1
|
Immunohistochemical Study of Smooth Muscle Cells and Elastin in Goose Lungs. FOLIA VETERINARIA 2022. [DOI: 10.2478/fv-2022-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
There are many differences (morphological, physiological and mechanical) between the lungs of birds and the lungs of mammals. Birds have a more efficient exchange of oxygen and carbon dioxide than mammals. In this article, we studied the presence of four antibodies (actin, α-smooth muscle actin, desmin and elastin) in the lungs of geese. Smooth muscle cells (SMCs) immunoreactive to actin, α-SMA and desmin were observed in the primary and secondary bronchi and arranged as a continuous layer. In the tertiary bronchus (parabronchus), immunoreactive cells on α-SMA and desmin were observed as aggregations of smooth muscle cells in the septum tips in atrial opening. A small number of α-SMA and desmin-positive cells were observed on the periphery of the parabronchi and between the air and blood capillaries. The elastic fibres were found in the large bronchi in connection with smooth muscle bands. In the parabronchi the elastic fibres form an elastic membrane lining the parabronchial lumen. In the blood vessels, the elastic fibres form the inner and outer elastic membrane. The individual elastic membranes connect neighbouring blood vessels.
Collapse
|
2
|
Yu L, Qiu C, Chen R. A narrative review of research advances in the study of molecular markers of airway smooth muscle cells. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:375. [PMID: 35434039 PMCID: PMC9011254 DOI: 10.21037/atm-22-800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/16/2022] [Indexed: 11/06/2022]
Abstract
Background and Objective Airway smooth muscle cells (ASMCs) are an important component of the airway. Their thickening and proliferation are important in pathological situations, such as airway remodeling in asthma, but their origin remains unclear. Therefore, characterizing molecular markers of ASMCs were sought to identify the source of increased ASMCs in asthmatic airway remodeling. Methods Articles for this review were derived from a review of the literature related to surface markers and biological properties of ASMCs and smooth muscle cells (SMCs) using PubMed, Google Scholar, and Web of Science. Key Content and Findings This review discusses several SMC molecular markers, describes the different developmental stages of SMCs that express different molecular markers, and summarizes several classical SMC molecular markers. However, the establishment of a specific molecular marker detection system for ASMCs still faces great challenges. Conclusions Although there is no recognized molecular marker detection system for ASMCs, and the study of the properties and sources of increased ASMCs in asthma airway remodeling is still in a state of exploration, the future is promising. Among the SMC markers described in this review, Myosin heavy chain 11 (MYH11) is a molecular marker for mature SMCs and Transgelin (TAGLN) is an early marker for SMC differentiation, and different molecular markers or combinations of molecular markers can be selected for the identification of the properties and sources of increased ASMCs in asthma airway remodeling according to the differentiation period and research needs.
Collapse
Affiliation(s)
- Li Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Rongchang Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| |
Collapse
|
3
|
Rippa AL, Alpeeva EV, Vasiliev AV, Vorotelyak EA. Alveologenesis: What Governs Secondary Septa Formation. Int J Mol Sci 2021; 22:ijms222212107. [PMID: 34829987 PMCID: PMC8618598 DOI: 10.3390/ijms222212107] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/30/2022] Open
Abstract
The simplification of alveoli leads to various lung pathologies such as bronchopulmonary dysplasia and emphysema. Deep insight into the process of emergence of the secondary septa during development and regeneration after pneumonectomy, and into the contribution of the drivers of alveologenesis and neo-alveolarization is required in an efficient search for therapeutic approaches. In this review, we describe the formation of the gas exchange units of the lung as a multifactorial process, which includes changes in the actomyosin cytoskeleton of alveocytes and myofibroblasts, elastogenesis, retinoic acid signaling, and the contribution of alveolar mesenchymal cells in secondary septation. Knowledge of the mechanistic context of alveologenesis remains incomplete. The characterization of the mechanisms that govern the emergence and depletion of αSMA will allow for an understanding of how the niche of fibroblasts is changing. Taking into account the intense studies that have been performed on the pool of lung mesenchymal cells, we present data on the typing of interstitial fibroblasts and their role in the formation and maintenance of alveoli. On the whole, when identifying cell subpopulations in lung mesenchyme, one has to consider the developmental context, the changing cellular functions, and the lability of gene signatures.
Collapse
|
4
|
Rubin L, Stabler CT, Schumacher-Klinger A, Marcinkiewicz C, Lelkes PI, Lazarovici P. Neurotrophic factors and their receptors in lung development and implications in lung diseases. Cytokine Growth Factor Rev 2021; 59:84-94. [PMID: 33589358 DOI: 10.1016/j.cytogfr.2021.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
Abstract
Although lung innervation has been described by many studies in humans and rodents, the regulation of the respiratory system induced by neurotrophins is not fully understood. Here, we review current knowledge on the role of neurotrophins and the expression and function of their receptors in neurogenesis, vasculogenesis and during the embryonic development of the respiratory tree and highlight key implications relevant to respiratory diseases.
Collapse
Affiliation(s)
- Limor Rubin
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | - Collin T Stabler
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, USA.
| | - Adi Schumacher-Klinger
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| | - Cezary Marcinkiewicz
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, USA.
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, USA.
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| |
Collapse
|
5
|
McGowan S. Understanding the developmental pathways pulmonary fibroblasts may follow during alveolar regeneration. Cell Tissue Res 2017; 367:707-719. [PMID: 28062913 DOI: 10.1007/s00441-016-2542-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/19/2016] [Indexed: 10/20/2022]
Abstract
Although pulmonary alveolar interstitial fibroblasts are less specialized than their epithelial and endothelial neighbors, they play essential roles during development and in response to lung injury. At birth, they must adapt to the sudden mechanical changes imposed by the onset of respiration and to a higher ambient oxygen concentration. In diseases such as bronchopulmonary dysplasia and interstitial fibrosis, their adaptive responses are overwhelmed leading to compromised gas-exchange function. Thus, although fibroblasts do not directly participate in gas-exchange, they are essential for creating and maintaining an optimal environment at the alveolar epithelial-endothelial interface. This review summarizes new information and concepts about the ontogeny differentiation, and function of alveolar fibroblasts. Alveolar development will be emphasized, because the development of strategies to evoke alveolar repair and regeneration hinges on thoroughly understanding the way that resident fibroblasts populate specific locations in which extracellular matrix must be produced and remodeled. Other recent reviews have described the disruption that diseases cause to the fibroblast niche and so my objective is to illustrate how the unique developmental origins and differentiation pathways could be harnessed favorably to augment certain fibroblast subpopulations and to optimize the conditions for alveolar regeneration.
Collapse
Affiliation(s)
- Stephen McGowan
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA. .,Division of Pulmonary, Critical Care, and Occupational Medicine, C33B GH, Department of Internal Medicine, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| |
Collapse
|
6
|
Swonger JM, Liu JS, Ivey MJ, Tallquist MD. Genetic tools for identifying and manipulating fibroblasts in the mouse. Differentiation 2016; 92:66-83. [PMID: 27342817 PMCID: PMC5079827 DOI: 10.1016/j.diff.2016.05.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/18/2023]
Abstract
The use of mouse genetic tools to track and manipulate fibroblasts has provided invaluable in vivo information regarding the activities of these cells. Recently, many new mouse strains have been described for the specific purpose of studying fibroblast behavior. Colorimetric reporter mice and lines expressing Cre are available for the study of fibroblasts in the organs prone to fibrosis, including heart, kidney, liver, lung, and skeletal muscle. In this review we summarize the current state of the models that have been used to define tissue resident fibroblast populations. While these complex genetic reagents provide unique insights into the process of fibrosis, they also require a thorough understanding of the caveats and limitations. Here, we discuss the specificity and efficiency of the available genetic models and briefly describe how they have been used to document the mechanisms of fibrosis.
Collapse
Affiliation(s)
- Jessica M Swonger
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Jocelyn S Liu
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Malina J Ivey
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Michelle D Tallquist
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| |
Collapse
|
7
|
Yamada M, Kurihara H, Kinoshita K, Sakai T. Temporal Expression of Alpha–Smooth Muscle Actin and Drebrin in Septal Interstitial Cells during Alveolar Maturation. J Histochem Cytochem 2016; 53:735-44. [PMID: 15928322 DOI: 10.1369/jhc.4a6483.2005] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In rat lung, the definitive alveoli are established during development by the outgrowth of secondary septa from the primary septa present in newborn; however, the mechanism of alveolar formation has not yet been fully clarified. In this study, we characterize the septal interstitial cells in developing alveoli. During the perinatal period, alpha-SMA–containing slender cells were found in the primitive alveolar septa. Alpha-SMA–containing cells were detected at the tips of the septa until postnatal day 21, when the alveolar formation was almost completed, but disappeared in adult. Immunoelectron microscopy demonstrated that alpha-SMA is localized mainly in the cellular protrusions, which are connected with the elastic fibers around the interstitial cells. Developmentally regulated brain protein (drebrin) is also located in the cell extensions containing alpha-SMA in immature alveolar interstitial cells. In adult lung, alpha-SMA–positive cells are located only at the alveolar ducts but are not found in the secondary septa. Desmin is expressed only in alpha-SMA–containing cells at the alveolar ducts but not in those at the tip of alveolar septa. These results suggest that a part of the septal interstitial cells are temporarily alpha-SMA– and drebrin-positive during maturation. Alpha-SMA– and drebrin-containing septal interstitial cells (termed septal myofibroblast-like cells) may play an important role in alveolar formation.
Collapse
Affiliation(s)
- Mie Yamada
- Department of Anatomy, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | | | | | | |
Collapse
|
8
|
Vrhovski B, McKay K, Schevzov G, Gunning PW, Weinberger RP. Smooth Muscle-specific α Tropomyosin Is a Marker of Fully Differentiated Smooth Muscle in Lung. J Histochem Cytochem 2016; 53:875-83. [PMID: 15995146 DOI: 10.1369/jhc.4a6504.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tropomyosin (Tm) is one of the major components of smooth muscle. Currently it is impossible to easily distinguish the two major smooth muscle (sm) forms of Tm at a protein level by immunohistochemistry due to lack of specific antibodies. α-sm Tm contains a unique 2a exon not found in any other Tm. We have produced a polyclonal antibody to this exon that specifically detects α-sm Tm. We demonstrate here the utility of this antibody for the study of smooth muscle. The tissue distribution of α-sm Tm was shown to be highly specific to smooth muscle. α-sm Tm showed an identical profile and tissue colocalization with α-sm actin both by Western blotting and immunohistochemistry. Using lung as a model organ system, we examined the developmental appearance of α-sm Tm in comparison to α-sm actin in both the mouse and human. α-sm Tm is a late-onset protein, appearing much later than actin in both species. There were some differences in onset of appearance in vascular and airway smooth muscle with airway appearing earlier. α-sm Tm can therefore be used as a good marker of mature differentiated smooth muscle cells. Along with α-sm actin and sm-myosin antibodies, α-sm Tm is a valuable tool for the study of smooth muscle.
Collapse
Affiliation(s)
- Bernadette Vrhovski
- The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia
| | | | | | | | | |
Collapse
|
9
|
Branchfield K, Li R, Lungova V, Verheyden JM, McCulley D, Sun X. A three-dimensional study of alveologenesis in mouse lung. Dev Biol 2015; 409:429-41. [PMID: 26632490 DOI: 10.1016/j.ydbio.2015.11.017] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 11/23/2015] [Accepted: 11/23/2015] [Indexed: 01/08/2023]
Abstract
Alveologenesis is the final step of lung maturation, which subdivides the alveolar region of the lung into smaller units called alveoli. Each of the nascent dividers serves as a new gas-exchange surface, and collectively they drastically increase the surface area for breathing. Disruption of alveologenesis results in simplification of alveoli, as is seen in premature infants diagnosed with bronchopulmonary dysplasia (BPD), a prevalent lung disease that is often associated with lifelong breathing deficiencies. To date, a majority of studies of alveologenesis rely on two-dimensional (2D) analysis of tissue sections. Given that an overarching theme of alveologenesis is thinning and extension of the epithelium and mesenchyme to facilitate gas exchange, often only a small portion of a cell or a cellular structure is represented in a single 2D plane. Here, we use a three-dimensional (3D) approach to examine the structural architecture and cellular composition of myofibroblasts, alveolar type 2 cells, elastin and lipid droplets in normal as well as BPD-like mouse lung. We found that 2D finger-like septal crests, commonly used to depict growing alveolar septae, are often artifacts of sectioning through fully established alveolar walls. Instead, a more accurate representation of growing septae are 3D ridges that are lined by platelet-derived growth factor receptor alpha (PDGFRA) and alpha smooth muscle actin (α-SMA)-expressing myofibroblasts, as well as the elastin fibers that they produce. Accordingly in 3D, both α-SMA and elastin were each found in connected networks underlying the 3D septal ridges rather than as isolated dots at the tip of 2D septal crests. Analysis through representative stages of alveologenesis revealed unappreciated dynamic changes in these patterns. PDGFRA-expressing cells are only α-SMA-positive during the first phase of alveologenesis, but not in the second phase, suggesting that the two phases of septae formation may be driven by distinct mechanisms. Thin elastin fibers are already present in the alveolar region prior to alveologenesis, suggesting that during alveologenesis, there is not only new elastin deposition, but also extensive remodeling to transform thin and uniformly distributed fibers into thick cables that rim the nascent septae. Analysis of several genetic as well as hyperoxia-induced models of BPD revealed that the myofibroblast organization is perturbed in all, regardless of whether the origin of defect is epithelial, mesenchymal, endothelial or environmental. Finally, analysis of relative position of PDGFRA-positive cells and alveolar type 2 cells reveal that during alveologenesis, these two cell types are not always adjacent to one another. This result suggests that the niche and progenitor relationship afforded by their close juxtaposition in the adult lung may be a later acquired property. These insights revealed by 3D reconstruction of the septae set the foundation for future investigations of the mechanisms driving normal alveologenesis, as well as causes of alveolar simplification in BPD.
Collapse
Affiliation(s)
- Kelsey Branchfield
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States
| | - Rongbo Li
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States
| | - Vlasta Lungova
- Department of Surgery, University of Wisconsin-Madison Madison, WI 53706, United States
| | - Jamie M Verheyden
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States
| | - David McCulley
- Department of Pediatrics University of Wisconsin-Madison Madison, WI 53706, United States
| | - Xin Sun
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States.
| |
Collapse
|
10
|
Pieretti AC, Ahmed AM, Roberts JD, Kelleher CM. A novel in vitro model to study alveologenesis. Am J Respir Cell Mol Biol 2014; 50:459-69. [PMID: 24066869 DOI: 10.1165/rcmb.2013-0056oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Many pediatric pulmonary diseases are associated with significant morbidity and mortality due to impairment of alveolar development. The lack of an appropriate in vitro model system limits the identification of therapies aimed at improving alveolarization. Herein, we characterize an ex vivo lung culture model that facilitates investigation of signaling pathways that influence alveolar septation. Postnatal Day 4 (P4) mouse pup lungs were inflated with 0.4% agarose, sliced, and cultured within a collagen matrix in medium that was optimized to support cell proliferation and promote septation. Lung slices were grown with and without 1D11, an active transforming growth factor-β-neutralizing antibody. After 4 days, the lung sections (designated P4 + 4) and noncultured lung sections were examined using quantitative morphometry to assess alveolar septation and immunohistochemistry to evaluate cell proliferation and differentiation. We observed that the P4 + 4 lung sections exhibited ex vivo alveolarization, as evidenced by an increase in septal density, thinning of septal walls, and a decrease in mean linear intercept comparable to P8, age-matched, uncultured lungs. Moreover, immunostaining showed ongoing cell proliferation and differentiation in cultured lungs that were similar to P8 controls. Cultured lungs exposed to 1D11 had a distinct phenotype of decreased septal density when compared with untreated P4 + 4 lungs, indicating the utility of investigating signaling in these lung slices. These results indicate that this novel lung culture system is optimized to permit the investigation of pathways involved in septation, and potentially the identification of therapeutic targets that enhance alveolarization.
Collapse
Affiliation(s)
- Alberto C Pieretti
- 1 Department of Pediatric Surgery, MassGeneral Hospital for Children, Boston, Massachusetts
| | | | | | | |
Collapse
|
11
|
Bachiller PR, Cornog KH, Kato R, Buys ES, Roberts JD. Soluble guanylate cyclase modulates alveolarization in the newborn lung. Am J Physiol Lung Cell Mol Physiol 2013; 305:L569-81. [PMID: 23934926 DOI: 10.1152/ajplung.00401.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide (NO) regulates lung development through incompletely understood mechanisms. NO controls pulmonary vascular smooth muscle cell (SMC) differentiation largely through stimulating soluble guanylate cyclase (sGC) to produce cGMP and increase cGMP-mediated signaling. To examine the role of sGC in regulating pulmonary development, we tested whether decreased sGC activity reduces alveolarization in the normal and injured newborn lung. For these studies, mouse pups with gene-targeted sGC-α1 subunit truncation were used because we determined that they have decreased pulmonary sGC enzyme activity. sGC-α1 knockout (KO) mouse pups were observed to have decreased numbers of small airway structures and lung volume compared with wild-type (WT) mice although lung septation and body weights were not different. However, following mild lung injury caused by breathing 70% O2, the sGC-α1 KO mouse pups had pronounced inhibition of alveolarization, as evidenced by an increase in airway mean linear intercept, reduction in terminal airway units, and decrease in lung septation and alveolar openings, as well as reduced somatic growth. Because cGMP regulates SMC phenotype, we also tested whether decreased sGC activity reduces lung myofibroblast differentiation. Cellular markers revealed that vascular SMC differentiation decreased, whereas myofibroblast activation increased in the hyperoxic sGC-α1 KO pup lung. These results indicate that lung development, particularly during hyperoxic injury, is impaired in mouse pups with diminished sGC activity. These studies support the investigation of sGC-targeting agents as therapies directed at improving development in the newborn lung exposed to injury.
Collapse
Affiliation(s)
- Patricia R Bachiller
- Jr., Cardiovascular Research Center, Massachusetts General Hospital - East, 149 13 St., Charlestown, MA 02129.
| | | | | | | | | |
Collapse
|
12
|
Ahlfeld SK, Gao Y, Wang J, Horgusluoglu E, Bolanis E, Clapp DW, Conway SJ. Periostin downregulation is an early marker of inhibited neonatal murine lung alveolar septation. ACTA ACUST UNITED AC 2013; 97:373-85. [PMID: 23723163 DOI: 10.1002/bdra.23149] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/12/2013] [Accepted: 04/18/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Extreme preterm birth exposes the saccular lung to multiple teratogens, which ultimately retard alveolar development. Specifically, therapeutic high level oxygen supplementation adversely affects the premature lungs and results in blunted alveolarization. Prolonged hyperoxic lung injury has previously been shown to upregulate the matricellular protein Periostin (Postn) and stimulate ectopic accumulation of alpha smooth muscle actin (αSMA) myofibroblasts. Therapies that promote lung septation are lacking largely due to a lack of reliable early biomarkers of injury. Thus, we determined if Postn expression correlated with the initial appearance of myofibroblasts in the saccular lung and was required for early alveolar development. METHODS Lung development in C57BL/6J mice following room-air (RA, 21%-O₂) or continuous hyperoxia (85%-O₂) from birth (P0) through postnatal day P14 was correlated with Postn and αSMA expression. Alveolarization in Postn knockout mice exposed to room-air, 60%-, and 85%-O₂ was also examined. RESULTS Postn was widely expressed in distal lung septa through P2 to P4 and peak expression coincided with accumulation of saccular myofibroblasts. Initially, 85%-O₂ prematurely downregulated Postn and αSMA expression and suppressed proliferation before the first evidence of distal lung simplification at P4. By P14, chronic 85%-O₂ resulted in secondary upregulation of Postn and αSMA in blunted septa. Myofibroblast differentiation and alveolar development was unaffected in Postn null mice and acute 85%-O₂ exposure equally inhibited septal formation in Postn null and wild-type littermates. CONCLUSION Postn expression is tightly correlated with the presence of αSMA-myofibroblasts and is a novel early biomarker of acutely inhibited alveolar septation during a crucial window of lung development.
Collapse
Affiliation(s)
- Shawn K Ahlfeld
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
13
|
Capelari DN, Sánchez SI, Ortega HH, Ciuffo GM, Fuentes LB. Effects of maternal captopril treatment during late pregnancy on neonatal lung development in rats. ACTA ACUST UNITED AC 2012; 177:97-106. [DOI: 10.1016/j.regpep.2012.05.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 03/10/2012] [Accepted: 05/05/2012] [Indexed: 11/28/2022]
|
14
|
Manoli SE, Smith LA, Vyhlidal CA, An CH, Porrata Y, Cardoso WV, Baron RM, Haley KJ. Maternal smoking and the retinoid pathway in the developing lung. Respir Res 2012; 13:42. [PMID: 22651576 PMCID: PMC3479035 DOI: 10.1186/1465-9921-13-42] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 04/30/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Maternal smoking is a risk factor for pediatric lung disease, including asthma. Animal models suggest that maternal smoking causes defective alveolarization in the offspring. Retinoic acid signaling modulates both lung development and postnatal immune function. Thus, abnormalities in this pathway could mediate maternal smoking effects. We tested whether maternal smoking disrupts retinoic acid pathway expression and functioning in a murine model. METHODS Female C57Bl/6 mice with/without mainstream cigarette smoke exposure (3 research cigarettes a day, 5 days a week) were mated to nonsmoking males. Cigarette smoke exposure continued throughout the pregnancy and after parturition. Lung tissue from the offspring was examined by mean linear intercept analysis and by quantitative PCR. Cell culture experiments using the type II cell-like cell line, A549, tested whether lipid-soluble cigarette smoke components affected binding and activation of retinoic acid response elements in vitro. RESULTS Compared to tobacco-naïve mice, juvenile mice with tobacco toxin exposure had significantly (P < 0.05) increased mean linear intercepts, consistent with an alveolarization defect. Tobacco toxin exposure significantly (P < 0.05) decreased mRNA and protein expression of retinoic acid signaling pathway elements, including retinoic acid receptor alpha and retinoic acid receptor beta, with the greatest number of changes observed between postnatal days 3-5. Lipid-soluble cigarette smoke components significantly (P < 0.05) decreased retinoic acid-induced binding and activation of the retinoic acid receptor response element in A549 cells. CONCLUSIONS A murine model of maternal cigarette smoking causes abnormal alveolarization in association with altered retinoic acid pathway element expression in the offspring. An in vitro cell culture model shows that lipid-soluble components of cigarette smoke decrease retinoic acid response element activation. It is feasible that disruption of retinoic acid signaling contributes to the pediatric lung dysfunction caused by maternal smoking.
Collapse
Affiliation(s)
- Sara E Manoli
- Department of Medicine, Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Chen L, Acciani T, Le Cras T, Lutzko C, Perl AKT. Dynamic regulation of platelet-derived growth factor receptor α expression in alveolar fibroblasts during realveolarization. Am J Respir Cell Mol Biol 2012; 47:517-27. [PMID: 22652199 DOI: 10.1165/rcmb.2012-0030oc] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Although the importance of platelet-derived growth factor receptor (PDGFR)-α signaling during normal alveogenesis is known, it is unclear whether this signaling pathway can regulate realveolarization in the adult lung. During alveolar development, PDGFR-α-expressing cells induce α smooth muscle actin (α-SMA) and differentiate to interstitial myofibroblasts. Fibroblast growth factor (FGF) signaling regulates myofibroblast differentiation during alveolarization, whereas peroxisome proliferator-activated receptor (PPAR)-γ activation antagonizes myofibroblast differentiation in lung fibrosis. Using left lung pneumonectomy, the roles of FGF and PPAR-γ signaling in differentiation of myofibroblasts from PDGFR-α-positive precursors during compensatory lung growth were assessed. FGF receptor (FGFR) signaling was inhibited by conditionally activating a soluble dominant-negative FGFR2 transgene. PPAR-γ signaling was activated by administration of rosiglitazone. Changes in α-SMA and PDGFR-α protein expression were assessed in PDGFR-α-green fluorescent protein (GFP) reporter mice using immunohistochemistry, flow cytometry, and real-time PCR. Immunohistochemistry and flow cytometry demonstrated that the cell ratio and expression levels of PDGFR-α-GFP changed dynamically during alveolar regeneration and that α-SMA expression was induced in a subset of PDGFR-α-GFP cells. Expression of a dominant-negative FGFR2 and administration of rosiglitazone inhibited induction of α-SMA in PDGFR-α-positive fibroblasts and formation of new septae. Changes in gene expression of epithelial and mesenchymal signaling molecules were assessed after left lobe pneumonectomy, and results demonstrated that inhibition of FGFR2 signaling and increase in PPAR-γ signaling altered the expression of Shh, FGF, Wnt, and Bmp4, genes that are also important for epithelial-mesenchymal crosstalk during early lung development. Our data demonstrate for the first time that a comparable epithelial-mesenchymal crosstalk regulates fibroblast phenotypes during alveolar septation.
Collapse
Affiliation(s)
- Leiling Chen
- Division of Pulmonary Biology, Perinatal Institute, Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | | | | | | | |
Collapse
|
16
|
Kikuchi A, Kishi A, Yamamoto M, Yamane S, Umezawa T, Ide Y, Abe S. Expression of Intermediate Filaments in the Development of Genioglossus Muscle. J HARD TISSUE BIOL 2012. [DOI: 10.2485/jhtb.21.421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
17
|
Rawlins EL, Perl AK. The a"MAZE"ing world of lung-specific transgenic mice. Am J Respir Cell Mol Biol 2011; 46:269-82. [PMID: 22180870 DOI: 10.1165/rcmb.2011-0372ps] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The purpose of this review is to give a comprehensive overview of transgenic mouse lines suitable for studying gene function and cellular lineage relationships in lung development, homeostasis, injury, and repair. Many of the mouse strains reviewed in this Perspective have been widely shared within the lung research community, and new strains are continuously being developed. There are many transgenic lines that target subsets of lung cells, but it remains a challenge for investigators to select the correct transgenic modules for their experiment. This review covers the tetracycline- and tamoxifen-inducible systems and focuses on conditional lines that target the epithelial cells. We point out the limitations of each strain so investigators can choose the system that will work best for their scientific question. Current mesenchymal and endothelial lines are limited by the fact that they are not lung specific. These lines are summarized in a brief overview. In addition, useful transgenic reporter mice for studying lineage relationships, promoter activity, and signaling pathways will complete our lung-specific conditional transgenic mouse shopping list.
Collapse
Affiliation(s)
- Emma L Rawlins
- Children's Hospital Medical Center, Divisions of Neonatology and Pulmonary Biology, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | | |
Collapse
|
18
|
Yun EJ, Vu TH. mSmile is necessary for bronchial smooth muscle and alveolar myofibroblast development. Anat Rec (Hoboken) 2011; 295:167-76. [PMID: 21956870 DOI: 10.1002/ar.21475] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 05/31/2011] [Accepted: 07/06/2011] [Indexed: 11/10/2022]
Abstract
Disrupted lung alveolar myofibroblast and bronchial smooth muscle (BSM) cell development may lead to pulmonary disorders such as bronchopulmonary dysplasia. The molecular mechanisms that regulate BSM and alveolar myofibroblast development are not fully understood. Here we show that mSmile (murine Smile), a novel transmembrane protein with tetratricopeptide repeats, functions in lung alveolar myofibroblast and BSM cell development. mSmile mutant mice exhibit early neonatal lethality with few mice surviving up to 3 weeks. Mutant lungs display both airway branching morphogenesis defect during fetal lung development and alveolarization defect after birth. These defects are associated with reduced numbers of BSM cells in the peribronchial subepithelial region and clefts and myofibroblasts in alveolar septae. Expression of fibroblast growth factor-10 and its down stream target Bmp-4, which are important for BSM formation, is decreased. In vitro, mSmile mutant embryonic fibroblasts show reduced receptor activation and induction of myofibroblast formation in response to Transforming growth factor-β (Tgf-β), indicating that mSmile may mediate myofibroblast development through modulation of Tgf-β signaling. These studies identify mSmile as a novel gene specifying both the BSM and lung alveolar myofibroblast lineages, contributing to our understanding of the biological control of the development of these cells, and may provide insights into the aberrant smooth muscle and alveolar myofibroblast development that occur in pathological conditions.
Collapse
Affiliation(s)
- Eun Jun Yun
- Department of Medicine and Lung Biology Center, University of California, San Francisco, 94143-2911, USA
| | | |
Collapse
|
19
|
McGowan SE, McCoy DM. Fibroblasts expressing PDGF-receptor-alpha diminish during alveolar septal thinning in mice. Pediatr Res 2011; 70:44-9. [PMID: 21659960 DOI: 10.1203/pdr.0b013e31821cfb5a] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In mice, secondary alveolar septal formation primarily occurs during a brief postnatal period and is accompanied by transient expansion of the interstitial lung fibroblast (LF) population. PDGF-A, which solely signals through PDGF-receptor-alpha (PDGF-Rα), is required for expansion, but the receptor's relevant downstream targets remain incompletely defined. We have evaluated the proliferation, apoptosis, and differential response to the selective protein tyrosine kinase inhibitor, imatinib, by pdgfrα-expressing LF (pdgfrα-LF) and compared them with their nonexpressing LF counterparts. Our objective was to determine whether diminished signaling through PDGF-Rα-mediated pathways regulates the decline in myofibroblasts, which accompanies septal thinning and ensures more efficient alveolar gas exchange. Using quantitative stereology and flow cytometry at postnatal d 12 and 14, we observed that imatinib caused a selective suppression of proliferation and an increase in apoptosis. The number of the alpha smooth muscle actin (αSMA) producing pdgfrα-LF was also reduced. Using cultures of neonatal mouse LF, we showed that imatinib did not suppress PDGF-Rα gene expression but reduced PDGF-A-mediated Akt phosphorylation, potentially explaining the increase in apoptosis. Our findings are relevant to bronchopulmonary dysplasia in which positive pressure ventilation interferes with myofibroblast depletion, septal thinning, and capillary maturation.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA.
| | | |
Collapse
|
20
|
Six1 transcription factor is critical for coordination of epithelial, mesenchymal and vascular morphogenesis in the mammalian lung. Dev Biol 2011; 353:242-58. [PMID: 21385574 DOI: 10.1016/j.ydbio.2011.02.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 02/23/2011] [Accepted: 02/28/2011] [Indexed: 01/12/2023]
Abstract
Six1 is a member of the six-homeodomain family of transcription factors. Six1 is expressed in multiple embryonic cell types and plays important roles in proliferation, differentiation and survival of precursor cells of different organs, yet its function during lung development was hitherto unknown. Herein we show that Six1(-/-) lungs are severely hypoplastic with greatly reduced epithelial branching and increased mesenchymal cellularity. Six1 is expressed at the distal epithelial tips of branching tubules as well as in the surrounding distal mesenchyme. Six1(-/-) lung epithelial cells show increased expression of differentiation markers, but loss of progenitor cell markers. Six1 overexpression in MLE15 lung epithelial cells in vitro inhibited cell differentiation, but increases the expression of progenitor cell markers. In addition, Six1(-/-) embryos and newborn mice exhibit mesenchymal overproliferation, decreased Fgf10 expression and severe defects in the smooth muscle component of the bronchi and major pulmonary vessels. These defects lead to rupture of major vessels in mutant lungs after birth. Treatment of Six1(-/-) epithelial explants in culture with recombinant Fgf10 protein restores epithelial branching. As Shh expression is abnormally increased in Six1(-/-) lungs, we also treated mutant mesenchymal explants with recombinant Shh protein and found that these explants were competent to respond to Shh and continued to grow in culture. Furthermore, inhibition of Shh signaling with cyclopamine stimulated Six1(-/-) lungs to grow and branch in culture. This study provides the first evidence for the requirement of Six1 in coordinating Shh-Fgf10 signaling in embryonic lung to ensure proper levels of proliferation and differentiation along the proximodistal axis of epithelial, mesenchymal and endothelial cells. These findings uncover novel and essential functions for Six1 as a critical coordinator of Shh-Fgf10 signaling during embryonic lung development. We propose that Six1 is hence critical for coordination of proper lung epithelial, mesenchymal and vascular development.
Collapse
|
21
|
Abstract
BACKGROUND It has been reported that the smooth muscles in fetal airways exhibit spontaneous phasic contractions throughout gestation. However, the mechanism of these spontaneous contractions is unknown. In the bowel wall, interstitial cells of Cajal (ICCs), which are derived from c-kit positive precursor cells, play an important role as pacemaker cells responsible for the spontaneous, rhythmic activity in the smooth muscle cells. In this study, we investigated the spatial and temporal expression patterns of c-kit positive cells in the embryonic lung and its relationship to the smooth muscle cells surrounding the trachea and the bronchus. METHODS Rat fetuses were removed from timed pregnant dams on embryonic days (E) 11.5, 13.5, 15.5, and 17.5. Immunohistochemical studies with anti c-kit antibody and anti α-SMA antibody were carried out using frozen sections. RESULTS A small number of c-kit positive cells were observed in the mesenchyme of the lung bud on day E 11.5. They were markedly increased in number on day E 13.5. On day E 15.5 and on day E 17.5, strong c-kit expressions were observed on the vascular wall and moderate expressions in the mesenchyme. C-kit positive cells co-localized with α-SMA positive smooth muscle cells surrounding the airway epithelium. CONCLUSION Co-localization of c-kit positive cells and airway smooth muscles in the fetal lung suggests that c-kit positive cells may play an important role in the spontaneous contractions of fetal airways. C-kit expressions in the fetal pulmonary vascular wall suggest that these cells may play an important role in vasculogenesis and angiogenesis of the embryonic lung.
Collapse
|
22
|
El-Hashash AHK, Al Alam D, Turcatel G, Bellusci S, Warburton D. Eyes absent 1 (Eya1) is a critical coordinator of epithelial, mesenchymal and vascular morphogenesis in the mammalian lung. Dev Biol 2010; 350:112-26. [PMID: 21129374 DOI: 10.1016/j.ydbio.2010.11.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 11/10/2010] [Accepted: 11/19/2010] [Indexed: 01/12/2023]
Abstract
The proper level of proliferation and differentiation along the proximodistal axis is crucial for lung organogenesis. Elucidation of the factors that control these processes will therefore provide important insights into embryonic lung development and regeneration. Eya1 is a transcription factor/protein phosphatase that regulates cell lineage specification and proliferation. Yet its functions during lung development are unknown. In this paper we show that Eya1(-/-) lungs are severely hypoplastic with reduced epithelial branching and increased mesenchymal cellularity. Eya1 is expressed at the distal epithelial tips of branching tubules as well as in the surrounding distal mesenchyme. Eya1(-/-) lung epithelial cells show loss of progenitor cell markers with increased expression of differentiation markers and cell cycle exit. In addition, Eya1(-/-) embryos and newborn mice exhibit severe defects in the smooth muscle component of the bronchi and major pulmonary vessels with decreased Fgf10 expression. These defects lead to rupture of the major vessels and hemorrhage into the lungs after birth. Treatment of Eya1(-/-) epithelial explants in culture with recombinant Fgf10 stimulates epithelial branching. Since Shh expression and activity are abnormally increased in Eya1(-/-) lungs, we tested whether genetically lowering Shh activity could rescue the Eya1(-/-) lung phenotype. Indeed, genetic reduction of Shh partially rescues Eya1(-/-) lung defects while restoring Fgf10 expression. This study provides the first evidence that Eya1 regulates Shh signaling in embryonic lung, thus ensuring the proper level of proliferation and differentiation along the proximodistal axis of epithelial, mesenchymal and endothelial cells. These findings uncover novel functions for Eya1 as a critical upstream coordinator of Shh-Fgf10 signaling during embryonic lung development. We conclude, therefore, that Eya1 function is critical for proper coordination of lung epithelial, mesenchymal and vascular development.
Collapse
Affiliation(s)
- Ahmed H K El-Hashash
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Keck School of Medicine of University of Southern California, 4650 Sunset Boulevard MS35, Los Angeles, CA 90027, USA
| | | | | | | | | |
Collapse
|
23
|
Singh SR, Billington CK, Sayers I, Hall IP. Can lineage-specific markers be identified to characterize mesenchyme-derived cell populations in the human airways? Am J Physiol Lung Cell Mol Physiol 2010; 299:L169-83. [PMID: 20435685 DOI: 10.1152/ajplung.00311.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchyme-derived cells in the airway wall including airway smooth muscle cells, fibroblasts, and myofibroblasts are known to play important roles in airway remodeling. The lack of specific phenotypical markers makes it difficult to define these cell populations in primary cultures. Most relevant studies to date have used animal airway tissues, vascular tissues, or transformed cell lines with only limited studies attempting to phenotypically characterize human airway mesenchymal cells. The objectives of this study were to evaluate reported markers and identify novel markers to define these cell types. We could not identify any specific marker to define these cell populations in vitro that permitted unequivocal identification using immunocytochemistry. However, characteristic filamentous alpha-smooth muscle actin distribution was observed in a significant ( approximately 25%) proportion of human airway smooth muscle cells, whereas this was not observed in airway fibroblasts. A significantly higher proportion of airway fibroblasts expressed alpha(1)- and alpha(2)-integrin receptors compared with human airway smooth muscle cells as assessed by fluorescence activated cell sorting. Global gene expression profiling identified aldo-keto reductase 1C3 (AKR1C3) and cathepsin K as being novel markers to define airway smooth muscle cells, whereas integrin-alpha(8) (ITGA8) and thromboxane synthase 1 (TBXAS1) were identified as novel airway fibroblast-specific markers, and these findings were validated by RT-PCR. Ex vivo studies in human airway tissue sections identified high-molecular weight caldesmon and alpha-smooth muscle actin as being expressed in smooth muscle bundles, whereas ITGA8 and TBXAS1 were absent from these.
Collapse
|
24
|
IMAMURA T, ISHIZUKA O, YAMAMOTO T, GOTOH M, NISHIZAWA O. Bone Marrow-Derived Cells Implanted into Freeze-Injured Urinary Bladders Reconstruct Functional Smooth Muscle Layers. Low Urin Tract Symptoms 2010; 2:1-10. [DOI: 10.1111/j.1757-5672.2010.00066.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
25
|
Das S, Du Z, Bassly S, Singer L, Vicencio AG. Effects of chronic hypercapnia in the neonatal mouse lung and brain. Pediatr Pulmonol 2009; 44:176-82. [PMID: 19142892 DOI: 10.1002/ppul.20971] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Permissive hypercapnia is increasingly utilized in the care of premature infants to prevent bronchopulmonary dysplasia. In a previous investigation, we described gene expression changes in the neonatal mouse lung exposed to chronic hypercapnia that might contribute to lung protection and accelerated maturation. However, it is unknown whether chronic hypercapnia increases alveolar formation, nor if it has detrimental effects in other developing organs such as the brain. OBJECTIVE To determine whether chronic hypercapnia accelerates early alveolar formation and increases neuronal cell injury in the developing mouse lung and brain, respectively. DESIGN Mouse pups were exposed to 8% CO(2) + 21% O(2) starting at postnatal day (P) 2 until P7. Control animals were maintained in room air. Animals were sacrificed at P4 or P7, and lungs and brains were excised and analyzed. RESULTS Exposure to 8% CO(2) resulted in an increased expression of alpha-smooth muscle actin (alpha-sma) which localized to the tips of developing alveolar buds, and also an increased number of alveolar buds at P7. Importantly, hypercapnic animals also demonstrated evidence of increased TUNEL-positive cells in the brain. CONCLUSIONS Exposure to chronic hypercapnia may lead to early initiation of alveolar budding in the neonatal mouse, but may also lead to increased TUNEL-positive cells in the developing brain.
Collapse
Affiliation(s)
- Sumon Das
- Department of Pediatrics, Division of Critical Care Medicine, Albert Einstein College of Medicine and Children's Hospital at Montefiore, Bronx, New York 10467, USA
| | | | | | | | | |
Collapse
|
26
|
Shan L, Subramaniam M, Emanuel RL, Degan S, Johnston P, Tefft D, Warburton D, Sunday ME. Centrifugal migration of mesenchymal cells in embryonic lung. Dev Dyn 2008; 237:750-7. [PMID: 18297731 DOI: 10.1002/dvdy.21462] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Murine lung development begins at embryonic day (E) 9.5. Normal lung structure and function depend on the patterns of localization of differentiated cells. Pulmonary mesenchymal cell lineages have been relatively unexplored. Importantly, there has been no prior evidence of clonality of any lung cells. Herein we use a definitive genetic approach to demonstrate a common origin for proximal and distal pulmonary mesenchymal cells. A retroviral library with 3,400 unique inserts was microinjected into the airway lumen of E11.5 lung buds. After 7-11 days of culture, buds were stained for placental alkaline phosphatase (PLAP). Most PLAP+ cells are peribronchial smooth muscle cells, initially localized laterally near the hilum, then migrating down airways to the subpleural region. Laser-capture microdissection and polymerase chain reaction confirm the clonal identities of PLAP+ cells proximally and distally. Our observation of this fundamental process during lung development opens new avenues for investigation of maladaptive mesenchymal responses in lung diseases.
Collapse
Affiliation(s)
- Lin Shan
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ha HY, Kim JB, Cho IH, Joo HJ, Kim KS, Lee KW, Sunwoo H, Im JY, Lee JK, Hong JH, Han PL. Morphogenetic lung defects of JSAP1-deficient embryos proceeds via the disruptions of the normal expressions of cytoskeletal and chaperone proteins. Proteomics 2008; 8:1071-80. [PMID: 18324732 DOI: 10.1002/pmic.200700815] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recent studies have shown that JNK/stress-activated protein kinase-associated protein 1 (JSAP1)-deficient mice die from respiratory failure shortly after birth. To understand the underlying mechanism, we investigated the histological appearances and cell type changes in developing jsap1(-/-) lungs between E12.5 and E18.5. At the light microscopic level, no overt abnormality was detected in jsap1(-/-) until E16.5. However, alveoli and airway formations that normally occur after E16.5 were poorly advanced in jsap1(-/-). Despite these morphological defects, surfactant secreting cells labeled by anti-SP-B or anti-SP-C were present in normal ranges in jsap1(-/-) lungs. Smooth muscle alpha-actin expressing cells were also developed in jsap1(-/-) lungs, although actin expression was decreased. The expressions of transcriptional factors, such as, nuclear factor Ib (Nfib), N-myc, and octamer transcriptional factor 1 (Oct-1), which play a critical role in lung morphogenesis, were found to be down-regulated, whereas signal transducer and activator of transcription 3 (Stat3), sonic hedgehog (Shh), and smoothened (Smo) were up-regulated, in jsap1(-/-) lungs at E17.5-E18.5 compared with those in jsap1(+/+) lungs. Proteomics analysis of E17.5 lung identified 39 proteins with altered expressions, which included actin, tropomyosin, myosin light chain, vimentin, heat shock protein (Hsp27), and Hsp84. These results suggest that JSAP1 is required for the normal expressions of cytoskeletal and chaperone proteins in the developing lung, and that impaired expressions of these proteins might cause morphogenetic defects observed in jsap1(-/-) lungs.
Collapse
Affiliation(s)
- Hye-Yeong Ha
- Division of Nano Sciences and Brain Disease Research Institute, Ewha Womans University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Imamura T, Kinebuchi Y, Ishizuka O, Seki S, Igawa Y, Nishizawa O. Implanted Mouse Bone Marrow-Derived Cells Reconstruct Layered Smooth Muscle Structures in Injured Urinary Bladders. Cell Transplant 2008; 17:267-78. [DOI: 10.3727/096368908784153850] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study is a preliminary investigation to determine if bone marrow-derived cells, when implanted into freeze-injured urinary bladders, differentiate into smooth muscle cells and reconstruct smooth muscle layers. Bone marrow cells were harvested from femurs of male ICR mice and cultured in collagen-coated dishes for 7 days. After 5 days of culture, the cells were transfected with green fluorescent protein (GFP) genes for identification in recipient tissues. Three days prior to implantation, the posterior urinary bladder walls of female nude mice were injured with an iron bar refrigerated by dry ice. Seven days after the culture and 3 days after the injury, adherent, proliferating GFP-labeled bone marrow-derived cells (1.0 × 105 cells) were implanted into the injured regions. For controls, a cell-free solution was injected. At 14 days after implantation, the experimental urinary bladders were analyzed by histological, gene expression, and cystometric investigations. Just prior to implantation, the injured regions did not have any smooth muscle layers. After 14 days, the implanted cells surviving in the recipient tissues were detected with GFP antibody. The implanted regions had distinct smooth muscle layers composed of regenerated smooth muscle marker-positive cells. The implanted GFP-labeled cells differentiated into smooth muscle cells that formed into layers. The differentiated cells contacted each other within the implanted region as well as smooth muscle cells of the host. As a result, the reconstructed smooth muscle layers were integrated into the host tissues. Control mice injected with cell-free solution developed only few smooth muscle cells and no layers. Cystometric investigations showed that mice with implanted the cells developed bladder contractions similar to normal mice, whereas control mice did not. In summary, mouse bone marrow-derived cells can reconstruct layered smooth muscle structures in injured bladders to remediate urinary dysfunction.
Collapse
Affiliation(s)
- Tetsuya Imamura
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yoshiaki Kinebuchi
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Osamu Ishizuka
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Satoshi Seki
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yasuhiko Igawa
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Osamu Nishizawa
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| |
Collapse
|
29
|
Nemati B, Atmodjo W, Gagnon S, Humes D, McKerlie C, Kaplan F, Sweezey NB. Glucocorticoid receptor disruption delays structural maturation in the lungs of newborn mice. Pediatr Pulmonol 2008; 43:125-33. [PMID: 18085690 DOI: 10.1002/ppul.20746] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In order to better understand the regulation of lung maturation by glucocorticoid-glucocorticoid receptor signaling, we studied glucocorticoid receptor (GR) hypomorphic mice with a mixed C57Bl6/129 sv background, in which disruption of exon 2 of the GR gene produces an N-terminal truncated GR protein. Four groups of mice were compared: homozygous mice that die at birth (non-survivors), homozygous mice that survive the neonatal period (survivors), heterozygotes and wild-type mice. Newborn non-survivors had 50% thicker airspace walls and a 46% decrease in the formation of secondary crests (the beginning of alveolar secondary septation) compared to either survivor or wild-type littermates (n = 9 mice in each group). The lung tissue to airspace ratio in homozygous mice not expressing wild-type GR (non-survivor and survivor) was increased compared to heterozygotes and wild-type mice that do express wild-type GR (0.91 +/- 0.08 vs. 0.49 +/- 0.02, n = 4 in each of the four subgroups), suggesting that complete morphological maturation of the lung is dependent on effective glucocorticoid signaling through a fully functional GR. Moreover, the relatively mature lung morphology of survivor versus non-survivor newborns suggests that a partial reduction in mesenchymal thickness is compatible with capillary remodeling, alveolar septation, and viable respiratory function after birth. Our findings suggest that in mice homozygous for disrupted GR, the severity of newborn respiratory insufficiency correlates with the degree of lung structural immaturity.
Collapse
Affiliation(s)
- Baharak Nemati
- Physiology and Experimental Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
30
|
Khan PA, Cloutier M, Piedboeuf B. Tracheal occlusion: A review of obstructing fetal lungs to make them grow and mature. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2007; 145C:125-38. [PMID: 17436297 DOI: 10.1002/ajmg.c.30127] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fetal lung growth and functional differentiation are affected strongly by the extent that pulmonary tissue is distended (expanded) by liquid that naturally fills developing future airspaces. Methods that prevent normal egress of this lung fluid through the trachea magnify mechanical stretching of lung parenchymal cells, thereby promoting lung development. Indeed, experimental observations demonstrate that in utero tracheal occlusion (TO) performed on fetuses during the late canalicular-early saccular stage potently stimulates pulmonary growth and maturation. In this review, we present the four principle non-human animal models of TO/obstruction and discuss them in relation to their utility in elucidating lung development, in remedying congenital diaphragmatic hernia (CDH) as well as in investigating the stretching effects on growth and remodeling of the fine vasculature.
Collapse
Affiliation(s)
- Paul A Khan
- CRCHUL Medical Research Centre, Laval University, Québec, Canada
| | | | | |
Collapse
|
31
|
Dickie R, Wang YT, Butler JP, Schulz H, Tsuda A. Distribution and Quantity of Contractile Tissue in Postnatal Development of Rat Alveolar Interstitium. Anat Rec (Hoboken) 2007; 291:83-93. [DOI: 10.1002/ar.20622] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Kim N, Vu TH. Parabronchial smooth muscle cells and alveolar myofibroblasts in lung development. ACTA ACUST UNITED AC 2006; 78:80-9. [PMID: 16622850 DOI: 10.1002/bdrc.20062] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Epithelial-mesenchymal interactions and extracellular matrix remodeling are key processes of embryonic lung development. Lung smooth muscle cells, which are derived from the mesenchyme, form a sheath around bronchi and blood vessels. During lung organogenesis, smooth muscle differentiation coincides with epithelial branching morphogenesis and closely follows developing airways spatially and temporally. The precise function of parabronchial smooth muscle (PBSM) cells in healthy adult lung remains unclear. However, PBSM may regulate epithelial branching morphogenesis during lung development by the induction of mechanical stress or through regulation of paracrine signaling pathways. Alveolar myofibroblasts are interstitial contractile cells that share features and may share an origin with smooth muscle cells. Alveolar myofibroblasts are essential for secondary septation, a process critical for the development of the gas-exchange region of the lung. Dysregulation of PBSM or alveolar myofibroblast development is thought to underlie the pathogenesis of many lung diseases, including bronchopulmonary dysplasia, asthma, and interstitial fibrosis. We review the current understanding of the regulation of PBSM and alveolar myofibroblast development, and discuss the role of PBSM in lung development. We specifically focus on the role of these cells in the context of fibroblast growth factor-10, sonic hedgehog, bone morphogenetic protein-4, retinoic acid, and Wnt signaling pathways in the regulation of lung branching morphogenesis.
Collapse
Affiliation(s)
- Namjin Kim
- Department of Medicine and Lung Biology Center, University of California, San Francisco, California 94143-2911, USA
| | | |
Collapse
|
33
|
Nadeau K, Jankov RP, Tanswell AK, Sweezey NB, Kaplan F. Lgl1 is suppressed in oxygen toxicity animal models of bronchopulmonary dysplasia and normalizes during recovery in air. Pediatr Res 2006; 59:389-95. [PMID: 16492977 DOI: 10.1203/01.pdr.0000198819.81785.f1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bronchopulmonary dysplasia (BPD), a major cause of morbidity in premature infants, is characterized by arrest of lung growth and inhibited alveologenesis. We had earlier cloned late-gestation lung 1 (LGL1), a glucocorticoid (GC)-induced, developmentally regulated gene in lung mesenchyme, and showed that reduced levels of late-gestation lung 1 protein (lgl1) inhibit lung branching. Maximal fetal expression of LGL1 is concordant with the onset of alveolar septation, suggesting an additional role for lgl1 in alveologenesis. At postnatal d 7, during the period of maximal septation in postnatal rat lung, lgl1 concentrates at the tips of budding secondary alveolar septa. We studied two models of impaired postnatal alveologenesis generated by exposure of newborn rats to 60% O2 for 2 wk or 95% O2 for 1 wk. A profound decrease of lgl1 expression with oxygen exposure was observed in both animal models. Animals exposed to 95% O2 for 1 wk recovered in air over a 3-wk period, associated with normalization of lgl1 levels. Changes in lung levels of alpha-actin (a marker of myofibroblast differentiation associated with alveologenesis) and the mesenchymal marker vimentin were significant but less marked. Our findings support a role for lgl1 in postnatal lung development. We speculate that deficiency of lgl1 contributes to the arrested alveolar partitioning observed in BPD and that recovery is associated with normalization of lgl1 levels.
Collapse
Affiliation(s)
- Katia Nadeau
- Montreal Children's Hospital Research Institute, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
34
|
Pua ZJ, Stonestreet BS, Cullen A, Shahsafaei A, Sadowska GB, Sunday ME. Histochemical analyses of altered fetal lung development following single vs multiple courses of antenatal steroids. J Histochem Cytochem 2005; 53:1469-79. [PMID: 15956023 PMCID: PMC3957547 DOI: 10.1369/jhc.5a6721.2005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Accepted: 05/04/2005] [Indexed: 11/22/2022] Open
Abstract
A single course of antenatal steroids is widely used during preterm labor to promote fetal lung maturation. However, little is known regarding efficacy and safety of multiple courses of antenatal steroids. In animal models and clinical trials, treatment with glucocorticoids can inhibit growth. The present study of single- vs multiple-course steroids in pregnant ewes analyzes the effects of steroids vs placebo on fetal lung histopathology. Single-course groups received dexamethasone (Dex) 6 mg or normal saline every 12 hr for 48 hr at 104-106 days of gestation (term = 150 days). Multiple-course groups received the first course at 76-78 days; this was repeated weekly for 5 weeks. At 108 days, lungs were analyzed using immunohistochemistry for alpha-smooth muscle actin, a myofibroblast marker and proliferating cell nuclear antigen. Cell injury/death was evaluated using TdT-mediated dUTP digoxigenin nick end labeling (TUNEL) analysis. Although fetal growth was restricted by either single or multiple courses of Dex, alveolar development was accelerated as measured by mean linear intercepts. Alveolar walls were thinner, developing septa were longer, and septal myofibroblasts were increased for both Dex groups compared with controls. Cell proliferation increased following multiple steroid courses, especially in the distal parenchyma, with a corresponding decrease in apoptosis. These observations suggest that Dex promotes alveolarization, whether given in single or multiple courses.
Collapse
Affiliation(s)
- Zarah J. Pua
- Department of Medicine and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, Massachusetts (ZJP, AC, MES)
| | - Barbara S. Stonestreet
- Department of Pediatrics, Women's & Infants’ Hospital and Brown University, Providence, Rhode Island (BSS, GBS)
| | - Anne Cullen
- Department of Medicine and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, Massachusetts (ZJP, AC, MES)
| | - Aliakbar Shahsafaei
- Department of Pathology, Brigham & Women's Hospital, Boston, Massachusetts (AS, MES)
| | - Grazyna B. Sadowska
- Department of Pediatrics, Women's & Infants’ Hospital and Brown University, Providence, Rhode Island (BSS, GBS)
| | - Mary E. Sunday
- Department of Medicine and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, Massachusetts (ZJP, AC, MES)
- Department of Pathology, Brigham & Women's Hospital, Boston, Massachusetts (AS, MES)
| |
Collapse
|
35
|
Chen P, Marsilio E, Goldstein RH, Yannas IV, Spector M. Formation of Lung Alveolar-Like Structures in Collagen–Glycosaminoglycan Scaffolds in Vitro. ACTA ACUST UNITED AC 2005; 11:1436-48. [PMID: 16259599 DOI: 10.1089/ten.2005.11.1436] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The objective of this study was to investigate the histology of tissue formed when fetal rat lung cells were grown in a collagen-glycosaminoglycan (GAG) tissue-engineering scaffold. The goal was the formation of lung histotypic structures in the tissue-engineering scaffolds in vitro. Achieving this goal would facilitate future investigations of the effects of selected scaffold design parameters on processes that may underlie aspects of lung regeneration in vivo. Lung cells were obtained from Sprague-Dawley rats after 16 and 19 days of gestation. These dissociated cells were seeded into type I collagen-chondroitin 6-sulfate matrices, 8 mm in diameter by 2 mm in thickness, cross-linked and sterilized by dehydrothermal treatment. Approximately 28 million cells were seeded into each spongelike sample. Histological and immunohistochemical studies were performed at termination periods of 2 days and 1, 2, and 3 weeks. The enzymatically dissociated 19-day gestation fetal rat lung cells formed and maintained alveolar-like structures, 50-60 microm in diameter, in the collagen- GAG scaffold. A novel finding was that all of the cell-seeded scaffolds underwent cell-mediated contraction that appeared to be associated with the finding by immunohistochemistry of expression of alpha-smooth muscle actin in some cells. These results demonstrate the capability of dissociated lung cells to form lung histotypic structures in collagen-GAG tissue-engineering scaffolds in vitro. This culture system may be of value in facilitating exploration of strategies for preparing such scaffolds for the regeneration of lung tissue in vivo.
Collapse
Affiliation(s)
- Patty Chen
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | | | | |
Collapse
|
36
|
Wang L, Chitano P, Murphy TM. Maturation of guinea pig tracheal strip stiffness. Am J Physiol Lung Cell Mol Physiol 2005; 289:L902-8. [PMID: 15951335 DOI: 10.1152/ajplung.00005.2005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previously, we showed the shortening velocity of guinea pig tracheal strips was the greatest in juvenile (3-wk-old) compared with infant (1-wk-old) and adult animals (3-mo-old). The greatest shortening velocity was associated with the least resistance to shortening calculated from force-velocity curves among the three age groups. It remained to be verified if the stiffness of tracheal tissue, a measure of tissue response to geometrical deformations, is different among the three age groups. We hypothesized that stiffness of intact tracheal strips is lowest in the juvenile group and that this can explain the ontogeny of airway smooth muscle resistance to shortening and shortening velocity. Static stiffness measured through stepwise deformations showed no age-related differences. Evaluation of tissue response to oscillatory deformations showed that the dynamic stiffness of unstimulated tracheal strips was 8.35 +/- 0.88, 4.15 +/- 1.09, and 8.21 +/- 1.57 kPa, and the phase angle was 10.3 +/- 2.93, 2.46 +/- 0.67, and 7.87 +/- 1.77 degrees in infant, juvenile, and adult, respectively. Unstimulated juvenile strips were significantly lower in dynamic stiffness and phase angle compared with unstimulated infant or adult strips. This maturational profile was independent of muscle strip preset length or oscillation mode/amplitude but was abolished at peak of contraction to either carbachol or electric field stimulation. These results suggest that the noncontractile components of tracheal strips are less stiff and contain fewer viscous/frictional elements in juvenile than in other age groups. This may provide a functional basis for reduced resistance to length changes in juvenile airway smooth muscle.
Collapse
Affiliation(s)
- Lu Wang
- Dept. of Pediatrics, Duke University Medical Center, Rm. 302, Bell Bldg., Box 2994, Durham, NC 27710, USA.
| | | | | |
Collapse
|
37
|
Abstract
Mechanical force is a critical modulator of smooth muscle (SM) function and gene expression. Very little is known, however, about its contribution to SM myogenesis. This review presents and discusses what has been learned about the role of mechanical force in inducing SM myogenesis and some of the signaling mechanisms involved in this process.
Collapse
Affiliation(s)
- Sandhya Jakkaraju
- Department of Pathology, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
38
|
Wagner TE, Frevert CW, Herzog EL, Schnapp LM. Expression of the integrin subunit alpha8 in murine lung development. J Histochem Cytochem 2003; 51:1307-15. [PMID: 14500699 DOI: 10.1177/002215540305101008] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The complex interplay between cells and extracellular matrix (ECM) proteins is critical for lung development. Integrins are key modulators of this interaction. The integrin subunit alpha 8 associates with the beta(1)-subunit to form an RGD-binding integrin. We previously showed that, in adult lung, alpha 8 is expressed in contractile interstitial cells and smooth muscle cells and is upregulated in lung injury. To gain insight into the function of alpha 8 during lung development, we examined the spatiotemporal expression of alpha 8 throughout murine lung development. We compared the distribution of alpha 8 with alpha-smooth muscle actin (alpha SMA), fibronectin (alpha 8 ligand), and cytokeratin. alpha 8 co-localized with alpha SMA and fibronectin in the peribronchial and perivascular regions. In all stages, alpha 8 immunoreactivity was detected diffusely in the mesenchyme except for cells surrounding distal, newly forming airways. alpha 8, alpha SMA, and fibronectin co-localized at tips of secondary septae in the alveolar stage. We conclude that alpha 8 is marker for lung mesenchymal cells starting early in development. alpha 8 is also a marker for smooth muscle cells, expressed as early as alpha SMA. Co-localization of alpha 8 with fibronectin suggests a role in branching morphogenesis. Furthermore, alpha 8 may participate in secondary septation by modulating signals from the extracellular matrix to alveolar myofibroblasts.
Collapse
Affiliation(s)
- Teresa E Wagner
- University of Washington, Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, Seattle, Washington, USA.
| | | | | | | |
Collapse
|
39
|
Yamamoto Y, Tanaka A, Kanamaru A, Tanaka S, Tsubone H, Atoji Y, Suzuki Y. Morphology of aging lung in F344/N rat: alveolar size, connective tissue, and smooth muscle cell markers. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2003; 272:538-47. [PMID: 12740948 DOI: 10.1002/ar.a.10172] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This study investigated the morphological changes of lungs in F344/N rats (9-36 months old). We initially examined general and quantitative morphological changes, and then we used immunohistochemistry to detect distributional changes in collagen subtypes (types I, III, and IV) and smooth muscle cell (SMC) markers (alpha-smooth muscle actin (ASMA), gamma-smooth muscle actin (GSMA), desmin, and vimentin) in the lungs. In 24-month-old rats, alveolar ducts and alveolar sacs were enlarged, and alveoli were wider and shallower than in younger animals. In old rats (>/=27 months), terminal and respiratory bronchioles and alveolar ducts were dilated and alveoli were more extended than in 24-month-old rats. No age-related distributional changes were observed for collagen types I, III, and IV as revealed by immunohistochemistry, or elastin as revealed by resorsin fuchsin. SMCs in the extra- and intrapulmonary bronchi were immunoreactive for ASMA, GSMA, and desmin, but not for vimentin at all ages. In old rats (>/=27 months), SMCs were loosely arranged in comparison with younger animals, and stainability for GSMA and desmin was decreased. In the respiratory bronchioles and alveolar ducts, a few cells immunoreactive for ASMA and vimentin were observed in the smooth muscle aggregations of the alveolar orifice in rats younger than 12 months. In older rats (>20 months), cells immunoreactive for ASMA and vimentin were increased in septal tips. In conclusion, extension of distal airways and immunohistochemical changes of SMC markers in F344/N rat lungs were evident by approximately 24 months of age, but there was no apparent change in connective tissue morphology.
Collapse
Affiliation(s)
- Yoshio Yamamoto
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, Gifu University, Gifu, Japan.
| | | | | | | | | | | | | |
Collapse
|
40
|
Hall SM, Hislop AA, Haworth SG. Origin, differentiation, and maturation of human pulmonary veins. Am J Respir Cell Mol Biol 2002; 26:333-40. [PMID: 11867341 DOI: 10.1165/ajrcmb.26.3.4698] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Recent studies on human embryonic and fetal lungs show that the pulmonary arteries form by vasculogenesis. Little is known of the early development of the pulmonary veins. Using immunohistochemical techniques and serial reconstruction, we studied 18 fetal and neonatal lungs. Sections were stained with antibodies specific for endothelium (CD31, von Willebrand factor) and smooth muscle (alpha and gamma smooth muscle actin, smooth muscle myosin, calponin, caldesmon, and desmin) and antibodies specific for the matrix glycoprotein tenascin, the receptor protein tyrosine kinase EphB4, and its ligand ephrinB2. Kiel University-raised antibody number 67 (Ki67) expression allowed qualitative assessment of cell replication. By 34 d gestation, there was continuity between the aortic sac, pulmonary arteries, capillaries, pulmonary veins, and atrium. The pulmonary veins formed by vasculogenesis in the mesenchyme surrounding the terminal buds during the pseudoglandular period and probably by angiogenesis in the canalicular and alveolar stages. EphB4 and ephrinB2 did not distinguish between presumptive venous and arterial endothelium as they do in mouse. All venous smooth muscle cells derived directly from the mesenchyme, gradually acquiring smooth muscle specific proteins from 56 d gestation. Thus, both pulmonary arteries and veins arise by vasculogenesis, but the origins of their smooth muscle cells and their cytoskeletal protein content are different.
Collapse
Affiliation(s)
- Susan M Hall
- Unit of Vascular Biology and Pharmacology, Institute of Child Health, University College, London, United Kingdom.
| | | | | |
Collapse
|
41
|
Yamada T, Suzuki E, Gejyo F, Ushiki T. Developmental changes in the structure of the rat fetal lung, with special reference to the airway smooth muscle and vasculature. ARCHIVES OF HISTOLOGY AND CYTOLOGY 2002; 65:55-69. [PMID: 12002611 DOI: 10.1679/aohc.65.55] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Structural changes in the developing rat lung were studied by a combined use of light microscopy including immunohistochemistry for a-smooth muscle actin (alpha-SMA) and scanning electron microscopy (SEM) using the KOH-collagenase digestion method. In the embryonic stage (E11-E13), the lung bud appeared as an outgrowth from the ventral wall of the foregut which grew caudally into the splanchnic mesoderm to form a pair of bronchial buds at the end. At E13, the airway smooth muscle cells first appeared around the bifurcation of the trachea. These smooth muscle cells were restricted to the dorsal surface of the tracheal epithelium, suggesting a difference in character between the dorsal and ventral sides of the mesenchymal cells in this region. During the pseudoglandular stage (E13-E18.5), the bronchial buds repeatedly gave off branches in the mesenchymal tissue. The smooth muscle cells in the bronchioles were spindle-shaped and arranged completely circularly around the epithelial tube, except that the terminal bud of bronchioles lacked the smooth muscles. The neck of the terminal bud was constantly surrounded by flat and irregularly-shaped immature smooth muscle cells, representing an early event in the smooth muscle cell differentiation from mesenchymal cells. In the canalicular to saccular stages (E18.5 to birth), the terminals of bronchioles became saccular, thus forming prospective alveolar acini. At birth, the alveolar wall became thinner than before birth, and the individual smooth muscle cells in bronchioles were elongated like a tape. As to the blood vessel differentiation, various sized sinusoidal spaces indicating the primitive blood vessels were already present in the mesenchymal tissue at E11.5. The endothelial cells of these sinusoidal spaces were irregularly shaped and sometimes extended their processes into the lumen. The network of tubular vessels appeared from E14.5. These vessels had tapering ends as well as transluminal trabeculae, suggesting that capillary growth proceeds by both the sprouting and partitioning (i.e., intussusception) of vessels in the pseudoglandular stage.
Collapse
Affiliation(s)
- Takaho Yamada
- Department of Cellular Function, Niigata University Graduate School of Medical and Dental Sciences, Japan.
| | | | | | | |
Collapse
|
42
|
Tanaka R, Al-Jamal R, Ludwig MS. Maturational changes in extracellular matrix and lung tissue mechanics. J Appl Physiol (1985) 2001; 91:2314-21. [PMID: 11641376 DOI: 10.1152/jappl.2001.91.5.2314] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The viscoelastic properties of the pulmonary parenchyma change rapidly postparturition. We compared changes in mechanical properties with changes in tissue composition of rat lung parenchymal strips in three groups of Sprague-Dawley rats: baby (B; 10-14 days), young (Y; approximately 3 wk), and adult (A; approximately 8 wk). Strips were suspended in an organ bath, and resistance (R), elastance (E), and hysteresivity (eta) were calculated during sinusoidal oscillations before and after the addition of acetylcholine (ACh) (10(-3) M). Strips were then fixed in formalin, and sections were stained with hematoxylin and eosin, Verhoff's elastic stain, or Van Gieson's picric acid-fuchsin stain for collagen. The volume proportion of collagen (%Col), the length density of elastic fibers (L(V)/Pr(alv)), and the arithmetic mean thickness of alveolar septae (T(a)) were calculated by morphometry. Tissue was also stained for alpha-smooth muscle actin (ASMA), and the volume proportion of ASMA (%ASMA) was calculated. Hyaluronic acid (HA) was quantitated by radioimmunoassay in separate strips. R and E in B strips were significantly higher, whereas eta was significantly smaller than in Y or A strips. Changes in these parameters with ACh were greater in B strips. T(a), %ASMA, and HA were greatest in B strips, whereas %Col and L(V)/Pr(alv) were least. There were significant positive correlations between R and E vs. T(a) and between percent change in R and eta post-ACh vs. T(a) and vs. %ASMA, and significant negative correlations between R and E vs. %Col and vs. L(V)/Pr(alv) and percent increase in all three mechanical parameters post-ACh vs. %Col. These data suggest that the relatively high stiffness, R, and contractile responsiveness of parenchymal tissues observed in newborns are not directly attributable to the amount of collagen and elastic fibers in the tissue, but rather they are related to the thickened alveolar wall and the relatively greater percent of contractile cells.
Collapse
Affiliation(s)
- R Tanaka
- Meakins-Christie Laboratories, Royal Victoria Hospital, McGill University, Montreal, Quebec, Canada H2X 2P2
| | | | | |
Collapse
|
43
|
Bragg AD, Moses HL, Serra R. Signaling to the epithelium is not sufficient to mediate all of the effects of transforming growth factor beta and bone morphogenetic protein 4 on murine embryonic lung development. Mech Dev 2001; 109:13-26. [PMID: 11677049 DOI: 10.1016/s0925-4773(01)00508-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Many studies have suggested that transforming growth factor beta (TGF-beta) and bone morphogenetic protein 4 (Bmp4) regulate early development of the lung. In this study, administration of growth factors directly into the lumen of lungs grown in organ culture was used to limit their activity to the epithelium and test the hypothesis that signaling to the epithelium is sufficient to mediate the known effects of TGF-beta and BMP-4 on early lung development. Addition of TGF-beta1, beta2, or beta3 to the medium surrounding lungs grown in organ culture resulted in decreased branching, reduced cell proliferation, accumulation of alpha-smooth muscle actin protein (alpha-SMA) in the mesenchyme, and decreased expression of a marker for respiratory epithelium, surfactant protein-C (Sp-C). When TGF-beta1 was restricted to the epithelium, accumulation of alpha-SMA and inhibition of Sp-C expression were not observed but branching and proliferation were inhibited. In contrast, branching was not inhibited in lungs where TGF-beta2 or TGF-beta3 were restricted to the epithelium suggesting differences in the mechanism of signaling by TGF-beta1, TGF-beta2 or TGF -beta3 in lung. Addition of Bmp4 to the medium surrounding lungs grown in organ culture stimulated cell proliferation and branching morphogenesis; however, direct injection of Bmp4 into the lung lumen had no effect on proliferation or branching. Based on these data and data from mesenchyme-free cultures, we propose that the mesenchyme influences growth factor signaling in the lung.
Collapse
Affiliation(s)
- A D Bragg
- Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | | | | |
Collapse
|
44
|
Haley KJ, Sunday ME, Osathanondh R, Du J, Vathanaprida C, Karpitsky VV, Krause JE, Lilly CM. Developmental expression of neurokinin A and functional neurokinin-2 receptors in lung. Am J Physiol Lung Cell Mol Physiol 2001; 280:L1348-58. [PMID: 11350816 DOI: 10.1152/ajplung.2001.280.6.l1348] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peribronchial smooth muscle constriction causes airway stretch, an important mechanical force in developing lung. Little is known about factors influencing these spontaneously active muscle elements. We measured contractile activity of neurokinin (NK) receptors on fetal intrapulmonary smooth muscle by tracheal perfusion assay ( n = 11). Injecting either capsaicin or the NK2 receptor agonist [NLE10]NKA resulted in significant ( P < 0.05) bronchoconstriction. A specific NK2 receptor antagonist inhibited constriction caused by endogenous tachykinins released by capsaicin. We then examined NK2 receptor ( n = 44) and NKA ( n = 23) ontogeny in human lung. NKA immunostaining was identified in peribronchial nerves in samples with gestational age >12 wk. NK2 receptor protein was identified in peribronchial and perivascular smooth muscle. These results indicate that endogenous tachykinins released by the developing lung act via NK2receptors to cause smooth muscle constriction. We speculate that tachykinins could modulate lung development.
Collapse
MESH Headings
- Adult
- Barium Compounds/pharmacology
- Bronchoconstriction/drug effects
- Bronchoconstriction/physiology
- Bronchoconstrictor Agents/pharmacology
- Capsaicin/pharmacology
- Chlorides/pharmacology
- Fetus
- Gene Expression Regulation, Developmental/physiology
- Humans
- Immunohistochemistry
- In Vitro Techniques
- Lung/cytology
- Lung/drug effects
- Lung/embryology
- Lung/metabolism
- Methacholine Chloride/pharmacology
- Muscle, Smooth/embryology
- Muscle, Smooth/innervation
- Muscle, Smooth/metabolism
- Neurokinin A/analogs & derivatives
- Neurokinin A/metabolism
- Neurokinin A/pharmacology
- Peptides/pharmacology
- Perfusion
- RNA, Messenger/metabolism
- Receptors, Neurokinin-1/agonists
- Receptors, Neurokinin-1/metabolism
- Receptors, Neurokinin-2/agonists
- Receptors, Neurokinin-2/genetics
- Receptors, Neurokinin-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Trachea/blood supply
- Trachea/drug effects
- Trachea/embryology
- Trachea/metabolism
Collapse
Affiliation(s)
- K J Haley
- Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Liu J, Beqaj S, Yang Y, Honoré B, Schuger L. Heterogeneous nuclear ribonucleoprotein-H plays a suppressive role in visceral myogenesis. Mech Dev 2001; 104:79-87. [PMID: 11404082 DOI: 10.1016/s0925-4773(01)00377-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mouse embryonic mesenchymal cells undergo spontaneous smooth muscle (SM) differentiation upon spreading/elongation in culture (Relan et al., J. Cell Biol. 147 (1999) 1341; Yang et al., Development 125 (1998) 2621; Yang et al., Development 126 (1999) 3027). Using these cells we generated a subtracted cDNA library to identify potential suppressors of SM myogenesis. One of the differentially expressed genes was heterogeneous nuclear ribonucleoprotein-H (hnRNP-H), which is involved in pre-mRNA alternative splicing. hnRNP-H was highly expressed in mesenchymal cells prior to the onset of SM differentiation, but its expression rapidly decreased in mesenchymal cells undergoing SM myogenesis. In vivo, the drop in hnRNP-H expression was restricted to visceral SM cells. Antisense oligodeoxynucleotide and antisense RNA were used to inhibit hnRNP-H synthesis in SM-differentiating mesenchymal cells and in embryonic lung explants. A decrease in hnRNP-H levels resulted in upregulation of SM-specific gene expression and increased bronchial SM development in lung explants. hnRNP-H overexpression in cell cultures had the opposite effect. These studies, therefore, indicate a novel role for hnRNP-H in the control of visceral myogenesis.
Collapse
Affiliation(s)
- J Liu
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
46
|
Tollet J, Everett AW, Sparrow MP. Spatial and temporal distribution of nerves, ganglia, and smooth muscle during the early pseudoglandular stage of fetal mouse lung development. Dev Dyn 2001; 221:48-60. [PMID: 11357193 DOI: 10.1002/dvdy.1124] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Neural tissue and smooth muscle appear early in the developing fetal lung, but little is known of their origin and subsequent distribution. To investigate the spatial and temporal distribution of nerves, ganglia, and airway smooth muscle during the early pseudoglandular stage, fetal mouse lungs at embryonic days (E) 11 to 14 were immunostained as whole-mounts and imaged by confocal microscopy. At E11, the primordial lung consisted of the future trachea and two budding epithelial tubules that were covered in smooth muscle to the base of the growing buds. The vagus and processes entering the lung were positive for the neural markers PGP 9.5 (protein gene product 9.5) and synapsin but no neurons were stained at this stage. An antibody to p75NTR revealed neural crest cells on the future trachea as well as in the vagus and in processes extending from the vagus to the lung. This finding indicates that even though neuronal precursors are already present at this stage, they are still migrating into the lung. By E12, neural tissue was abundant in the proximal part of the lung and nerves followed the smooth muscle-covered tubules to the base of the growing buds. At E13 and E14, a neural network of interconnected ganglia, innervated by the vagus, covered the trachea. The postganglionic nerves mainly followed the smooth muscle-covered tubules, but some extended out into the mesenchyme beyond the epithelial buds. Furthermore, we show in a model of cultured lung explants that neural tissue and smooth muscle persist and continue to grow and differentiate in vitro. By using fluorescent markers and confocal microscopy, we present the developing lung as a dynamic structure with smooth muscle and neural tissue in a prime position to influence growth and development.
Collapse
Affiliation(s)
- J Tollet
- Department of Physiology, University of Western Australia, Nedlands, Western Australia, Australia.
| | | | | |
Collapse
|
47
|
Halayko AJ, Solway J. Molecular mechanisms of phenotypic plasticity in smooth muscle cells. J Appl Physiol (1985) 2001; 90:358-68. [PMID: 11133929 DOI: 10.1152/jappl.2001.90.1.358] [Citation(s) in RCA: 193] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Morphological, functional, molecular and cell biology studies have revealed a striking multifunctional nature of individual smooth muscle cells (SMC). SMCs manifest phenotypic plasticity in response to changes in environment and functional requirements, acquiring a range of structural and functional properties bounded by two extremes, called "synthetic" and "contractile." Each phenotypic state is characterized by expression of a unique set of structural, contractile, and receptor proteins and isoforms that correlate with differing patterns of gene expression. Recent studies have identified signaling pathways and transcription factors (e.g., RhoA GTPase/ROCK, also known as Rho kinase, and serum response factor) that regulate the transcriptional activities of genes encoding proteins associated with the contractile apparatus. Mechanical plasticity of contractile-state smooth muscle further extends SMC functional diversity. This may also be regulated, in part, by the RhoA GTPase/ROCK pathway, via reorganization of cytoskeletal and contractile proteins. Future studies that define transcriptional and posttranscriptional mechanisms of SMC plasticity are necessary to fully understand the role of SMC in the pathogenesis and morbidity of human diseases of the airways, vasculature, and gastrointestinal tract.
Collapse
Affiliation(s)
- A J Halayko
- Department of Physiology and Section of Respiratory Diseases, University of Manitoba, Winnipeg, Manitoba, Canada R3A 1R8.
| | | |
Collapse
|
48
|
Yang Y, Beqaj S, Kemp P, Ariel I, Schuger L. Stretch-induced alternative splicing of serum response factor promotes bronchial myogenesis and is defective in lung hypoplasia. J Clin Invest 2000; 106:1321-30. [PMID: 11104785 PMCID: PMC387248 DOI: 10.1172/jci8893] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Smooth muscle (SM) develops only in organs and sites that sustain mechanical tensions. Therefore, we determined the role of stretch in mouse and human bronchial myogenesis. Sustained stretch induced expression of SM proteins in undifferentiated mesenchymal cells and accelerated the differentiation of cells undergoing myogenesis. Moreover, bronchial myogenesis was entirely controlled in lung organ cultures by the airway intraluminal pressure. Serum response factor (SRF) is a transcription factor critical for the induction of muscle-specific gene expression. Recently, a SRF-truncated isoform produced by alternative splicing of exon 5 has been identified (SRFDelta5). Here we show that undifferentiated mesenchymal cells synthesize both SRF and SRFDelta5 but that SRFDelta5 synthesis is suppressed during bronchial myogenesis in favor of increased SRF production. Stretch induces the same change in SRF alternative splicing, and its myogenic effect is abrogated by overexpressing SRFDelta5. Furthermore, human hypoplastic lungs related to conditions that hinder cell stretching continue to synthesize SRFDelta5 and show a marked decrease in bronchial and interstitial SM cells and their ECM product, tropoelastin. Taken together, our findings indicate that stretch plays a critical role in SM myogenesis and suggest that its decrease precludes normal bronchial muscle development.
Collapse
Affiliation(s)
- Y Yang
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | |
Collapse
|
49
|
Hall SM, Hislop AA, Pierce CM, Haworth SG. Prenatal origins of human intrapulmonary arteries: formation and smooth muscle maturation. Am J Respir Cell Mol Biol 2000; 23:194-203. [PMID: 10919986 DOI: 10.1165/ajrcmb.23.2.3975] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Recent studies on the morphogenesis of the pulmonary arteries have focused on nonhuman species such as the chick and the mouse. Using immunohistochemical techniques, we have studied 16 lungs from human embryos and fetuses from 28 d of gestation to newborn, using serial sections stained with a panel of antibodies specific for endothelium, smooth muscle, and extracellular matrix proteins. Cell replication was also assessed. Serial reconstruction showed a continuity of circulation between the heart and the capillary plexus from at least 38 d of gestation. The intrapulmonary arteries appeared to be derived from a continuous expansion of the primary capillary plexus that is from within the mesenchyme, by vasculogenesis. The arteries formed by continuous coalescence of endothelial tubes alongside the newly formed airway. Findings were consistent with the pulmonary arterial smooth muscle cells being derived from three sites in a temporally distinct sequence: the earliest from the bronchial smooth muscle, later from the mesenchyme surrounding the arteries, and last from the endothelial cells. Despite their different origins, all smooth muscle cells followed the same sequence of expression of smooth muscle-specific cytoskeletal proteins with increasing age. The order of appearance of these maturing proteins was from the subendothelial cells outward across the vessel wall and from hilum to periphery. The airways would seem to act as a template for pulmonary artery development. This study provides a framework for studying the signaling mechanisms controlling the various aspects of lung development.
Collapse
Affiliation(s)
- S M Hall
- Unit of Vascular Biology and Pharmacology, Cardiovascular and Respiratory Sciences, Institute of Child Health, University College of London, London, United Kingdom
| | | | | | | |
Collapse
|
50
|
Wright C, Strauss S, Toole K, Burt AD, Robson SC. Composition of the pulmonary interstitium during normal development of the human fetus. Pediatr Dev Pathol 1999; 2:424-31. [PMID: 10441619 DOI: 10.1007/s100249900145] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Normal lung development is dependent on epithelial-mesenchymal interactions. This study was undertaken to examine the structure of the interstitium of the developing human fetal lung, concentrating particularly on the first and second trimesters. Lung tissue was obtained at autopsy from nonmalformed, nonmacerated cases of spontaneous abortion (n = 15), stillbirth (n = 9), and very early neonatal death (n = 5) (range of gestations, 10-42 weeks). Paraffin-embedded tissue sections were examined using immunohistochemical methods to determine expression of collagens I, III, IV, V, and VI; the glycoproteins fibronectin and laminin; and the intermediate filaments vimentin, alpha-smooth muscle actin (alphaSMA), and desmin. Collagens III and VI and cells expressing alphaSMA were present consistently at points of airway branching and secondary crest formation, indicating a role for these components in the initiation and stabilization of airway branches in the developing lung. Desmin expression by stromal cells succeeded alphaSMA temporally, and may represent a marker of terminal smooth muscle differentiation within the airway; it was not detected in the vascular tree. Other components were widely expressed throughout the extracellular matrix, including basement membranes, at all gestations. The spatial and temporal patterns of expression of components of the lung interstitium provide clues to the mechanisms underlying normal human lung development and possible insights into the pathogenesis of fetal and neonatal lung disease.
Collapse
Affiliation(s)
- C Wright
- Department of Pathology, University of Newcastle upon Tyne, Newcastle upon Tyne, NE1 4LP, UK
| | | | | | | | | |
Collapse
|