s(-1)) for H2O2 reduction determined for purified peroxiredoxin II (Prx2) and the high abundance of this protein indicate that under physiological conditions it consumes practically all the H2O2. However, this is inconsistent with extensive evidence that Prx2's contribution to H2O2 elimination is comparable to that of catalase. Models modified such that Prx2's effective peroxidase activity is just 10(5) M(-1) s(-1) agree near quantitatively with extensive experimental observations. This low effective activity is probably due to a strong but readily reversible inhibition of Prx2's peroxidatic activity in intact cells, implying that the main role of Prx2 in human erythrocytes is not to eliminate peroxide substrates. Simulations of the responses to physiological H2O2 stimuli highlight that a design combining abundant Prx2 with a low effective peroxidase activity spares NADPH while improving potential signaling properties of the Prx2/thioredoxin/thioredoxin reductase system.
Collapse
Affiliation(s)
- Rui Benfeitas
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Gianluca Selvaggio
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Fernando Antunes
- Departamento de Química e Bioquímica and Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro M B M Coelho
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Armindo Salvador
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Coimbra Chemistry Center, University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
22
|
Matrix biology of idiopathic pulmonary fibrosis: a workshop report of the national heart, lung, and blood institute. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1643-51. [PMID: 24726499 DOI: 10.1016/j.ajpath.2014.02.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 02/12/2014] [Accepted: 02/18/2014] [Indexed: 01/17/2023]
Abstract
A hallmark of idiopathic pulmonary fibrosis (IPF) is excessive and disordered deposition of extracellular matrix. Although the lung extracellular matrix normally plays an essential role in development and maintenance of lung tissue through reciprocal interactions with resident cells, the disordered matrix in the diseased lung is increasingly recognized as an active and important contributor to IPF pathogenesis. This working group summary from a recently conducted National Heart, Lung, and Blood Institute strategic planning workshop for IPF research highlights recent advances, challenges, and opportunities in the study of matrix biology in IPF. Particular attention is given to the composition and mechanical properties of the matrix in normal and diseased lungs, and the biochemical and biomechanical influences exerted by pathological matrix. Recently developed model systems are also summarized as key tools for advancing our understanding of matrix biology in IPF. Emerging approaches to therapeutically target the matrix in preclinical and clinical settings are discussed, as are important concepts, such as alterations of the matrix with aging and the potential for the resolution of fibrosis. Specific recommendations for future studies in matrix biology of IPF are also proposed.
Collapse
|
23
|
Frevert U, Nacer A, Cabrera M, Movila A, Leberl M. Imaging Plasmodium immunobiology in the liver, brain, and lung. Parasitol Int 2013; 63:171-86. [PMID: 24076429 DOI: 10.1016/j.parint.2013.09.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 08/28/2013] [Accepted: 09/18/2013] [Indexed: 01/10/2023]
Abstract
Plasmodium falciparum malaria is responsible for the deaths of over half a million African children annually. Until a decade ago, dynamic analysis of the malaria parasite was limited to in vitro systems with the typical limitations associated with 2D monocultures or entirely artificial surfaces. Due to extremely low parasite densities, the liver was considered a black box in terms of Plasmodium sporozoite invasion, liver stage development, and merozoite release into the blood. Further, nothing was known about the behavior of blood stage parasites in organs such as the brain where clinical signs manifest and the ensuing immune response of the host that may ultimately result in a fatal outcome. The advent of fluorescent parasites, advances in imaging technology, and availability of an ever-increasing number of cellular and molecular probes have helped illuminate many steps along the pathogenetic cascade of this deadly tropical parasite.
Collapse
Affiliation(s)
- Ute Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, 341 E 25 Street, New York, NY 10010, USA.
| | | | | | | | | |
Collapse
|
24
|
Abstract
The distal airways are covered with a heterogeneous layer of cells known as the alveolar epithelium. Alveolar epithelial cells provide the major barrier between the airspace and fluid filled tissue compartments. As such, regulation of the alveolar epithelium is critical to maintain a healthy lung and for optimal gas exchange. In this chapter, we discuss functional roles for alveolar epithelial cells with particular emphasis on intercellular junctions and communication. As a thin layer of cells directly exposed to atmospheric oxygen, alveoli are particularly sensitive to oxidant insults. Alcohol significantly diminishes the normal antioxidant reserves of the alveolar epithelium, thereby rendering it sensitized for an exaggerated damage response to acute and chronic injuries. The effects of alcohol on alveolar epithelia are discussed along with open questions and potential therapeutic targets to prevent the pathophysiology of alcoholic lung disease.
Collapse
|
25
|
Imaging inflammatory leukocyte recruitment in kidney, lung and liver—challenges to the multi‐step paradigm. Immunol Cell Biol 2013; 91:281-9. [DOI: 10.1038/icb.2012.83] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
26
|
Morty RE, Matalon S. “Real-time visualization of lung function: from micro to macro”. Am J Physiol Lung Cell Mol Physiol 2013; 304:L1-3. [DOI: 10.1152/ajplung.00279.2012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Rory E. Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University Hospital Giessen and Marburg, Giessen, Germany; and
| | - Sadis Matalon
- Department of Anesthesiology, University of Alabama at Birmingham, Alabama
| |
Collapse
|
27
|
Wang L, Yin J, Nickles HT, Ranke H, Tabuchi A, Hoffmann J, Tabeling C, Barbosa-Sicard E, Chanson M, Kwak BR, Shin HS, Wu S, Isakson BE, Witzenrath M, de Wit C, Fleming I, Kuppe H, Kuebler WM. Hypoxic pulmonary vasoconstriction requires connexin 40-mediated endothelial signal conduction. J Clin Invest 2012; 122:4218-30. [PMID: 23093775 PMCID: PMC3484430 DOI: 10.1172/jci59176] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 08/30/2012] [Indexed: 12/21/2022] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a physiological mechanism by which pulmonary arteries constrict in hypoxic lung areas in order to redirect blood flow to areas with greater oxygen supply. Both oxygen sensing and the contractile response are thought to be intrinsic to pulmonary arterial smooth muscle cells. Here we speculated that the ideal site for oxygen sensing might instead be at the alveolocapillary level, with subsequent retrograde propagation to upstream arterioles via connexin 40 (Cx40) endothelial gap junctions. HPV was largely attenuated by Cx40-specific and nonspecific gap junction uncouplers in the lungs of wild-type mice and in lungs from mice lacking Cx40 (Cx40-/-). In vivo, hypoxemia was more severe in Cx40-/- mice than in wild-type mice. Real-time fluorescence imaging revealed that hypoxia caused endothelial membrane depolarization in alveolar capillaries that propagated to upstream arterioles in wild-type, but not Cx40-/-, mice. Transformation of endothelial depolarization into vasoconstriction involved endothelial voltage-dependent α1G subtype Ca2+ channels, cytosolic phospholipase A2, and epoxyeicosatrienoic acids. Based on these data, we propose that HPV originates at the alveolocapillary level, from which the hypoxic signal is propagated as endothelial membrane depolarization to upstream arterioles in a Cx40-dependent manner.
Collapse
MESH Headings
- Animals
- Calcium Channels/metabolism
- Connexins/genetics
- Connexins/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Human Umbilical Vein Endothelial Cells
- Humans
- Hypoxia/genetics
- Hypoxia/metabolism
- Hypoxia/pathology
- Hypoxia/physiopathology
- Lung/blood supply
- Lung/metabolism
- Lung/pathology
- Lung/physiopathology
- Mice
- Mice, Knockout
- Muscle, Smooth/metabolism
- Muscle, Smooth/pathology
- Muscle, Smooth/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phospholipases A2, Cytosolic/metabolism
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Signal Transduction
- Vasoconstriction
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Liming Wang
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jun Yin
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hannah T. Nickles
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hannes Ranke
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Arata Tabuchi
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Hoffmann
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Christoph Tabeling
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Eduardo Barbosa-Sicard
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Marc Chanson
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Brenda R. Kwak
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hee-Sup Shin
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Songwei Wu
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Brant E. Isakson
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Martin Witzenrath
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Cor de Wit
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Ingrid Fleming
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hermann Kuppe
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Wolfgang M. Kuebler
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
McLaughlin RA, Yang X, Quirk BC, Lorenser D, Kirk RW, Noble PB, Sampson DD. Static and dynamic imaging of alveoli using optical coherence tomography needle probes. J Appl Physiol (1985) 2012; 113:967-74. [DOI: 10.1152/japplphysiol.00051.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Imaging of alveoli in situ has for the most part been infeasible due to the high resolution required to discern individual alveoli and limited access to alveoli beneath the lung surface. In this study, we present a novel technique to image alveoli using optical coherence tomography (OCT). We propose the use of OCT needle probes, where the distal imaging probe has been miniaturized and encased within a hypodermic needle (as small as 30-gauge, outer diameter 310 μm), allowing insertion deep within the lung tissue with minimal tissue distortion. Such probes enable imaging at a resolution of ∼12 μm within a three-dimensional cylindrical field of view with diameter ∼1.5 mm centered on the needle tip. The imaging technique is demonstrated on excised lungs from three different species: adult rats, fetal sheep, and adult pigs. OCT needle probes were used to image alveoli, small bronchioles, and blood vessels, and results were matched to histological sections. We also present the first dynamic OCT images acquired with an OCT needle probe, allowing tracking of individual alveoli during simulated cyclical lung inflation and deflation.
Collapse
Affiliation(s)
- Robert A. McLaughlin
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, University of Western Australia, Crawley, Western Australia, Australia
| | - Xiaojie Yang
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, University of Western Australia, Crawley, Western Australia, Australia
| | - Bryden C. Quirk
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, University of Western Australia, Crawley, Western Australia, Australia
| | - Dirk Lorenser
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, University of Western Australia, Crawley, Western Australia, Australia
| | - Rodney W. Kirk
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B. Noble
- Centre for Neonatal Research and Education, School of Women's and Infants' Health, University of Western Australia, Crawley, Western Australia, Australia; and
| | - David D. Sampson
- Optical + Biomedical Engineering Laboratory, School of Electrical, Electronic and Computer Engineering, University of Western Australia, Crawley, Western Australia, Australia
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Crawley Western Australia, Australia
| |
Collapse
|
29
|
Kellner M, Heidrich M, Beigel R, Lorbeer RA, Knudsen L, Ripken T, Heisterkamp A, Meyer H, Kühnel MP, Ochs M. Imaging of the mouse lung with scanning laser optical tomography (SLOT). J Appl Physiol (1985) 2012; 113:975-83. [DOI: 10.1152/japplphysiol.00026.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The current study focuses on the use of scanning laser optical tomography (SLOT) in imaging of the mouse lung ex vivo. SLOT is a highly efficient fluorescence microscopy technique allowing rapid scanning of samples of a size of several millimeters, thus enabling volumetric visualization by using intrinsic contrast mechanisms of previously fixed lung lobes. Here, we demonstrate the imaging of airways, blood vessels, and parenchyma from whole, optically cleared mouse lung lobes with a resolution down to the level of single alveoli using absorption and autofluorescence scan modes. The internal structure of the lung can then be analyzed nondestructively and quantitatively in three-dimensional datasets in any preferred planar orientation. Moreover, the procedure preserves the microscopic structure of the lung and allows for subsequent correlative histologic studies. In summary, the current study has shown that SLOT is a valuable technique to study the internal structure of the mouse lung.
Collapse
Affiliation(s)
- Manuela Kellner
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Marko Heidrich
- Biomedical Optics Department, Laser Zentrum Hannover e.V., Hannover, Germany
| | - Rebecca Beigel
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover, Germany
| | | | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, Hannover, Germany
| | - Tammo Ripken
- Biomedical Optics Department, Laser Zentrum Hannover e.V., Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover, Germany
| | - Alexander Heisterkamp
- REBIRTH Cluster of Excellence, Hannover, Germany
- German Center for Lung Research, Hannover, Germany
- Institute of Applied Optics, Friedrich-Schiller-University Jena, Jena, Germany; and
| | - Heiko Meyer
- Biomedical Optics Department, Laser Zentrum Hannover e.V., Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover, Germany
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Mark Philipp Kühnel
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover, Germany
- German Center for Lung Research, Hannover, Germany
| |
Collapse
|
30
|
Kar R, Batra N, Riquelme MA, Jiang JX. Biological role of connexin intercellular channels and hemichannels. Arch Biochem Biophys 2012; 524:2-15. [PMID: 22430362 PMCID: PMC3376239 DOI: 10.1016/j.abb.2012.03.008] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/16/2012] [Accepted: 03/06/2012] [Indexed: 12/11/2022]
Abstract
Gap junctions (GJ) and hemichannels (HC) formed from the protein subunits called connexins are transmembrane conduits for the exchange of small molecules and ions. Connexins and another group of HC-forming proteins, pannexins comprise the two families of transmembrane proteins ubiquitously distributed in vertebrates. Most cell types express more than one connexin or pannexin. While connexin expression and channel activity may vary as a function of physiological and pathological states of the cell and tissue, only a few studies suggest the involvement of pannexin HC in acquired pathological conditions. Importantly, genetic mutations in connexin appear to interfere with GJ and HC function which results in several diseases. Thus connexins could serve as potential drug target for therapeutic intervention. Growing evidence suggests that diseases resulting from HC dysfunction might open a new direction for development of specific HC reagents. This review provides a comprehensive overview of the current studies of GJ and HC formed by connexins and pannexins in various tissue and organ systems including heart, central nervous system, kidney, mammary glands, ovary, testis, lens, retina, inner ear, bone, cartilage, lung and liver. In addition, present knowledge of the role of GJ and HC in cell cycle progression, carcinogenesis and stem cell development is also discussed.
Collapse
Affiliation(s)
| | | | - Manuel A Riquelme
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900
| | - Jean X. Jiang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900
| |
Collapse
|
31
|
Hobi N, Ravasio A, Haller T. Interfacial stress affects rat alveolar type II cell signaling and gene expression. Am J Physiol Lung Cell Mol Physiol 2012; 303:L117-29. [PMID: 22610352 DOI: 10.1152/ajplung.00340.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous work from our group (Ravasio A, Hobi N, Bertocchi C, Jesacher A, Dietl P, Haller T. Am J Physiol Cell Physiol 300: C1456-C1465, 2011.) showed that contact of alveolar epithelial type II cells with an air-liquid interface (I(AL)) leads to a paradoxical situation. It is a potential threat that can cause cell injury, but also a Ca(2+)-dependent stimulus for surfactant secretion. Both events can be explained by the impact of interfacial tensile forces on cellular structures. Here, the strength of this mechanical stimulus became also apparent in microarray studies by a rapid and significant change on the transcriptional level. Cells challenged with an I(AL) in two different ways showed activation/inactivation of cellular pathways involved in stress response and defense, and a detailed Pubmatrix search identified genes associated with several lung diseases and injuries. Altogether, they suggest a close relationship of interfacial stress sensation with current models in alveolar micromechanics. Further similarities between I(AL) and cell stretch were found with respect to the underlying signaling events. The source of Ca(2+) was extracellular, and the transmembrane Ca(2+) entry pathway suggests the involvement of a mechanosensitive channel. We conclude that alveolar type II cells, due to their location and morphology, are specific sensors of the I(AL), but largely protected from interfacial stress by surfactant release.
Collapse
Affiliation(s)
- Nina Hobi
- Department of Physiology and Medical Physics, Division of Physiology, Innsbruck Medical University, Austria
| | | | | |
Collapse
|
32
|
Westphalen K, Monma E, Islam MN, Bhattacharya J. Acid contact in the rodent pulmonary alveolus causes proinflammatory signaling by membrane pore formation. Am J Physiol Lung Cell Mol Physiol 2012; 303:L107-16. [PMID: 22561462 DOI: 10.1152/ajplung.00206.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although gastric acid aspiration causes rapid lung inflammation and acute lung injury, the initiating mechanisms are not known. To determine alveolar epithelial responses to acid, we viewed live alveoli of the isolated lung by fluorescence microscopy, then we microinjected the alveoli with HCl at pH of 1.5. The microinjection caused an immediate but transient formation of molecule-scale pores in the apical alveolar membrane, resulting in loss of cytosolic dye. However, the membrane rapidly resealed. There was no cell damage and no further dye loss despite continuous HCl injection. Concomitantly, reactive oxygen species (ROS) increased in the adjacent perialveolar microvascular endothelium in a Ca(2+)-dependent manner. By contrast, ROS did not increase in wild-type mice in which we gave intra-alveolar injections of polyethylene glycol (PEG)-catalase, in mice overexpressing alveolar catalase, or in mice lacking functional NADPH oxidase (Nox2). Together, our findings indicate the presence of an unusual proinflammatory mechanism in which alveolar contact with acid caused membrane pore formation. The effect, although transient, was nevertheless sufficient to induce Ca(2+) entry and Nox2-dependent H(2)O(2) release from the alveolar epithelium. These responses identify alveolar H(2)O(2) release as the signaling mechanism responsible for lung inflammation induced by acid and suggest that intra-alveolar PEG-catalase might be therapeutic in acid-induced lung injury.
Collapse
Affiliation(s)
- Kristin Westphalen
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | | | | | | |
Collapse
|
33
|
Samapati R, Yang Y, Yin J, Stoerger C, Arenz C, Dietrich A, Gudermann T, Adam D, Wu S, Freichel M, Flockerzi V, Uhlig S, Kuebler WM. Lung endothelial Ca2+ and permeability response to platelet-activating factor is mediated by acid sphingomyelinase and transient receptor potential classical 6. Am J Respir Crit Care Med 2012; 185:160-70. [PMID: 22246702 DOI: 10.1164/rccm.201104-0717oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
RATIONALE Platelet-activating factor (PAF) increases lung vascular permeability within minutes by activation of acid sphingomyelinase (ASM) and a subsequent nitric oxide (NO)-inhibitable and Ca(2+)-dependent loss in barrier function. OBJECTIVES To elucidate the molecular mechanisms underlying this response. METHODS In isolated perfused rat and mouse lungs, endothelial Ca(2+) concentration ([Ca(2+)](i)) was quantified by real-time fluorescence imaging, and caveolae of endothelial cells were isolated and probed for Ca(2+) entry channels. Regulation of transient receptor potential classical (TRPC) 6-mediated currents in lung endothelial cells was assessed by patch clamp technique. MEASUREMENTS AND MAIN RESULTS PAF increased lung weight gain and endothelial [Ca(2+)](i). This response was abrogated by inhibitors of ASM or in ASM-deficient mice, and replicated by lung perfusion with exogenous ASM or C2-ceramide. PAF increased the caveolar abundance of TRPC6 channels, which was similarly blocked by ASM inhibition. PAF-induced increases in lung endothelial [Ca(2+)](i), vascular filtration coefficient, and edema formation were attenuated by the TRPC inhibitor SKF96365 and in TRPC6-deficient mice, whereas direct activation of TRPC6 replicated the [Ca(2+)](i) and edema response to PAF. The exogenous NO donor PapaNONOate or the cyclic guanosine 3',5'-monophosphate analog 8Br-cGMP blocked the endothelial [Ca(2+)](i) and permeability response to PAF, in that they directly blocked TRPC6 channels without interfering with their PAF-induced recruitment to caveolae. CONCLUSIONS The present findings outline a new signaling cascade in the induction of PAF-induced lung edema, in that stimulation of ASM causes recruitment of TRPC6 channels to caveolae, thus allowing for Ca(2+) influx and subsequent increases in endothelial permeability that are amplified in the absence of endothelial NO synthesis.
Collapse
Affiliation(s)
- Rudi Samapati
- Institute for Physiology, Charité - Universitätsmedizin Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Terpolilli NA, Kim SW, Thal SC, Kataoka H, Zeisig V, Nitzsche B, Klaesner B, Zhu C, Schwarzmaier S, Meissner L, Mamrak U, Engel DC, Drzezga A, Patel RP, Blomgren K, Barthel H, Boltze J, Kuebler WM, Plesnila N. Inhalation of nitric oxide prevents ischemic brain damage in experimental stroke by selective dilatation of collateral arterioles. Circ Res 2011; 110:727-38. [PMID: 22207711 DOI: 10.1161/circresaha.111.253419] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Stroke is the third most common cause of death in industrialized countries. The main therapeutic target is the ischemic penumbra, potentially salvageable brain tissue that dies within the first few hours after blood flow cessation. Hence, strategies to keep the penumbra alive until reperfusion occurs are needed. OBJECTIVE To study the effect of inhaled nitric oxide on cerebral vessels and cerebral perfusion under physiological conditions and in different models of cerebral ischemia. METHODS AND RESULTS This experimental study demonstrates that inhaled nitric oxide (applied in 30% oxygen/70% air mixture) leads to the formation of nitric oxide carriers in blood that distribute throughout the body. This was ascertained by in vivo microscopy in adult mice. Although under normal conditions inhaled nitric oxide does not affect cerebral blood flow, after experimental cerebral ischemia induced by transient middle cerebral artery occlusion it selectively dilates arterioles in the ischemic penumbra, thereby increasing collateral blood flow and significantly reducing ischemic brain damage. This translates into significantly improved neurological outcome. These findings were validated in independent laboratories using two different mouse models of cerebral ischemia and in a clinically relevant large animal model of stroke. CONCLUSIONS Inhaled nitric oxide thus may provide a completely novel strategy to improve penumbral blood flow and neuronal survival in stroke or other ischemic conditions.
Collapse
Affiliation(s)
- Nicole A Terpolilli
- Laboratory of Experimental Neurosurgery, University of Munich Medical Center-Grosshadern, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Roan E, Waters CM. What do we know about mechanical strain in lung alveoli? Am J Physiol Lung Cell Mol Physiol 2011; 301:L625-35. [PMID: 21873445 PMCID: PMC3213982 DOI: 10.1152/ajplung.00105.2011] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 08/23/2011] [Indexed: 12/25/2022] Open
Abstract
The pulmonary alveolus, terminal gas-exchange unit of the lung, is composed of alveolar epithelial and endothelial cells separated by a thin basement membrane and interstitial space. These cells participate in the maintenance of a delicate system regulated not only by biological factors but also by the mechanical environment of the lung, which undergoes dynamic deformation during breathing. Clinical and animal studies as well as cell culture studies point toward a strong influence of mechanical forces on lung cells and tissues including effects on growth and repair, surfactant release, injury, and inflammation. However, despite substantial advances in our understanding of lung mechanics over the last half century, there are still many unanswered questions regarding the micromechanics of the alveolus and how it deforms during lung inflation. Therefore, the aims of this review are to draw a multidisciplinary account of the mechanics of the alveolus on the basis of its structure, biology, and chemistry and to compare estimates of alveolar deformation from previous studies.
Collapse
Affiliation(s)
- Esra Roan
- Departments of Biomedical Engineering and Mechanical Engineering, University of Memphis, Memphis, Tennessee 38163-0001, USA
| | | |
Collapse
|
36
|
Two-photon imaging within the murine thorax without respiratory and cardiac motion artifact. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:75-82. [PMID: 21703395 DOI: 10.1016/j.ajpath.2011.03.048] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 03/21/2011] [Accepted: 03/29/2011] [Indexed: 11/22/2022]
Abstract
Intravital microscopy has been recognized for its ability to make physiological measurements at cellular and subcellular levels while maintaining the complex natural microenvironment. Two-photon microscopy (TPM), using longer wavelengths than single-photon excitation, has extended intravital imaging deeper into tissues, with minimal phototoxicity. However, due to a relatively slow acquisition rate, TPM is especially sensitive to motion artifact, which presents a challenge when imaging tissues subject to respiratory and cardiac movement. Thoracoabdominal organs that cannot be exteriorized or immobilized during TPM have generally required the use of isolated, pump-perfused preparations. However, this approach entails significant alteration of normal physiology, such as a lack of neural inputs, increased vascular resistance, and leukocyte activation. We adapted techniques of intravital microscopy that permitted TPM of organs maintained within the thoracoabdominal cavity of living, breathing rats or mice. We obtained extended intravital TPM imaging of the intact lung, arguably the organ most susceptible to both respiratory and cardiac motion. Intravital TPM detected the development of lung microvascular endothelial activation manifested as increased leukocyte adhesion and plasma extravasation in response to oxidative stress inducers PMA or soluble cigarette smoke extract. The pulmonary microvasculature and alveoli in the intact animal were imaged with comparable detail and fidelity to those in pump-perfused animals, opening the possibility for TPM of other thoracoabdominal organs under physiological and pathophysiological conditions.
Collapse
|
37
|
Rowlands DJ, Islam MN, Das SR, Huertas A, Quadri SK, Horiuchi K, Inamdar N, Emin MT, Lindert J, Ten VS, Bhattacharya S, Bhattacharya J. Activation of TNFR1 ectodomain shedding by mitochondrial Ca2+ determines the severity of inflammation in mouse lung microvessels. J Clin Invest 2011; 121:1986-99. [PMID: 21519143 DOI: 10.1172/jci43839] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 02/09/2011] [Indexed: 12/25/2022] Open
Abstract
Shedding of the extracellular domain of cytokine receptors allows the diffusion of soluble receptors into the extracellular space; these then bind and neutralize their cytokine ligands, thus dampening inflammatory responses. The molecular mechanisms that control this process, and the extent to which shedding regulates cytokine-induced microvascular inflammation, are not well defined. Here, we used real-time confocal microscopy of mouse lung microvascular endothelium to demonstrate that mitochondria are key regulators of this process. The proinflammatory cytokine soluble TNF-α (sTNF-α) increased mitochondrial Ca2+, and the purinergic receptor P2Y2 prolonged the response. Concomitantly, the proinflammatory receptor TNF-α receptor-1 (TNFR1) was shed from the endothelial surface. Inhibiting the mitochondrial Ca2+ increase blocked the shedding and augmented inflammation, as denoted by increases in endothelial expression of the leukocyte adhesion receptor E-selectin and in microvascular leukocyte recruitment. The shedding was also blocked in microvessels after knockdown of a complex III component and after mitochondria-targeted catalase overexpression. Endothelial deletion of the TNF-α converting enzyme (TACE) prevented the TNF-α receptor shedding response, which suggests that exposure of microvascular endothelium to sTNF-α induced a Ca2+-dependent increase of mitochondrial H2O2 that caused TNFR1 shedding through TACE activation. These findings provide what we believe to be the first evidence that endothelial mitochondria regulate TNFR1 shedding and thereby determine the severity of sTNF-α-induced microvascular inflammation.
Collapse
Affiliation(s)
- David J Rowlands
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Tyurina YY, Tyurin VA, Kapralova VI, Wasserloos K, Mosher M, Epperly MW, Greenberger JS, Pitt BR, Kagan VE. Oxidative lipidomics of γ-radiation-induced lung injury: mass spectrometric characterization of cardiolipin and phosphatidylserine peroxidation. Radiat Res 2011; 175:610-21. [PMID: 21338246 DOI: 10.1667/rr2297.1] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Oxidative damage plays a significant role in the pathogenesis of γ-radiation-induced lung injury. Endothelium is a preferred target for early radiation-induced damage and apoptosis. Given the newly discovered role of oxidized phospholipids in apoptotic signaling, we performed oxidative lipidomics analysis of phospholipids in irradiated mouse lungs and cultured mouse lung endothelial cells. C57BL/6NHsd female mice were subjected to total-body irradiation (10 Gy, 15 Gy) and euthanized 24 h thereafter. Mouse lung endothelial cells were analyzed 48 h after γ irradiation (15 Gy). We found that radiation-induced apoptosis in vivo and in vitro was accompanied by non-random oxidation of phospholipids. Cardiolipin and phosphatidylserine were the major oxidized phospholipids, while more abundant phospholipids (phosphatidylcholine, phosphatidylethanolamine) remained non-oxidized. Electrospray ionization mass spectrometry analysis revealed the formation of cardiolipin and phosphatidylserine oxygenated molecular species in the irradiated lung and cells. Analysis of fatty acids after hydrolysis of cardiolipin and phosphatidylserine by phospholipase A(2) revealed the presence of mono-hydroperoxy and/or mono-hydroxy/mono-epoxy, mono-hydroperoxy/mono-oxo molecular species of linoleic acid. We speculate that cyt c-driven oxidations of cardiolipin and phosphatidylserine associated with the execution of apoptosis in pulmonary endothelial cells are important contributors to endothelium dysfunction in γ-radiation-induced lung injury.
Collapse
Affiliation(s)
- Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Bridgeside Point, 100 Technology Drive, Suite 350, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Erzurum S, Rounds SI, Stevens T, Aldred M, Aliotta J, Archer SL, Asosingh K, Balaban R, Bauer N, Bhattacharya J, Bogaard H, Choudhary G, Dorn GW, Dweik R, Fagan K, Fallon M, Finkel T, Geraci M, Gladwin MT, Hassoun PM, Humbert M, Kaminski N, Kawut SM, Loscalzo J, McDonald D, McMurtry IF, Newman J, Nicolls M, Rabinovitch M, Shizuru J, Oka M, Polgar P, Rodman D, Schumacker P, Stenmark K, Tuder R, Voelkel N, Sullivan E, Weinshilboum R, Yoder MC, Zhao Y, Gail D, Moore TM. Strategic plan for lung vascular research: An NHLBI-ORDR Workshop Report. Am J Respir Crit Care Med 2010; 182:1554-62. [PMID: 20833821 DOI: 10.1164/rccm.201006-0869ws] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The Division of Lung Diseases of the National Heart, Lung, and Blood Institute, with the Office of Rare Diseases Research, held a workshop to identify priority areas and strategic goals to enhance and accelerate research that will result in improved understanding of the lung vasculature, translational research needs, and ultimately the care of patients with pulmonary vascular diseases. Multidisciplinary experts with diverse experience in laboratory, translational, and clinical studies identified seven priority areas and discussed limitations in our current knowledge, technologies, and approaches. The focus for future research efforts include the following: (1) better characterizing vascular genotype-phenotype relationships and incorporating systems biology approaches when appropriate; (2) advancing our understanding of pulmonary vascular metabolic regulatory signaling in health and disease; (3) expanding our knowledge of the biologic relationships between the lung circulation and circulating elements, systemic vascular function, and right heart function and disease; (4) improving translational research for identifying disease-modifying therapies for the pulmonary hypertensive diseases; (5) establishing an appropriate and effective platform for advancing translational findings into clinical studies testing; and (6) developing the specific technologies and tools that will be enabling for these goals, such as question-guided imaging techniques and lung vascular investigator training programs. Recommendations from this workshop will be used within the Lung Vascular Biology and Disease Extramural Research Program for planning and strategic implementation purposes.
Collapse
Affiliation(s)
- Serpil Erzurum
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nava RG, Li W, Gelman AE, Krupnick AS, Miller MJ, Kreisel D. Two-photon microscopy in pulmonary research. Semin Immunopathol 2010; 32:297-304. [PMID: 20589501 DOI: 10.1007/s00281-010-0209-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 05/28/2010] [Indexed: 12/12/2022]
Abstract
As the lung is constantly exposed to both innocuous and potentially noxious antigens, a thorough understanding of both innate and adaptive immune responses in this organ is of the essence. Imaging modalities such as magnetic resonance imaging, positron emission tomography, and confocal microscopy have expanded our knowledge about various molecular processes and cellular responses in the lung. Two-photon microscopy has evolved into a powerful tool to observe cellular interactions in real time and has markedly expanded our understanding of the immune system. Recently, two-photon microscopy has also been utilized to image the murine lung. As immune responses in the lung differ from those in other non-lymphoid tissues, this technique holds great promise to advance our knowledge of the biology that underlies a wide spectrum of pulmonary diseases.
Collapse
Affiliation(s)
- Ruben G Nava
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | | | | |
Collapse
|
42
|
Zhou C, Chen H, King JA, Sellak H, Kuebler WM, Yin J, Townsley MI, Shin HS, Wu S. Alpha1G T-type calcium channel selectively regulates P-selectin surface expression in pulmonary capillary endothelium. Am J Physiol Lung Cell Mol Physiol 2010; 299:L86-97. [PMID: 20435690 DOI: 10.1152/ajplung.00331.2009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Regulated P-selectin surface expression provides a rapid measure for endothelial transition to a proinflammatory phenotype. In general, P-selectin surface expression results from Weibel-Palade body (WPb) exocytosis. Yet, it is unclear whether pulmonary capillary endothelium possesses WPbs or regulated P-selectin surface expression and, if so, how inflammatory stimuli initiate exocytosis. We used immunohistochemistry, immunofluorescence labeling, ultrastructural assessment, and an isolated perfused lung model to demonstrate that capillary endothelium lacks WPbs but possesses P-selectin. Thrombin stimulated P-selectin surface expression in both extra-alveolar vessel and alveolar capillary endothelium. Only in capillaries was the thrombin-stimulated P-selectin surface expression considerably mitigated by pharmacologic blockade of the T-type channel or genetic knockout of the T-type channel alpha(1G)-subunit. Depolarization of endothelial plasma membrane via high K(+) perfusion capable of eliciting cytosolic Ca(2+) transients also provoked P-selectin surface expression in alveolar capillaries that was abolished by T-type channel blockade or alpha(1G) knockout. Our findings reveal an intracellular WPb-independent P-selectin pool in pulmonary capillary endothelium, where the regulated P-selectin surface expression is triggered by Ca(2+) transients evoked through activation of the alpha(1G) T-type channel.
Collapse
Affiliation(s)
- Chun Zhou
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, Alabama 36688-0002, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hasegawa A, Hayashi K, Kishimoto H, Yang M, Tofukuji S, Suzuki K, Nakajima H, Hoffman RM, Shirai M, Nakayama T. Color-coded real-time cellular imaging of lung T-lymphocyte accumulation and focus formation in a mouse asthma model. J Allergy Clin Immunol 2010; 125:461-468.e6. [PMID: 20031194 DOI: 10.1016/j.jaci.2009.09.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 07/26/2009] [Accepted: 08/02/2009] [Indexed: 10/19/2022]
Abstract
BACKGROUND A critical role for CD4(+)T(H)2 cells in the pathogenesis of acute asthma has been demonstrated in the studies of human asthma as well as of animal models of asthma. T(H)2-cell migration into the lung is crucial for the initiation of asthma phenotype, but the dynamics of this process are poorly understood because it has been difficult to visualize this process. OBJECTIVE Our aim was to image the cellular dynamics of the migration of T(H)2 cells into the lung of living animals in a mouse model of asthma and identify the cellular processes required for the initiation of the asthma phenotype. METHODS We developed a color-coded real-time imaging model of cell migration into the lung using green fluorescent protein (GFP) and red fluorescent protein (RFP) transgenic CD4 T cells. RESULTS Selective accumulation of antigen-specific CD4 T cells in the lungs was quantitatively imaged in a mouse model of asthma. The inhibition of accumulation by dexamethasone was imaged. Accumulating GFP(+) T(H)2 cells formed foci in the lungs from 6 to 20 hours after antigen inhalation. This process was also inhibited by the administration of anti-intercellular adhesion molecule 1 or anti-vascular cell adhesion molecule 1 mAbs. Two days after inhalation of antigen, GFP(+) T(H)2 cells were detected in the area of eosinophil infiltration. CONCLUSION Focus formation generated by accumulating antigen-specific T(H)2 cells in the lung appeared to be a critical process in the initiation of the asthma phenotype. This new model enables the study of in vivo cell biology of airway inflammation and novel drug discovery for lung inflammatory diseases.
Collapse
Affiliation(s)
- Akihiro Hasegawa
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Lung function is inextricably linked to mechanics. On short timescales every breath generates dynamic cycles of cell and matrix stretch, along with convection of fluids in the airways and vasculature. Perturbations such airway smooth muscle shortening or surfactant dysfunction rapidly alter respiratory mechanics, with profound influence on lung function. On longer timescales, lung development, maturation, and remodeling all strongly depend on cues from the mechanical environment. Thus mechanics has long played a central role in our developing understanding of lung biology and respiratory physiology. This concise review focuses on progress over the past 5 years in elucidating the molecular origins of lung mechanical behavior, and the cellular signaling events triggered by mechanical perturbations that contribute to lung development, homeostasis, and injury. Special emphasis is placed on the tools and approaches opening new avenues for investigation of lung behavior at integrative cellular and molecular scales. We conclude with a brief summary of selected opportunities and challenges that lie ahead for the lung mechanobiology research community.
Collapse
|
45
|
Alveolar dynamics in acute lung injury: heterogeneous distension rather than cyclic opening and collapse. Crit Care Med 2009; 37:2604-11. [PMID: 19623041 DOI: 10.1097/ccm.0b013e3181a5544d] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES : To analyze alveolar dynamics in healthy and acid-injured lungs of ventilated mice. Protective ventilation is potentially lifesaving in patients with acute lung injury. However, optimization of ventilation strategies is hampered by an incomplete understanding of the effects of mechanical ventilation at the alveolar level. DESIGN : In anesthetized and ventilated Balb/c mice, subpleural alveoli were visualized by darkfield intravital microscopy and optical coherence tomography. SETTING : Animal research laboratory. SUBJECTS : Male Balb/c mice. INTERVENTIONS : Lung injury was induced by intratracheal instillation of hydrochloric acid. In control animals and mice with lung injury, ventilation pressures were varied between 0 and 24 cm H2O at baseline, 60 mins, and 120 mins, and alveolar distension and cyclic opening and collapse of alveolar clusters were analyzed. MEASUREMENTS AND MAIN RESULTS : In normal lungs, alveolar clusters distend with increasing ventilation pressure in a sigmoid relationship. Although an increase in ventilation pressure from 0 to 24 cm H2O increases alveolar size by 41.5 +/- 2.3% in normal lungs, alveolar distension is reduced to 20.6 +/- 2.2% 120 mins after induction of lung injury by acid aspiration. Cyclic opening and collapse of alveolar clusters are neither observed in normal nor acid-injured lungs. Alveolar compliance is highest in small and distensible alveolar clusters, which are also most prone to acid-induced injury. CONCLUSIONS : Over the applied pressure range, volume changes in control and acid-injured mouse lungs result predominantly from alveolar distension rather than cyclic opening and collapse of alveolar clusters. Preferential loss of compliance in small alveolar clusters redistributes tidal volume to larger alveoli, which increases spatial heterogeneity in alveolar inflation and may promote alveolar overdistension.
Collapse
|
46
|
Johnson LN, Koval M. Cross-talk between pulmonary injury, oxidant stress, and gap junctional communication. Antioxid Redox Signal 2009; 11:355-67. [PMID: 18816185 PMCID: PMC2933150 DOI: 10.1089/ars.2008.2183] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Gap junction channels interconnect several different types of cells in the lung, ranging from the alveolar epithelium to the pulmonary vasculature, each of which expresses a unique subset of gap junction proteins (connexins). Major lung functions regulated by gap junctional communication include coordination of ciliary beat frequency and inflammation. Gap junctions help enable the alveolus to regulate surfactant secretion as an integrated system, in which type I cells act as mechanical sensors that transmit calcium transients to type II cells. Thus, disruption of epithelial gap junctional communication, particularly during acute lung injury, can interfere with these processes and increase the severity of injury. Consistent with this, connexin expression is altered during lung injury, and connexin-deficiency has a negative impact on the injury response and lung-growth control. It has recently been shown that alcohol abuse is a significant risk factor associated with acute respiratory distress syndrome. Oxidant stress and hormone-signaling cascades in the lung induced by prolonged alcohol ingestion are discussed, as well as the effects of these pathways on connexin expression and function.
Collapse
Affiliation(s)
- Latoya N Johnson
- Division of Pulmonary, Allergy and Critical Care Medicine, and Emory Alcohol and Lung Biology Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
47
|
Lo Celso C, Klein RJ, Scadden DT. Analysis of the hematopoietic stem cell niche. ACTA ACUST UNITED AC 2008; Chapter 2:Unit 2A.5. [PMID: 18785177 DOI: 10.1002/9780470151808.sc02a05s3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Hematopoietic stem cells (HSCs) continuously replenish all blood cell lineages not only to maintain the normal rapid turnover of differentiated cells but also to respond to injury and stress. Cell-extrinsic mechanisms are critical determinants of the fine balance between HSC self-renewal and differentiation. The bone marrow microenvironment has emerged as a new area of intense study to identify which of its many components constitute the HSC niche and regulate HSC fate. While HSCs have been isolated, characterized and used in clinical practice for many years thanks to the development of very specific assays and technology (i.e., bone marrow transplants and fluorescence activated cell sorting), study of the HSC niche has evolved by combining experimental designs developed in different fields. In this unit we describe a collection of protocols spanning a wide range of techniques that can help every researcher tackling questions regarding the nature of the HSC niche.
Collapse
|
48
|
Endothelium–platelet interactions in inflammatory lung disease. Vascul Pharmacol 2008; 49:141-50. [DOI: 10.1016/j.vph.2008.06.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 04/21/2008] [Accepted: 06/13/2008] [Indexed: 01/15/2023]
|
49
|
Yin J, Hoffmann J, Kaestle SM, Neye N, Wang L, Baeurle J, Liedtke W, Wu S, Kuppe H, Pries AR, Kuebler WM. Negative-feedback loop attenuates hydrostatic lung edema via a cGMP-dependent regulation of transient receptor potential vanilloid 4. Circ Res 2008; 102:966-74. [PMID: 18323527 DOI: 10.1161/circresaha.107.168724] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the formation of hydrostatic lung edema is generally attributed to imbalanced Starling forces, recent data show that lung endothelial cells respond to increased vascular pressure and may thus regulate vascular permeability and edema formation. In combining real-time optical imaging of the endothelial Ca(2+) concentration ([Ca(2+)](i)) and NO production with filtration coefficient (K(f)) measurements in the isolated perfused lung, we identified a series of endothelial responses that constitute a negative-feedback loop to protect the microvascular barrier. Elevation of lung microvascular pressure was shown to increase endothelial [Ca(2+)](i) via activation of transient receptor potential vanilloid 4 (TRPV4) channels. The endothelial [Ca(2+)](i) transient increased K(f) via activation of myosin light-chain kinase and simultaneously stimulated NO synthesis. In TRPV4 deficient mice, pressure-induced increases in endothelial [Ca(2+)](i), NO synthesis, and lung wet/dry weight ratio were largely blocked. Endothelial NO formation limited the permeability increase by a cGMP-dependent attenuation of the pressure-induced [Ca(2+)](i) response. Inactivation of TRPV4 channels by cGMP was confirmed by whole-cell patch-clamp of pulmonary microvascular endothelial cells and intravital imaging of endothelial [Ca(2+)](i). Hence, pressure-induced endothelial Ca(2+) influx via TRPV4 channels increases lung vascular permeability yet concomitantly activates an NO-mediated negative-feedback loop that protects the vascular barrier by a cGMP-dependent attenuation of the endothelial [Ca(2+)](i) response. The identification of this novel regulatory pathway gives rise to new treatment strategies, as demonstrated in vivo in rats with acute myocardial infarction in which inhibition of cGMP degradation by the phosphodiesterase 5 inhibitor sildenafil reduced hydrostatic lung edema.
Collapse
Affiliation(s)
- Jun Yin
- Institute of Physiology, Charité-Universitaetsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, 14195 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tabuchi A, Mertens M, Kuppe H, Pries AR, Kuebler WM. Intravital microscopy of the murine pulmonary microcirculation. J Appl Physiol (1985) 2008; 104:338-46. [DOI: 10.1152/japplphysiol.00348.2007] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intravital microscopy (IVM) is considered as the gold standard for in vivo investigations of dynamic microvascular regulation. The availability of transgenic and knockout animals has propelled the development of murine IVM models for various organs, but technical approaches to the pulmonary microcirculation are still scarce. In anesthetized and ventilated BALB/c mice, we established a microscopic access to the surface of the right upper lung lobe by surgical excision of a window of 7- to 10-mm diameter from the right thoracic wall. The window was covered by a transparent polyvinylidene membrane and sealed with α-cyanoacrylate. Removal of intrathoracic air via a transdiaphragmal intrapleural catheter coupled the lung surface to the window membrane. IVM preparations were hemodynamically stable for at least 120 min, with mean arterial blood pressure above 70 mmHg, and mean arterial Po2 and arterial Pco2 in the range of 90–100 Torr and 30–40 Torr, respectively. Imaged lungs did not show any signs of acute lung injury or edema. Following infusion of FITC dextran, subpleural pulmonary arterioles and venules of up to 50-μm diameter and alveolar capillary networks could be visualized during successive expiratory plateau phases over a period of at least 2 h. Vasoconstrictive responses to hypoxia (11% O2) or infusion of the thromboxane analog U-46619 were prominent in medium-sized arterioles (30- to 50-μm diameter), minor in small arterioles <30 μm, and absent in venules. The presented IVM model may constitute a powerful new tool for investigations of pulmonary microvascular responses in mice.
Collapse
|