301
|
Jain K, Smyth AR. Current dilemmas in antimicrobial therapy in cystic fibrosis. Expert Rev Respir Med 2013; 6:407-22. [PMID: 22971066 DOI: 10.1586/ers.12.39] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The majority of cystic fibrosis (CF)-related morbidity and mortality is caused by pulmonary damage due to recurrent and chronic infections. Considerable improvements in the survival of individuals with CF have been achieved in recent decades, some of which may be due to better management of common pathogens such as Staphylococcus aureus and Pseudomonas aeruginosa. While the search continues for the optimal approach for prophylaxis, eradication and maintenance treatment of infections, there are several unanswered questions, posing dilemmas related to various therapeutic choices. Microbes pose additional challenges by adapting to CF lungs and developing treatment resistance. Several new, highly antimicrobial-resistant pathogens have emerged. Their pathogenic role in the progression of CF lung disease is not yet clear and effective treatment approaches have not been defined. There is an urgent need for well-designed comparative clinical trials of new antibiotic strategies.
Collapse
Affiliation(s)
- Kamini Jain
- School of Clinical Sciences, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
302
|
CFTR-deficient pigs display peripheral nervous system defects at birth. Proc Natl Acad Sci U S A 2013; 110:3083-8. [PMID: 23382208 DOI: 10.1073/pnas.1222729110] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Peripheral nervous system abnormalities, including neuropathy, have been reported in people with cystic fibrosis. These abnormalities have largely been attributed to secondary manifestations of the disease. We tested the hypothesis that disruption of the cystic fibrosis transmembrane conductance regulator (CFTR) gene directly influences nervous system function by studying newborn CFTR(-/-) pigs. We discovered CFTR expression and activity in Schwann cells, and loss of CFTR caused ultrastructural myelin sheath abnormalities similar to those in known neuropathies. Consistent with neuropathic changes, we found increased transcripts for myelin protein zero, a gene that, when mutated, can cause axonal and/or demyelinating neuropathy. In addition, axon density was reduced and conduction velocities of the trigeminal and sciatic nerves were decreased. Moreover, in vivo auditory brainstem evoked potentials revealed delayed conduction of the vestibulocochlear nerve. Our data suggest that loss of CFTR directly alters Schwann cell function and that some nervous system defects in people with cystic fibrosis are likely primary.
Collapse
|
303
|
Prather RS, Lorson M, Ross JW, Whyte JJ, Walters E. Genetically engineered pig models for human diseases. Annu Rev Anim Biosci 2013; 1:203-19. [PMID: 25387017 DOI: 10.1146/annurev-animal-031412-103715] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although pigs are used widely as models of human disease, their utility as models has been enhanced by genetic engineering. Initially, transgenes were added randomly to the genome, but with the application of homologous recombination, zinc finger nucleases, and transcription activator-like effector nuclease (TALEN) technologies, now most any genetic change that can be envisioned can be completed. To date these genetic modifications have resulted in animals that have the potential to provide new insights into human diseases for which a good animal model did not exist previously. These new animal models should provide the preclinical data for treatments that are developed for diseases such as Alzheimer's disease, cystic fibrosis, retinitis pigmentosa, spinal muscular atrophy, diabetes, and organ failure. These new models will help to uncover aspects and treatments of these diseases that were otherwise unattainable. The focus of this review is to describe genetically engineered pigs that have resulted in models of human diseases.
Collapse
Affiliation(s)
- Randall S Prather
- Division of Animal Science, National Swine Resource and Research Center, and
| | | | | | | | | |
Collapse
|
304
|
Abstract
Cystic fibrosis is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) molecule; these mutations result in a defect in chloride secretion in epithelial cell layers. The disease is characterized by severe gastrointestinal and pulmonary symptoms, but it is the pulmonary symptoms that dominate the clinical course of the disease and determine patients' life expectancy. These pulmonary symptoms include reduced mucociliary clearance, chronic inflammation, and recurrent and chronic pulmonary infections with Pseudomonas aeruginosa, Staphylococcus aureus, Burkholderia cepacia, and Haemophilus influenzae. Recent studies have shown that sphingolipids, especially ceramide, play a crucial role in the pathogenesis of cystic fibrosis. These studies have demonstrated that ceramide accumulates in the lungs of cystic fibrosis patients and mice, causing inflammation and high susceptibility to bacterial infections. The results of initial clinical studies suggest that interfering with sphingolipids may be a novel treatment strategy for cystic fibrosis.
Collapse
Affiliation(s)
- Heike Grassmé
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | | | | |
Collapse
|
305
|
The pig as a model for investigating the role of neutrophil serine proteases in human inflammatory lung diseases. Biochem J 2012; 447:363-70. [PMID: 22860995 PMCID: PMC3492928 DOI: 10.1042/bj20120818] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The serine proteases released by activated polymorphonuclear neutrophils [NSPs (neutrophil serine proteases)] contribute to a variety of inflammatory lung diseases, including CF (cystic fibrosis). They are therefore key targets for the development of efficient inhibitors. Although rodent models have contributed to our understanding of several diseases, we have previously shown that they are not appropriate for testing anti-NSP therapeutic strategies [Kalupov, Brillard-Bourdet, Dade, Serrano, Wartelle, Guyot, Juliano, Moreau, Belaaouaj and Gauthier (2009) J. Biol. Chem. 284, 34084–34091). Thus NSPs must be characterized in an animal model that is much more likely to predict how therapies will act in humans in order to develop protease inhibitors as drugs. The recently developed CFTR−/− (CFTR is CF transmembrane conductance regulator) pig model is a promising alternative to the mouse model of CF [Rogers, Stoltz, Meyerholz, Ostedgaard, Rokhlina, Taft, Rogan, Pezzulo, Karp, Itani et al. (2008) Science 321, 1837–1841]. We have isolated blood neutrophils from healthy pigs and determined their responses to the bacterial pathogens Pseudomonas aeruginosa and Staphylococcus aureus, and the biochemical properties of their NSPs. We used confocal microscopy and antibodies directed against their human homologues to show that the three NSPs (elastase, protease 3 and cathepsin G) are enzymatically active and present on the surface of triggered neutrophils and NETs (neutrophil extracellular traps). All of the porcine NSPs are effectively inhibited by human NSP inhibitors. We conclude that there is a close functional resemblance between porcine and human NSPs. The pig is therefore a suitable animal model for testing new NSP inhibitors as anti-inflammatory agents in neutrophil-associated diseases such as CF.
Collapse
|
306
|
Mayer ML, Blohmke CJ, Falsafi R, Fjell CD, Madera L, Turvey SE, Hancock REW. Rescue of Dysfunctional Autophagy Attenuates Hyperinflammatory Responses from Cystic Fibrosis Cells. THE JOURNAL OF IMMUNOLOGY 2012; 190:1227-38. [DOI: 10.4049/jimmunol.1201404] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
307
|
Sinn PL, Cooney AL, Oakland M, Dylla DE, Wallen TJ, Pezzulo AA, Chang EH, McCray PB. Lentiviral vector gene transfer to porcine airways. MOLECULAR THERAPY-NUCLEIC ACIDS 2012. [PMID: 23187455 PMCID: PMC3511674 DOI: 10.1038/mtna.2012.47] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this study, we investigated lentiviral vector development and transduction efficiencies in well-differentiated primary cultures of pig airway epithelia (PAE) and wild-type pigs in vivo. We noted gene transfer efficiencies similar to that observed for human airway epithelia (HAE). Interestingly, feline immunodeficiency virus (FIV)-based vectors transduced immortalized pig cells as well as pig primary cells more efficiently than HIV-1–based vectors. PAE express TRIM5α, a well-characterized species-specific lentiviral restriction factor. We contrasted the restrictive properties of porcine TRIM5α against FIV- and HIV-based vectors using gain and loss of function approaches. We observed no effect on HIV-1 or FIV conferred transgene expression in response to porcine TRIM5α overexpression or knockdown. To evaluate the ability of GP64-FIV to transduce porcine airways in vivo, we delivered vector expressing mCherry to the tracheal lobe of the lung and the ethmoid sinus of 4-week-old pigs. One week later, epithelial cells expressing mCherry were readily detected. Our findings indicate that pseudotyped FIV vectors confer similar tropisms in porcine epithelia as observed in human HAE and provide further support for the selection of GP64 as an appropriate envelope pseudotype for future preclinical gene therapy studies in the porcine model of cystic fibrosis (CF).
Collapse
Affiliation(s)
- Patrick L Sinn
- Department of Pediatrics, Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | | | | | | | | | | | | |
Collapse
|
308
|
Walters EM, Wolf E, Whyte JJ, Mao J, Renner S, Nagashima H, Kobayashi E, Zhao J, Wells KD, Critser JK, Riley LK, Prather RS. Completion of the swine genome will simplify the production of swine as a large animal biomedical model. BMC Med Genomics 2012; 5:55. [PMID: 23151353 PMCID: PMC3499190 DOI: 10.1186/1755-8794-5-55] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 10/28/2011] [Indexed: 12/25/2022] Open
Abstract
Background Anatomic and physiological similarities to the human make swine an excellent large animal model for human health and disease. Methods Cloning from a modified somatic cell, which can be determined in cells prior to making the animal, is the only method available for the production of targeted modifications in swine. Results Since some strains of swine are similar in size to humans, technologies that have been developed for swine can be readily adapted to humans and vice versa. Here the importance of swine as a biomedical model, current technologies to produce genetically enhanced swine, current biomedical models, and how the completion of the swine genome will promote swine as a biomedical model are discussed. Conclusions The completion of the swine genome will enhance the continued use and development of swine as models of human health, syndromes and conditions.
Collapse
Affiliation(s)
- Eric M Walters
- National Swine Resource and Research Center, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
309
|
Uc A, Giriyappa R, Meyerholz DK, Griffin M, Ostedgaard LS, Tang XX, Abu-El-Haija M, Stoltz DA, Ludwig P, Pezzulo A, Abu-El-Haija M, Taft P, Welsh MJ. Pancreatic and biliary secretion are both altered in cystic fibrosis pigs. Am J Physiol Gastrointest Liver Physiol 2012; 303:G961-8. [PMID: 22936270 PMCID: PMC3469695 DOI: 10.1152/ajpgi.00030.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The pancreas, liver, and gallbladder are commonly involved in cystic fibrosis (CF), and acidic, dehydrated, and protein-rich secretions are characteristic findings. Pancreatic function studies in humans have been done by sampling the jejunal fluid. However, it has been difficult to separately study the function of pancreatic and biliary systems in humans with CF, because jejunal fluid contains a mixture of bile and pancreatic fluids. In contrast, pancreatic and biliary ducts open separately into the porcine intestine; therefore, biliary and pancreatic fluid can be individually analyzed in CF pigs. We studied newborn wild-type (WT) and CF pigs and found that CFTR was localized to the pancreatic ducts. We collected bile and pancreatic fluid and analyzed pancreatic enzymes with activity assays and immunoblot. Pancreatic enzyme expression was significantly decreased in CF compared with WT pigs. The volume and pH of pancreatic fluid were significantly lower and protein concentration was >5-fold higher in CF pigs. Secretin stimulation increased pancreatic fluid volume and pH in WT, but not CF, pigs. Baseline bile volume did not differ between WT and CF pigs, but volume did not increase in response to secretin in CF pigs. Bile pH was lower and protein concentration was twofold higher in CF pigs. These results indicate that pancreatic and biliary secretions are altered in CF pigs. Abnormal pancreatic and biliary secretion in CF may have important implications in disease pathogenesis.
Collapse
Affiliation(s)
- Aliye Uc
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA.
| | - Radhamma Giriyappa
- 1Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa;
| | - David K. Meyerholz
- 2Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa;
| | - Michelle Griffin
- 1Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa;
| | - Lynda S. Ostedgaard
- 3Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - Xiao Xiao Tang
- 3Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - Marwa Abu-El-Haija
- 1Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa;
| | - David A. Stoltz
- 3Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - Paula Ludwig
- 3Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - Alejandro Pezzulo
- 3Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - Maisam Abu-El-Haija
- 1Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa;
| | - Peter Taft
- 3Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - Michael J. Welsh
- 3Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; and ,4Howard Hughes Medical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
310
|
Tucker TA, Fortenberry JA, Zsembery A, Schwiebert LM, Schwiebert EM. The ΔF508-CFTR mutation inhibits wild-type CFTR processing and function when co-expressed in human airway epithelia and in mouse nasal mucosa. BMC PHYSIOLOGY 2012; 12:12. [PMID: 22999299 PMCID: PMC3507716 DOI: 10.1186/1472-6793-12-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 09/04/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND Rescue or correction of CFTR function in native epithelia is the ultimate goal of CF therapeutics development. Wild-type (WT) CFTR introduction and replacement is also of particular interest. Such therapies may be complicated by possible CFTR self-assembly into an oligomer or multimer. RESULTS Surprisingly, functional CFTR assays in native airway epithelia showed that the most common CFTR mutant, ΔF508-CFTR (ΔF-CFTR), inhibits WT-CFTR when both forms are co-expressed. To examine more mechanistically, both forms of CFTR were transfected transiently in varying amounts into IB3-1 CF human airway epithelial cells and HEK-293 human embryonic kidney cells null for endogenous CFTR protein expression. Increasing amounts of ΔF-CFTR inhibited WT-CFTR protein processing and function in CF human airway epithelial cells but not in heterologous HEK-293 cells. Stably expressed ΔF-CFTR in clones of the non-CF human airway epithelial cell line, CALU-3, also showed reduction in cAMP-stimulated anion secretion and in WT-CFTR processing. An ultimate test of this dominant negative-like effect of ΔF-CFTR on WT-CFTR was the parallel study of two different CF mouse models: the ΔF-CFTR mouse and the bitransgenic CFTR mouse corrected in the gut but null in the lung and airways. WT/ΔF heterozygotes had an intermediate phenotype with regard to CFTR agonist responses in in vivo nasal potential difference (NPD) recordings and in Ussing chamber recordings of short-circuit current (ISC) in vitro on primary tracheal epithelial cells isolated from the same mice. In contrast, CFTR bitransgenic +/- heterozygotes had no difference in their responses versus +/+ wild-type mice. CONCLUSIONS Taken altogether, these data suggest that ΔF-CFTR and WT-CFTR co-assemble into an oligomeric macromolecular complex in native epithelia and share protein processing machinery and regulation at the level of the endoplasmic reticulum (ER). As a consequence, ΔF-CFTR slows WT-CFTR protein processing and limits its expression and function in the apical membrane of native airway epithelia. Implications of these data for the relative health of CF heterozygous carriers, for CFTR protein processing in native airway epithelia, and for the relative efficacy of different CF therapeutic approaches is significant and is discussed.
Collapse
Affiliation(s)
- Torry A Tucker
- Departments of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35294-0005, USA
- Gregory Fleming James Cystic Fibrosis (CF) Research Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, 35294-0005 AL, USA
- Department of Biochemistry, University of Texas Health Sciences Center at Tyler, Tyler, TX, USA
| | - James A Fortenberry
- Gregory Fleming James Cystic Fibrosis (CF) Research Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, 35294-0005 AL, USA
| | - Akos Zsembery
- Departments of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35294-0005, USA
- Gregory Fleming James Cystic Fibrosis (CF) Research Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, 35294-0005 AL, USA
- Department of Experimental Human Physiology, Semmelweis University, Budapest, Hungary
| | - Lisa M Schwiebert
- Departments of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35294-0005, USA
- Gregory Fleming James Cystic Fibrosis (CF) Research Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, 35294-0005 AL, USA
| | - Erik M Schwiebert
- Departments of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35294-0005, USA
- Gregory Fleming James Cystic Fibrosis (CF) Research Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, 35294-0005 AL, USA
- DiscoveryBioMed, Inc, Birmingham, AL, USA
| |
Collapse
|
311
|
Collawn JF, Matalon S. The role of CFTR in transepithelial liquid transport in pig alveolar epithelia. Am J Physiol Lung Cell Mol Physiol 2012; 303:L489-91. [PMID: 22797251 PMCID: PMC3468479 DOI: 10.1152/ajplung.00216.2012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
312
|
Veit G, Bossard F, Goepp J, Verkman AS, Galietta LJV, Hanrahan JW, Lukacs GL. Proinflammatory cytokine secretion is suppressed by TMEM16A or CFTR channel activity in human cystic fibrosis bronchial epithelia. Mol Biol Cell 2012; 23:4188-202. [PMID: 22973054 PMCID: PMC3484098 DOI: 10.1091/mbc.e12-06-0424] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Functional expression of either CFTR or the calcium-activated chloride channel TMEM16A attenuates expression and secretion of the proinflammatory cytokines IL-6, IL-8, and CXCL1/2 in respiratory epithelia. Thus augmented proinflammatory cytokine secretion caused by defective anion transport may contribute to lung inflammation in cystic fibrosis. Cystic fibrosis (CF) is caused by the functional expression defect of the CF transmembrane conductance regulator (CFTR) chloride channel at the apical plasma membrane. Impaired bacterial clearance and hyperactive innate immune response are hallmarks of the CF lung disease, yet the existence of and mechanism accounting for the innate immune defect that occurs before infection remain controversial. Inducible expression of either CFTR or the calcium-activated chloride channel TMEM16A attenuated the proinflammatory cytokines interleukin-6 (IL-6), IL-8, and CXCL1/2 in two human respiratory epithelial models under air–liquid but not liquid–liquid interface culture. Expression of wild-type but not the inactive G551D-CFTR indicates that secretion of the chemoattractant IL-8 is inversely proportional to CFTR channel activity in cftr∆F508/∆F508 immortalized and primary human bronchial epithelia. Similarly, direct but not P2Y receptor–mediated activation of TMEM16A attenuates IL-8 secretion in respiratory epithelia. Thus augmented proinflammatory cytokine secretion caused by defective anion transport at the apical membrane may contribute to the excessive and persistent lung inflammation in CF and perhaps in other respiratory diseases associated with documented down-regulation of CFTR (e.g., chronic obstructive pulmonary disease). Direct pharmacological activation of TMEM16A offers a potential therapeutic strategy to reduce the inflammation of CF airway epithelia.
Collapse
Affiliation(s)
- Guido Veit
- Department of Physiology and Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montréal, QC H3G 1Y6, Canada
| | | | | | | | | | | | | |
Collapse
|
313
|
Synthesis of porcine pCLCA2 protein during late differentiation of keratinocytes of epidermis and hair follicle inner root sheath. Cell Tissue Res 2012; 350:445-53. [PMID: 22968961 DOI: 10.1007/s00441-012-1482-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 07/12/2012] [Indexed: 10/27/2022]
Abstract
Despite the discovery of the widely expressed CLCA (chloride channel regulators, calcium-activated) proteins more than 15 years ago, their seemingly diverse functions are still poorly understood. With the recent generation of porcine animal models for cystic fibrosis (CF), members of the porcine CLCA family are becoming of interest as possible modulators of the disease in the pig. Here, we characterize pCLCA2, the porcine ortholog of the human hCLCA2 and the murine mCLCA5, which are the only CLCA members expressed in the skin. Immunohistochemical studies with a specific antibody against pCLCA2 have revealed a highly restricted pCLCA2 protein expression in the skin. The protein is strictly co-localized with filaggrin and trichohyalin in the granular layer of the epidermis and the inner root sheath of the hair follicles, respectively. No differences have been observed between the expression patterns of wild-type pigs and CF transmembrane conductance regulator(-/-) pigs. We speculate that pCLCA2 plays an as yet undefined role in the structural integrity of the skin or, possibly, in specialized functions of the epidermis, including barrier or defense mechanisms.
Collapse
|
314
|
Bauer C, Adam R, Stoltz DA, Beichel RR. Computer-aided analysis of airway trees in micro-CT scans of ex vivo porcine lung tissue. Comput Med Imaging Graph 2012; 36:601-9. [PMID: 22959430 DOI: 10.1016/j.compmedimag.2012.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 08/01/2012] [Accepted: 08/13/2012] [Indexed: 10/27/2022]
Abstract
We present a highly automated approach to obtain detailed structural models of airway trees from ex vivo porcine lung tissue imaged with a high resolution micro-CT scanner. Such information is an important prerequisite to systematically study models of lung disease that affect airway morphology. The method initially identifies all tubular airway-like structures in the lung. In a second processing step, these structures are grouped into a connected airway tree by utilizing prior knowledge about the airway trees branching pattern. The method was evaluated on 12 micro-CT scans from four tracheal lobes of piglets imaged at three different inflation levels. For this study, two control piglets and two cystic fibrosis piglets were used. For systematic validation of our approach, an airway nomenclature was developed for the pig airway tree. Out of more than 3500 airway tree segments assessed during evaluation, 88.45% were correctly identified by the method. No false positive airway branches were found. A detailed performance analysis for different airway tree hierarchy levels, lung inflation levels and piglets with/without cystic fibrosis is presented in the paper.
Collapse
Affiliation(s)
- Christian Bauer
- Department of Electrical and Computer Engineering, The University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | |
Collapse
|
315
|
Chang EH, Pezzulo AA, Meyerholz DK, Potash AE, Wallen TJ, Reznikov LR, Sieren JC, Karp PH, Ernst S, Moninger TO, Gansemer ND, McCray PB, Stoltz DA, Welsh MJ, Zabner J. Sinus hypoplasia precedes sinus infection in a porcine model of cystic fibrosis. Laryngoscope 2012; 122:1898-905. [PMID: 22711071 PMCID: PMC3449319 DOI: 10.1002/lary.23392] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 03/30/2012] [Accepted: 04/09/2012] [Indexed: 01/15/2023]
Abstract
OBJECTIVES/HYPOTHESIS Chronic sinusitis is nearly universal in humans with cystic fibrosis (CF) and is accompanied by sinus hypoplasia (small sinuses). However, whether impaired sinus development is a primary feature of loss of the cystic fibrosis transmembrane conductance regulator (CFTR) or a secondary consequence of chronic infection remains unknown. Our objective was to study the early pathogenesis of sinus disease in CF. STUDY DESIGN Animal/basic science research. METHODS Sinus development was studied in a porcine CF model. RESULTS Porcine sinus epithelia expressed CFTR and exhibited transepithelial anion transport. Disruption of the CFTR gene eliminated both. Sinuses of newborn CF pigs were not infected and showed no evidence of inflammation, yet were hypoplastic at birth. Older CF pigs spontaneously developed sinus disease similar to that seen in humans with CF. CONCLUSIONS These results define a role for CFTR in sinus development and suggest the potential of the CF pig as a genetic model of CF-sinus disease in which to test therapeutic strategies to minimize sinus-related CF morbidity.
Collapse
Affiliation(s)
- Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
316
|
Serial analysis of the gut and respiratory microbiome in cystic fibrosis in infancy: interaction between intestinal and respiratory tracts and impact of nutritional exposures. mBio 2012; 3:mBio.00251-12. [PMID: 22911969 PMCID: PMC3428694 DOI: 10.1128/mbio.00251-12] [Citation(s) in RCA: 256] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pulmonary damage caused by chronic colonization of the cystic fibrosis (CF) lung by microbial communities is the proximal cause of respiratory failure. While there has been an effort to document the microbiome of the CF lung in pediatric and adult patients, little is known regarding the developing microflora in infants. We examined the respiratory and intestinal microbiota development in infants with CF from birth to 21 months. Distinct genera dominated in the gut compared to those in the respiratory tract, yet some bacteria overlapped, demonstrating a core microbiota dominated by Veillonella and Streptococcus. Bacterial diversity increased significantly over time, with evidence of more rapidly acquired diversity in the respiratory tract. There was a high degree of concordance between the bacteria that were increasing or decreasing over time in both compartments; in particular, a significant proportion (14/16 genera) increasing in the gut were also increasing in the respiratory tract. For 7 genera, gut colonization presages their appearance in the respiratory tract. Clustering analysis of respiratory samples indicated profiles of bacteria associated with breast-feeding, and for gut samples, introduction of solid foods even after adjustment for the time at which the sample was collected. Furthermore, changes in diet also result in altered respiratory microflora, suggesting a link between nutrition and development of microbial communities in the respiratory tract. Our findings suggest that nutritional factors and gut colonization patterns are determinants of the microbial development of respiratory tract microbiota in infants with CF and present opportunities for early intervention in CF with altered dietary or probiotic strategies. While efforts have been focused on assessing the microbiome of pediatric and adult cystic fibrosis (CF) patients to understand how chronic colonization by these microbes contributes to pulmonary damage, little is known regarding the earliest development of respiratory and gut microflora in infants with CF. Our findings suggest that colonization of the respiratory tract by microbes is presaged by colonization of the gut and demonstrated a role of nutrition in development of the respiratory microflora. Thus, targeted dietary or probiotic strategies may be an effective means to change the course of the colonization of the CF lung and thereby improve patient outcomes.
Collapse
|
317
|
Hartl D, Gaggar A, Bruscia E, Hector A, Marcos V, Jung A, Greene C, McElvaney G, Mall M, Döring G. Innate immunity in cystic fibrosis lung disease. J Cyst Fibros 2012; 11:363-82. [PMID: 22917571 DOI: 10.1016/j.jcf.2012.07.003] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 06/29/2012] [Accepted: 07/02/2012] [Indexed: 12/16/2022]
Abstract
Chronic lung disease determines the morbidity and mortality of cystic fibrosis (CF) patients. The pulmonary immune response in CF is characterized by an early and non-resolving activation of the innate immune system, which is dysregulated at several levels. Here we provide a comprehensive overview of innate immunity in CF lung disease, involving (i) epithelial dysfunction, (ii) pathogen sensing, (iii) leukocyte recruitment, (iv) phagocyte impairment, (v) mechanisms linking innate and adaptive immunity and (iv) the potential clinical relevance. Dissecting the complex network of innate immune regulation and associated pro-inflammatory cascades in CF lung disease may pave the way for novel immune-targeted therapies in CF and other chronic infective lung diseases.
Collapse
Affiliation(s)
- D Hartl
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
318
|
Abstract
Chris Karp discusses the negative impact of cold stress on mouse models of disease. Mus musculus enjoys pride of place at the center of contemporary biomedical research. Despite being the current model system of choice for in vivo mechanistic analysis, mice have clear limitations. The literature is littered with examples of therapeutic approaches that showed promise in mouse models but failed in clinical trials. More generally, mice often provide poor mimics of the human diseases being modeled. Available data suggest that the cold stress to which laboratory mice are ubiquitously subjected profoundly affects mouse physiology in ways that impair the modeling of human homeostasis and disease. Experimental attention to this key, albeit largely ignored, environmental variable is likely to have a broad transformative effect on biomedical research.
Collapse
Affiliation(s)
- Christopher L Karp
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio 45229, USA
| |
Collapse
|
319
|
Pezzulo AA, Tang XX, Hoegger MJ, Abou Alaiwa MH, Ramachandran S, Moninger TO, Karp PH, Wohlford-Lenane CL, Haagsman HP, van Eijk M, Bánfi B, Horswill AR, Stoltz DA, McCray PB, Welsh MJ, Zabner J. Reduced airway surface pH impairs bacterial killing in the porcine cystic fibrosis lung. Nature 2012; 487:109-13. [PMID: 22763554 PMCID: PMC3390761 DOI: 10.1038/nature11130] [Citation(s) in RCA: 619] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/05/2012] [Indexed: 12/18/2022]
Abstract
Cystic fibrosis (CF) is a life-shortening disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene 1. Although bacterial lung infection and the resulting inflammation cause most of the morbidity and mortality, how loss of CFTR first disrupts airway host defense has remained uncertain 2–6. We asked what abnormalities impair eradication when a bacterium lands on the pristine surface of a newborn CF airway? To investigate these defects, we interrogated the viability of individual bacteria immobilized on solid grids and placed on the airway surface. As a model we studied CF pigs, which spontaneously develop hallmark features of CF lung disease 7,8. At birth, their lungs lack infection and inflammation, but have a reduced ability to eradicate bacteria 8. Here we show that in newborn wild-type pigs, the thin layer of airway surface liquid (ASL) rapidly killed bacteria in vivo, when removed from the lung, and in primary epithelial cultures. Lack of CFTR reduced bacterial killing. We found that ASL pH was more acidic in CF, and reducing pH inhibited the antimicrobial activity of ASL. Reducing ASL pH diminished bacterial killing in wild-type pigs, and increasing ASL pH rescued killing in CF pigs. These results directly link the initial host defense defect to loss of CFTR, an anion channel that facilitates HCO3− transport 9–13. Without CFTR, airway epithelial HCO3− secretion is defective, ASL pH falls and inhibits antimicrobial function, and thereby impairs killing of bacteria that enter the newborn lung. These findings suggest that increasing ASL pH might prevent the initial infection in patients with CF and that assaying bacterial killing could report on the benefit of therapeutic interventions.
Collapse
Affiliation(s)
- Alejandro A Pezzulo
- Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
320
|
Mou H, Zhao R, Sherwood R, Ahfeldt T, Lapey A, Wain J, Sicilian L, Izvolsky K, Musunuru K, Cowan C, Rajagopal J. Generation of multipotent lung and airway progenitors from mouse ESCs and patient-specific cystic fibrosis iPSCs. Cell Stem Cell 2012; 10:385-97. [PMID: 22482504 DOI: 10.1016/j.stem.2012.01.018] [Citation(s) in RCA: 240] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 12/15/2011] [Accepted: 01/19/2012] [Indexed: 12/15/2022]
Abstract
Deriving lung progenitors from patient-specific pluripotent cells is a key step in producing differentiated lung epithelium for disease modeling and transplantation. By mimicking the signaling events that occur during mouse lung development, we generated murine lung progenitors in a series of discrete steps. Definitive endoderm derived from mouse embryonic stem cells (ESCs) was converted into foregut endoderm, then into replicating Nkx2.1+ lung endoderm, and finally into multipotent embryonic lung progenitor and airway progenitor cells. We demonstrated that precisely-timed BMP, FGF, and WNT signaling are required for NKX2.1 induction. Mouse ESC-derived Nkx2.1+ progenitor cells formed respiratory epithelium (tracheospheres) when transplanted subcutaneously into mice. We then adapted this strategy to produce disease-specific lung progenitor cells from human Cystic Fibrosis induced pluripotent stem cells (iPSCs), creating a platform for dissecting human lung disease. These disease-specific human lung progenitors formed respiratory epithelium when subcutaneously engrafted into immunodeficient mice.
Collapse
Affiliation(s)
- Hongmei Mou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, 02114, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
321
|
Abu-El-Haija M, Ramachandran S, Meyerholz DK, Abu-El-Haija M, Griffin M, Giriyappa RL, Stoltz DA, Welsh MJ, McCray PB, Uc A. Pancreatic damage in fetal and newborn cystic fibrosis pigs involves the activation of inflammatory and remodeling pathways. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:499-507. [PMID: 22683312 DOI: 10.1016/j.ajpath.2012.04.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/04/2012] [Accepted: 04/12/2012] [Indexed: 01/28/2023]
Abstract
Pancreatic disease has onset in utero in humans with cystic fibrosis (CF), and progresses over time to complete destruction of the organ. The exact mechanisms leading to pancreatic damage in CF are incompletely understood. Inflammatory cells are present in the pancreas of newborn pigs with CF (CF pigs) and humans, which suggests that inflammation may have a role in the destructive process. We wondered whether tissue inflammation and genes associated with inflammatory pathways were increased in the pancreas of fetal CF pigs [83 to 90 days gestation (normal pig gestation is ~114 days)] and newborn pigs. Compared with fetal pigs without CF (non-CF pigs), in fetal CF pigs, the pancreas exhibited patchy inflammation and acinar atrophy, with progression in distribution and severity in neonatal CF pigs. Large-scale transcript profiling revealed that the pancreas in fetal and newborn CF pigs exhibited significantly increased expression of proinflammatory, complement cascade, and profibrotic genes when compared with fetal and newborn non-CF pigs. Acinar cells exhibited increased apoptosis in the pancreas of fetal and newborn CF pigs. α-Smooth muscle actin and transforming growth factor β1 were increased in both fetal and newborn CF pig pancreas, suggesting activation of profibrotic pathways. Cell proliferation and mucous cell metaplasia were detected in newborn, but not fetal, CF pigs, indicating that they were not an initiator of pathogenesis but a response. Proinflammatory, complement cascade, proapoptotic, and profibrotic pathways are activated in CF pig pancreas, and likely contribute to the destructive process.
Collapse
Affiliation(s)
- Maisam Abu-El-Haija
- Department of Pediatrics, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
Collawn JF, Lazrak A, Bebok Z, Matalon S. The CFTR and ENaC debate: how important is ENaC in CF lung disease? Am J Physiol Lung Cell Mol Physiol 2012; 302:L1141-6. [PMID: 22492740 PMCID: PMC3379041 DOI: 10.1152/ajplung.00036.2012] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 04/02/2012] [Indexed: 01/07/2023] Open
Abstract
Cystic fibrosis (CF) is caused by the loss of the cystic fibrosis transmembrane conductance regulator (CFTR) function and results in a respiratory phenotype that is characterized by dehydrated mucus and bacterial infections that affect CF patients throughout their lives. Much of the morbidity and mortality in CF results from a failure to clear bacteria from the lungs. What causes the defect in the bacterial clearance in the CF lung has been the subject of an ongoing debate. Here we discuss the arguments for and against the role of the epithelial sodium channel, ENaC, in the development of CF lung disease.
Collapse
Affiliation(s)
- James F Collawn
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, 35294-0005, USA.
| | | | | | | |
Collapse
|
323
|
Li X, Comellas AP, Karp PH, Ernst SE, Moninger TO, Gansemer ND, Taft PJ, Pezzulo AA, Rector MV, Rossen N, Stoltz DA, McCray PB, Welsh MJ, Zabner J. CFTR is required for maximal transepithelial liquid transport in pig alveolar epithelia. Am J Physiol Lung Cell Mol Physiol 2012; 303:L152-60. [PMID: 22637155 DOI: 10.1152/ajplung.00116.2012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A balance between alveolar liquid absorption and secretion is critical for maintaining optimal alveolar subphase liquid height and facilitating gas exchange in the alveolar space. However, the role of cystic fibrosis transmembrane regulator protein (CFTR) in this homeostatic process has remained elusive. Using a newly developed porcine model of cystic fibrosis, in which CFTR is absent, we investigated ion transport properties and alveolar liquid transport in isolated type II alveolar epithelial cells (T2AECs) cultured at the air-liquid interface. CFTR was distributed exclusively to the apical surface of cultured T2AECs. Alveolar epithelia from CFTR(-/-) pigs failed to increase liquid absorption in response to agents that increase cAMP, whereas cAMP-stimulated liquid absorption in CFTR(+/-) epithelia was similar to that in CFTR(+/+) epithelia. Expression of recombinant CFTR restored stimulated liquid absorption in CFTR(-/-) T2AECs but had no effect on CFTR(+/+) epithelia. In ex vivo studies of nonperfused lungs, stimulated liquid absorption was defective in CFTR(-/-) alveolar epithelia but similar between CFTR(+/+) and CFTR(+/-) epithelia. When epithelia were studied at the air-liquid interface, elevating cAMP levels increased subphase liquid height in CFTR(+/+) but not in CFTR(-/-) T2AECs. Our findings demonstrate that CFTR is required for maximal liquid absorption under cAMP stimulation, but it is not the rate-limiting factor. Furthermore, our data define a role for CFTR in liquid secretion by T2AECs. These insights may help to develop new treatment strategies for pulmonary edema and respiratory distress syndrome, diseases in which lung liquid transport is disrupted.
Collapse
Affiliation(s)
- Xiaopeng Li
- Department of Internal Medicine, University of Iowa, Iowa City, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
324
|
Fronius M, Clauss WG, Althaus M. Why Do We have to Move Fluid to be Able to Breathe? Front Physiol 2012; 3:146. [PMID: 22661953 PMCID: PMC3357553 DOI: 10.3389/fphys.2012.00146] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 05/01/2012] [Indexed: 12/11/2022] Open
Abstract
The ability to breathe air represents a fundamental step in vertebrate evolution that was accompanied by several anatomical and physiological adaptations. The morphology of the air-blood barrier is highly conserved within air-breathing vertebrates. It is formed by three different plies, which are represented by the alveolar epithelium, the basal lamina, and the endothelial layer. Besides these conserved morphological elements, another common feature of vertebrate lungs is that they contain a certain amount of fluid that covers the alveolar epithelium. The volume and composition of the alveolar fluid is regulated by transepithelial ion transport mechanisms expressed in alveolar epithelial cells. These transport mechanisms have been reviewed extensively. Therefore, the present review focuses on the properties and functional significance of the alveolar fluid. How does the fluid enter the alveoli? What is the fate of the fluid in the alveoli? What is the function of the alveolar fluid in the lungs? The review highlights the importance of the alveolar fluid, its volume and its composition. Maintenance of the fluid volume and composition within certain limits is critical to facilitate gas exchange. We propose that the alveolar fluid is an essential element of the air-blood barrier. Therefore, it is appropriate to refer to this barrier as being formed by four plies, namely (1) the thin fluid layer covering the apical membrane of the epithelial cells, (2) the epithelial cell layer, (3) the basal membrane, and (4) the endothelial cells.
Collapse
Affiliation(s)
- Martin Fronius
- Molecular Cell Physiology, Institute of Animal Physiology, Justus-Liebig-University Giessen Giessen, Germany
| | | | | |
Collapse
|
325
|
He L, Skirkanich J, Moronetti L, Lewis R, Lamitina T. The cystic-fibrosis-associated ΔF508 mutation confers post-transcriptional destabilization on the C. elegans ABC transporter PGP-3. Dis Model Mech 2012; 5:930-9. [PMID: 22569626 PMCID: PMC3484874 DOI: 10.1242/dmm.008987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Membrane proteins make up ∼30% of the proteome. During the early stages of maturation, this class of proteins can experience localized misfolding in distinct cellular compartments, such as the cytoplasm, endoplasmic reticulum (ER) lumen and ER membrane. ER quality control (ERQC) mechanisms monitor folding and determine whether a membrane protein is appropriately folded or is misfolded and warrants degradation. ERQC plays crucial roles in human diseases, such as cystic fibrosis, in which deletion of a single amino acid (F508) results in the misfolding and degradation of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl– channel. We introduced the ΔF508 mutation into Caenorhabditis elegans PGP-3, a 12-transmembrane ABC transporter with 15% identity to CFTR. When expressed in intestinal epithelial cells, PGP-3wt was stable and efficiently trafficked to the apical plasma membrane through a COPII-dependent mechanism. However, PGP-3ΔF508 was post-transcriptionally destabilized, resulting in reduced total and apical membrane protein levels. Genetic or physiological activation of the osmotic stress response pathway, which causes accumulation of the chemical chaperone glycerol, stabilized PGP-3ΔF508. Efficient degradation of PGP-3ΔF508 required the function of several C. elegans ER-associated degradation (ERAD) homologs, suggesting that destabilization occurs through an ERAD-type mechanism. Our studies show that the ΔF508 mutation causes post-transcriptional destabilization and degradation of PGP-3 in C. elegans epithelial cells. This model, combined with the power of C. elegans genetics, provides a new opportunity to genetically dissect metazoan ERQC.
Collapse
Affiliation(s)
- Liping He
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
326
|
Entezari M, Weiss DJ, Sitapara R, Whittaker L, Wargo MJ, Li J, Wang H, Yang H, Sharma L, Phan BD, Javdan M, Chavan SS, Miller EJ, Tracey KJ, Mantell LL. Inhibition of high-mobility group box 1 protein (HMGB1) enhances bacterial clearance and protects against Pseudomonas Aeruginosa pneumonia in cystic fibrosis. Mol Med 2012; 18:477-85. [PMID: 22314397 DOI: 10.2119/molmed.2012.00024] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 01/31/2012] [Indexed: 01/08/2023] Open
Abstract
Pulmonary infection with Pseudomonas aeruginosa and neutrophilic lung inflammation significantly contribute to morbidity and mortality in cystic fibrosis (CF). High-mobility group box 1 protein (HMGB1), a ubiquitous DNA binding protein that promotes inflammatory tissue injury, is significantly elevated in CF sputum. However, its mechanistic and potential therapeutic implications in CF were previously unknown. We found that HMGB1 levels were significantly elevated in bronchoalveolar lavage fluids (BALs) of CF patients and cystic fibrosis transmembrane conductance regulator (CFTR )(-/-) mice. Neutralizing anti-HMGB1 monoclonal antibody (mAb) conferred significant protection against P. aeruginosa-induced neutrophil recruitment, lung injury and bacterial infection in both CFTR(-/-) and wild-type mice. Alveolar macrophages isolated from mice treated with anti-HMGB1 mAb had improved phagocytic activity, which was suppressed by direct exposure to HMGB1. In addition, BAL from CF patients significantly impaired macrophage phagocytotic function, and this impairment was attenuated by HMGB1-neutralizing antibodies. The HMGB1-mediated suppression of bacterial phagocytosis was attenuated in macrophages lacking toll-like receptor (TLR)-4, suggesting a critical role for TLR4 in signaling HMGB1-mediated macrophage dysfunction. These studies demonstrate that the elevated levels of HMGB1 in CF airways are critical for neutrophil recruitment and persistent presence of P. aeruginosa in the lung. Thus, HMGB1 may provide a therapeutic target for reducing bacterial infection and lung inflammation in CF.
Collapse
Affiliation(s)
- Maria Entezari
- Cardiopulmonary Toxicology, Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Allied Health Professions, Queens, New York 11439, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
327
|
Fisher JT, Liu X, Yan Z, Luo M, Zhang Y, Zhou W, Lee BJ, Song Y, Guo C, Wang Y, Lukacs GL, Engelhardt JF. Comparative processing and function of human and ferret cystic fibrosis transmembrane conductance regulator. J Biol Chem 2012; 287:21673-85. [PMID: 22570484 DOI: 10.1074/jbc.m111.336537] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The most common cystic fibrosis transmembrane conductance regulator (CFTR) gene mutation is ΔF508, and this causes cystic fibrosis (CF). New CF models in the pig and ferret have been generated that develop lung, pancreatic, liver, and intestinal pathologies that reflect disease in CF patients. Species-specific biology in the processing of CFTR has demonstrated that pig and mouse ΔF508-CFTR proteins are more effectively processed to the apical membrane of airway epithelia than human ΔF508-CFTR. The processing behavior of ferret WT- and ΔF508-CFTR proteins remains unknown, and such information is important to predicting the utility of a ΔF508-CFTR ferret. To this end, we sought to compare processing, membrane stability, and function of human and ferret WT- and ΔF508-CFTR proteins in a heterologous expression system using HT1080, HEK293T, BHK21, and Cos7 cells as well as human and ferret CF polarized airway epithelia. Analysis of the protein processing and stability by metabolic pulse-chase and surface On-Cell Western blots revealed that WT-fCFTR half-life and membrane stability were increased relative to WT-hCFTR. Furthermore, in BHK21, Cos7, and CuFi cells, human and ferret ΔF508-CFTR processing was negligible, whereas low levels of processing of ΔF508-fCFTR could be seen in HT1080 and HEK293T cells. Only the WT-fCFTR, but not ΔF508-fCFTR, produced functional cAMP-inducible chloride currents in both CF human and ferret airway epithelia. Further elucidation of the mechanism responsible for elevated fCFTR protein stability may lead to new therapeutic approaches to augment CFTR function. These findings also suggest that generation of a ferret CFTR(ΔF508/ΔF508) animal model may be useful.
Collapse
Affiliation(s)
- John T Fisher
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
328
|
Rosenfeld M, Emerson J, McNamara S, Thompson V, Ramsey BW, Morgan W, Gibson RL. Risk factors for age at initial Pseudomonas acquisition in the cystic fibrosis epic observational cohort. J Cyst Fibros 2012; 11:446-53. [PMID: 22554417 DOI: 10.1016/j.jcf.2012.04.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 04/03/2012] [Accepted: 04/05/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND Risk factors for initial Pseudomonas aeruginosa (Pa) acquisition, particularly environmental exposures, are poorly understood. We aimed to identify such risk factors in order to inform prevention strategies and identify high-risk populations. METHODS The study cohort included all participants in the U.S. EPIC Observational Study who had no prior Pa-positive respiratory cultures (N=889). Cox proportional hazard models were used to test the effects of factors on age at first Pa-positive respiratory culture. RESULTS Cystic fibrosis (CF) genotype functional class had an important effect on age at initial Pa acquisition (hazard ratio (HR) comparing minimal to residual CFTR function 2.87 (95% CI 1.88, 4.39)). None of the modifiable risk factors evaluated, including cigarette smoke, hot tub use, breastfeeding, or daycare, was associated with age at Pa acquisition. Similarly, newborn screening was not associated with age at Pa acquisition (HR 0.85, 95% CI 0.66, 1.09). Key associations were validated in a CF Foundation National Patient Registry replication cohort. CONCLUSIONS Given the ubiquitous presence of Pa in the environment, it may be that many imposed lifestyle changes will have less impact on age at initial Pa acquisition than genetic determinants.
Collapse
Affiliation(s)
- Margaret Rosenfeld
- Department of Pediatrics, Seattle Children's Hospital and University of Washington, Seattle, WA 98105, USA.
| | | | | | | | | | | | | | | |
Collapse
|
329
|
Han MK, Huang YJ, Lipuma JJ, Boushey HA, Boucher RC, Cookson WO, Curtis JL, Erb-Downward J, Lynch SV, Sethi S, Toews GB, Young VB, Wolfgang MC, Huffnagle GB, Martinez FJ. Significance of the microbiome in obstructive lung disease. Thorax 2012; 67:456-63. [PMID: 22318161 PMCID: PMC3578398 DOI: 10.1136/thoraxjnl-2011-201183] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The composition of the lung microbiome contributes to both health and disease, including obstructive lung disease. Because it has been estimated that over 70% of the bacterial species on body surfaces cannot be cultured by currently available techniques, traditional culture techniques are no longer the gold standard for microbial investigation. Advanced techniques that identify bacterial sequences, including the 16S ribosomal RNA gene, have provided new insights into the depth and breadth of microbiota present both in the diseased and normal lung. In asthma, the composition of the microbiome of the lung and gut during early childhood development may play a key role in the development of asthma, while specific airway microbiota are associated with chronic asthma in adults. Early bacterial stimulation appears to reduce asthma susceptibility by helping the immune system develop lifelong tolerance to innocuous antigens. By contrast, perturbations in the microbiome from antibiotic use may increase the risk for asthma development. In chronic obstructive pulmonary disease, bacterial colonisation has been associated with a chronic bronchitic phenotype, increased risk of exacerbations, and accelerated loss of lung function. In cystic fibrosis, studies utilising culture-independent methods have identified associations between decreased bacterial community diversity and reduced lung function; colonisation with Pseudomonas aeruginosa has been associated with the presence of certain CFTR mutations. Genomic analysis of the lung microbiome is a young field, but has the potential to define the relationship between lung microbiome composition and disease course. Whether we can manipulate bacterial communities to improve clinical outcomes remains to be seen.
Collapse
Affiliation(s)
- Meilan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, 3916 Taubman Center, Box 5360, 1500 E. Medical Center Drive, Ann Arbor, MI 48109-5360, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Airway epithelial cells--hyperabsorption in CF? Int J Biochem Cell Biol 2012; 44:1232-5. [PMID: 22542896 DOI: 10.1016/j.biocel.2012.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Revised: 03/06/2012] [Accepted: 04/16/2012] [Indexed: 11/23/2022]
Abstract
Airway epithelial cells transport electrolytes and are central to the disease cystic fibrosis (CF), which is an inherited transport defect affecting smaller airways and a number of other epithelial organs. Clinically, CF is dominated by a chronic lung disease, the main cause of morbidity and mortality. Airway obstruction by thick mucus and chronic infection by Pseudomonas aeruginosa eventually lead to loss of pulmonary function. Loss of function of CFTR Cl(-) channels was found to be the cause for CF. However, intensive research on the detailed mechanism of CF lung disease for more than 25 years produced a bewildering number of hypotheses and an endless discussion whether reduced Cl(-) secretion, primarily located in airway submucosal glands, or dehydration of the airways, driven by a hyperabsorption of Na(+) ions, is the primary cause of the disease. Recent results suggest a fine-tuned regulation of the airway fluid layer, but how significant really are Cl(-) and Na(+) transport?
Collapse
|
331
|
Komor U, Bielecki P, Loessner H, Rohde M, Wolf K, Westphal K, Weiss S, Häussler S. Biofilm formation by Pseudomonas aeruginosa in solid murine tumors - a novel model system. Microbes Infect 2012; 14:951-8. [PMID: 22542841 DOI: 10.1016/j.micinf.2012.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/20/2012] [Accepted: 04/02/2012] [Indexed: 01/12/2023]
Abstract
The ability of opportunistic bacterial pathogens to grow in biofilms is decisive in the pathogenesis of chronic infectious diseases. Growth within biofilms does not only protect the bacteria against the host immune system but also from the killing by antimicrobial agents. Here, we introduce a mouse model in which intravenously administered planktonic Pseudomonas aeruginosa bacteria are enriched in transplantable subcutaneous mouse tumors. Electron microscopy images provide evidence that such bacteria reside in the tumor tissue within biofilm structures. Immunohistology furthermore demonstrated that infection of the tumor tissue elicits a host response characterized by strong neutrophilic influx. Interestingly, the biofilm defective PA14 pqsA transposon mutant formed less biofilm in vivo and was more susceptible to clearance by intravenous ciprofloxacin treatment as compared to the wild-type control. In conclusion, we have established an experimentally tractable model that may serve to identify novel bacterial and host factors important for in vivo biofilm formation and to re-evaluate bactericidal and anti-biofilm effects of currently used and novel antibacterial compounds.
Collapse
Affiliation(s)
- Uliana Komor
- Molecular Immunology, Helmholtz Center for Infection Research, Inhoffenstrasse 7, Braunschweig 38124, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
332
|
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) functions as a channel that regulates the transport of ions and the movement of water across the epithelial barrier. Mutations in CFTR, which form the basis for the clinical manifestations of cystic fibrosis, affect the epithelial innate immune function in the lung, resulting in exaggerated and ineffective airway inflammation that fails to eradicate pulmonary pathogens. Compounding the effects of excessive neutrophil recruitment, the mutant CFTR channel does not transport antioxidants to counteract neutrophil-associated oxidative stress. Whereas mutant CFTR expression in leukocytes outside of the lung does not markedly impair their function, the expected regulation of inflammation in the airways is clearly deficient in cystic fibrosis. The resulting bacterial infections, which are caused by organisms that have substantial genetic and metabolic flexibility, can resist multiple classes of antibiotics and evade phagocytic clearance. The development of animal models that approximate the human pulmonary phenotypes-airway inflammation and spontaneous infection-may provide the much-needed tools to establish how CFTR regulates mucosal immunity and to test directly the effect of pharmacologic potentiation and correction of mutant CFTR function on bacterial clearance.
Collapse
|
333
|
A review of pathophysiology and management of fetuses and neonates with meconium ileus for the pediatric surgeon. J Pediatr Surg 2012; 47:772-81. [PMID: 22498395 DOI: 10.1016/j.jpedsurg.2012.02.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Meconium ileus (MI) is the earliest clinical manifestation of cystic fibrosis (CF), occurring in up to 20% of patients with CF. Our aim was to review and integrate current knowledge about the diagnosis and management of fetuses and neonates with MI that may aid the pediatric surgeon in caring for these patients. METHODS We identified areas of interest including pathophysiology, prenatal diagnosis, nonoperative and operative management, postoperative management, and prognosis. We performed a Medline search using the search term meconium ileus for English language articles published in the last 20 years. We reviewed reference lists to identify other articles of historical significance. RESULTS Meconium ileus is primarily associated with CF transmembrane (conductance) regulator mutations F508del, G542X, W1282X, R553X, and G551D, and modifier genes have been found to explain approximately 17% of the phenotypic variability. Mouse, pig, and ferret models for CF demonstrate neonatal bowel obstruction mimicking MI. Sonographic findings of hyperechoic masses and dilated bowel in a high-risk fetus are suggestive of MI. Less than 7% of low-risk fetuses with hyperechoic bowel will have MI. Contemporary series of noninvasive management with Gastrografin enema report success rates of 36% to 39%, significantly lower than historical values. The optimal surgical technique remains controversial, although primary anastomosis results in surgical complication rates between 21% and 31%, higher than those noted with delayed anastomosis. Pulmonary function for patients with CF and MI at 15 and 25 years old is similar to those without MI, although height and weight percentiles may be lower. CONCLUSIONS This review for pediatric surgeons presents an examination of the literature and synthesizes current information about the pathophysiology, prenatal diagnosis, nonoperative and operative management, postoperative management, and prognosis of the patient with CF and MI.
Collapse
|
334
|
Gould NS, Min E, Martin RJ, Day BJ. CFTR is the primary known apical glutathione transporter involved in cigarette smoke-induced adaptive responses in the lung. Free Radic Biol Med 2012; 52:1201-6. [PMID: 22266045 PMCID: PMC3920665 DOI: 10.1016/j.freeradbiomed.2012.01.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 12/14/2011] [Accepted: 01/04/2012] [Indexed: 10/14/2022]
Abstract
One of the most abundant antioxidants in the lung is glutathione (GSH), a low-molecular-weight thiol, which functions to attenuate both oxidative stress and inflammation. GSH is concentrated in the epithelial lining fluid (ELF) of the lung and can be elevated in response to the increased oxidant burden from cigarette smoke (CS). However, the transporter(s) responsible for the increase in ELF GSH with cigarette smoke is not known. Three candidate apical GSH transporters in the lung are CFTR, BCRP, and MRP2, but their potential roles in ELF GSH transport in response to CS have not been investigated. In vitro, the inhibition of CFTR, BCRP, or MRP2 resulted in decreased GSH efflux in response to cigarette smoke extract. In vivo, mice deficient in CFTR, BCRP, or MRP2 were exposed to either air or acute CS. CFTR-deficient mice had reduced basal and CS-induced GSH in the ELF, whereas BCRP or MRP2 deficiency had no effect on ELF GSH basal or CS-exposed levels. Furthermore, BCRP or MRP2 deficiency had little effect on lung tissue GSH. These data indicate that CFTR is predominantly involved in maintaining basal ELF GSH and increasing ELF GSH in response to CS.
Collapse
Affiliation(s)
- Neal S. Gould
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO
| | - Elysia Min
- Department of Medicine, National Jewish Health, Denver, CO
| | - Richard J. Martin
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Medicine, University of Colorado Denver, Aurora, CO
| | - Brian J. Day
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO
- Department of Medicine, University of Colorado Denver, Aurora, CO
- Department of Immunology, University of Colorado Denver, Aurora, CO
| |
Collapse
|
335
|
Asokan A, Schaffer DV, Jude Samulski R. The AAV vector toolkit: poised at the clinical crossroads. Mol Ther 2012; 20:699-708. [PMID: 22273577 PMCID: PMC3321598 DOI: 10.1038/mt.2011.287] [Citation(s) in RCA: 343] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 12/02/2011] [Indexed: 12/14/2022] Open
Abstract
The discovery of naturally occurring adeno-associated virus (AAV) isolates in different animal species and the generation of engineered AAV strains using molecular genetics tools have yielded a versatile AAV vector toolkit. Promising results in preclinical animal models of human disease spurred the much awaited transition toward clinical application, and early successes in phase I/II clinical trials for a broad spectrum of genetic diseases have recently been reported. As the gene therapy community forges ahead with cautious optimism, both preclinical and clinical studies using first generation AAV vectors have highlighted potential challenges. These include cross-species variation in vector tissue tropism and gene transfer efficiency, pre-existing humoral immunity to AAV capsids and vector dose-dependent toxicity in patients. A battery of second generation AAV vectors, engineered through rational and combinatorial approaches to address the aforementioned concerns, are now available. This review will provide an overview of preclinical studies with the ever-expanding AAV vector portfolio in large animal models and an update on new lead AAV vector candidates poised for clinical translation.
Collapse
Affiliation(s)
- Aravind Asokan
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David V Schaffer
- Department of Chemical Engineering, University of California, Berkeley, California, USA
- The Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| | - R Jude Samulski
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
336
|
Abstract
Cystic fibrosis (CF) is an inherited chronic disease that remains a common cause of morbidity and mortality in affected patients, mostly in the young. A wealth of knowledge has been gained into the genetics, pathophysiology, and clinical manifestation of the disease. In parallel with these new insights into the disease, novel treatments have been developed or are under development that have had a major impact on quality of life and survival. Improvement in the delivery of care to patients in CF centers, using a team-based approach, and constant review of process, and by quality improvement projects, have also had an impact on outcomes in CF.
Collapse
Affiliation(s)
- Jason Lobo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, 27599-7020, USA
| | | | | |
Collapse
|
337
|
Lacruz RS, Smith CE, Moffatt P, Chang EH, Bromage TG, Bringas P, Nanci A, Baniwal SK, Zabner J, Welsh MJ, Kurtz I, Paine ML. Requirements for ion and solute transport, and pH regulation during enamel maturation. J Cell Physiol 2012; 227:1776-85. [PMID: 21732355 DOI: 10.1002/jcp.22911] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Transcellular bicarbonate transport is suspected to be an important pathway used by ameloblasts to regulate extracellular pH and support crystal growth during enamel maturation. Proteins that play a role in amelogenesis include members of the ABC transporters (SLC gene family and CFTR). A number of carbonic anhydrases (CAs) have also been identified. The defined functions of these genes are likely interlinked during enamel mineralization. The purpose of this study is to quantify relative mRNA levels of individual SLC, Cftr, and CAs in enamel cells obtained from secretory and maturation stages on rat incisors. We also present novel data on the enamel phenotypes for two animal models, a mutant porcine (CFTR-ΔF508) and the NBCe1-null mouse. Our data show that two SLCs (AE2 and NBCe1), Cftr, and Car2, Car3, Car6, and Car12 are all significantly up-regulated at the onset of the maturation stage of amelogenesis when compared to the secretory stage. The remaining SLCs and CA gene transcripts showed negligible expression or no significant change in expression from secretory to maturation stages. The enamel of CFTR-ΔF508 adult pigs was hypomineralized and showed abnormal crystal growth. NBCe1-null mice enamel was structurally defective and had a marked decrease in mineral content relative to wild-type. These data demonstrate the importance of many non-matrix proteins to amelogenesis and that the expression levels of multiple genes regulating extracellular pH are modulated during enamel maturation in response to an increased need for pH buffering during hydroxyapatite crystal growth.
Collapse
Affiliation(s)
- Rodrigo S Lacruz
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California 90033, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
338
|
A novel neutrophil derived inflammatory biomarker of pulmonary exacerbation in cystic fibrosis. J Cyst Fibros 2012; 11:100-7. [DOI: 10.1016/j.jcf.2011.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 09/21/2011] [Accepted: 09/27/2011] [Indexed: 11/17/2022]
|
339
|
Oakland M, Sinn PL, McCray PB. Advances in cell and gene-based therapies for cystic fibrosis lung disease. Mol Ther 2012; 20:1108-15. [PMID: 22371844 DOI: 10.1038/mt.2012.32] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis (CF) is a disease characterized by airway infection, inflammation, remodeling, and obstruction that gradually destroy the lungs. Direct delivery of the cystic fibrosis transmembrane conductance regulator (CFTR) gene to airway epithelia may offer advantages, as the tissue is accessible for topical delivery of vectors. Yet, physical and host immune barriers in the lung present challenges for successful gene transfer to the respiratory tract. Advances in gene transfer approaches, tissue engineering, and novel animal models are generating excitement within the CF research field. This review discusses current challenges and advancements in viral and nonviral vectors, cell-based therapies, and CF animal models.
Collapse
Affiliation(s)
- Mayumi Oakland
- Department of Microbiology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
340
|
Ramsey BW, Banks-Schlegel S, Accurso FJ, Boucher RC, Cutting GR, Engelhardt JF, Guggino WB, Karp CL, Knowles MR, Kolls JK, LiPuma JJ, Lynch S, McCray PB, Rubenstein RC, Singh PK, Sorscher E, Welsh M. Future directions in early cystic fibrosis lung disease research: an NHLBI workshop report. Am J Respir Crit Care Med 2012; 185:887-92. [PMID: 22312017 DOI: 10.1164/rccm.201111-2068ws] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Since the 1989 discovery that mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF), there has been substantial progress toward understanding the molecular basis for CF lung disease, leading to the discovery and development of new therapeutic approaches. However, the earliest impact of the loss of CFTR function on airway physiology and structure and its relationship to initial infection and inflammation are poorly understood. Universal newborn screening for CF in the United States represents an unprecedented opportunity for investigating CF clinical manifestations very early in life. Recently developed animal models with pulmonary phenotypic manifestations also provide a window into the early consequences of this genetic disorder. For these reasons, the National Heart, Lung, and Blood Institute (NHLBI) convened a working group of extramural experts, entitled "Future Research Directions in Early CF Lung Disease" on September 21-22, 2010, to identify future research directions of great promise in CF. The priority areas identified included (1) exploring pathogenic mechanisms of early CF lung disease; (2) leveraging newborn screening to elucidate the natural history of early lung disease; (3) developing a spectrum of biomarkers of early lung disease that reflects CF pathophysiology, clinical outcome, and response to treatment; (4) exploring the role of genetics/genomics (e.g., modifier genes, gene-environmental interactions, and epigenetics) in early CF pathogenesis; (5) defining early microbiological events in CF lung disease; and (6) elucidating the initial airway inflammatory, remodeling, and repair mechanisms in CF lung disease.
Collapse
|
341
|
Callaghan M, McClean S. Bacterial host interactions in cystic fibrosis. Curr Opin Microbiol 2012; 15:71-7. [DOI: 10.1016/j.mib.2011.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 11/02/2011] [Accepted: 11/02/2011] [Indexed: 01/01/2023]
|
342
|
Cao H, Molday RS, Hu J. Gene therapy: light is finally in the tunnel. Protein Cell 2012; 2:973-89. [PMID: 22231356 DOI: 10.1007/s13238-011-1126-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 11/27/2011] [Indexed: 01/23/2023] Open
Abstract
After two decades of ups and downs, gene therapy has recently achieved a milestone in treating patients with Leber's congenital amaurosis (LCA). LCA is a group of inherited blinding diseases with retinal degeneration and severe vision loss in early infancy. Mutations in several genes, including RPE65, cause the disease. Using adeno-associated virus as a vector, three independent teams of investigators have recently shown that RPE65 can be delivered to retinal pigment epithelial cells of LCA patients by subretinal injections resulting in clinical benefits without side effects. However, considering the whole field of gene therapy, there are still major obstacles to clinical applications for other diseases. These obstacles include innate and immune barriers to vector delivery, toxicity of vectors and the lack of sustained therapeutic gene expression. Therefore, new strategies are needed to overcome these hurdles for achieving safe and effective gene therapy. In this article, we shall review the major advancements over the past two decades and, using lung gene therapy as an example, discuss the current obstacles and possible solutions to provide a roadmap for future gene therapy research.
Collapse
Affiliation(s)
- Huibi Cao
- Programme in Physiology and Experimental Medicine, Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5G, 1X8, Canada
| | | | | |
Collapse
|
343
|
Abstract
PURPOSE OF REVIEW Cystic fibrosis is the first human genetic disease to benefit from the directed engineering of three different species of animal models (mice, pigs, and ferrets). Recent studies on the cystic fibrosis pig and ferret models are providing new information about the pathophysiology of cystic fibrosis in various organ systems. Additionally, new conditional cystic fibrosis transmembrane conductance regulator (CFTR) knockout mice are teaching unexpected lessons about CFTR function in surprising cellular locations. Comparisons between these animal models and the human condition are key to dissecting the complexities of disease pathophysiology in cystic fibrosis. RECENT FINDINGS Cystic fibrosis pigs and ferrets have provided new models to study the spontaneous development of disease in the lung and pancreas, two organs that are largely spared overt spontaneous disease in cystic fibrosis mice. New cystic fibrosis mouse models are now interrogating CFTR functions involved in growth and inflammation at an organ-based level using conditional knockout technology. Together, these models are providing new insights on the human condition. SUMMARY Basic and clinical cystic fibrosis research will benefit greatly from the comparative pathophysiology of cystic fibrosis mice, pigs, and ferrets. Both similarities and differences between these three cystic fibrosis models will inform pathophysiologically important mechanisms of CFTR function in humans and aid in the development of both organ-specific and general therapies for cystic fibrosis.
Collapse
|
344
|
Plog S, Grötzsch T, Klymiuk N, Kobalz U, Gruber AD, Mundhenk L. The porcine chloride channel calcium-activated family member pCLCA4a mirrors lung expression of the human hCLCA4. J Histochem Cytochem 2012; 60:45-56. [PMID: 22205680 PMCID: PMC3283134 DOI: 10.1369/0022155411426455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 09/17/2011] [Indexed: 11/22/2022] Open
Abstract
Pig models of cystic fibrosis (CF) have recently been established that are expected to mimic the human disease closer than mouse models do. The human CLCA (originally named chloride channels, calcium-activated) member hCLCA4 is considered a potential modifier of disease severity in CF, but its murine ortholog, mCLCA6, is not expressed in the mouse lung. Here, we have characterized the genomic structure, protein processing, and tissue expression patterns of the porcine ortholog to hCLCA4, pCLCA4a. The genomic structure and cellular protein processing of pCLCA4a were found to closely mirror those of hCLCA4 and mCLCA6. Similar to human lung, pCLCA4a mRNA was strongly expressed in porcine lungs, and the pCLCA4a protein was immunohistochemically detected on the apical membranes of tracheal and bronchial epithelial cells. This stands in sharp contrast to mouse mCLCA6, which has been detected exclusively in intestinal epithelia but not the murine lung. The results may add to the understanding of species-specific differences in the CF phenotype and support the notion that the CF pig model may be more suitable than murine models to study the role of hCLCA4.
Collapse
Affiliation(s)
- Stephanie Plog
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
345
|
Sequential targeting of CFTR by BAC vectors generates a novel pig model of cystic fibrosis. J Mol Med (Berl) 2011; 90:597-608. [PMID: 22170306 DOI: 10.1007/s00109-011-0839-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 11/24/2011] [Accepted: 11/25/2011] [Indexed: 01/16/2023]
Abstract
Cystic fibrosis (CF) is the most common lethal inherited disease in Caucasians and is caused by mutations in the CFTR gene. The disease is incurable and medical treatment is limited to the amelioration of symptoms or secondary complications. A comprehensive understanding of the disease mechanisms and the development of novel treatment options require appropriate animal models. Existing CF mouse models fail to reflect important aspects of human CF. We thus generated a CF pig model by inactivating the CFTR gene in primary porcine cells by sequential targeting using modified bacterial artificial chromosome vectors. These cells were then used to generate homozygous CFTR mutant piglets by somatic cell nuclear transfer. The homozygous CFTR mutants lack CFTR protein expression and display severe malformations in the intestine, respiratory tract, pancreas, liver, gallbladder, and male reproductive tract. These phenotypic abnormalities closely resemble both the human CF pathology as well as alterations observed in a recently published CF pig model which was generated by a different gene targeting strategy. Our new CF pig model underlines the value of the CFTR-deficient pig for gaining new insight into the disease mechanisms of CF and for the development and evaluation of new therapeutic strategies. This model will furthermore increase the availability of CF pigs to the scientific community.
Collapse
|
346
|
Abu-El-Haija M, Sinkora M, Meyerholz DK, Welsh MJ, McCray, Jr. PB, Butler J, Uc A. An activated immune and inflammatory response targets the pancreas of newborn pigs with cystic fibrosis. Pancreatology 2011; 11:506-15. [PMID: 22057257 PMCID: PMC3224519 DOI: 10.1159/000332582] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 08/29/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIMS In cystic fibrosis (CF), pancreatic disease begins in utero and progresses over time to complete destruction of the organ. Although inflammatory cells have been detected in the pancreas of humans and pigs with CF, their subtypes have not been characterized. METHODS Using four-color flow cytometry, we analyzed the surface antigens of leukocytes in pancreas, blood, and mesenteric lymph nodes (MLN) of newborn pigs with CF (CFTR(-/-) and CFTR(Δ)(F508/)(Δ)(F508)) and in those without CF (CFTR(+/-), CFTR(+/)(Δ)(F508), CFTR(+/+)). Pancreatic histopathology was examined with HE stain. RESULTS CF pig pancreas had patchy distribution of inflammatory cells with neutrophils/macrophages in dilated acini, and lymphocytes in the interstitium compared to non-CF. B cells, effector (MHC-II(+)) and cytotoxic (CD2(+)CD8(+)) γδ T cells, activated (MHC-II(+) and/or CD25(+)) and effector (CD4(+)CD8(+)) αβ T helper cells, effector natural killer cells (MHC-II(+)CD3(-)CD8(+)), and monocytes/macrophages and neutrophils were increased in the CF pig pancreas compared to pigs without CF. Blood and MLN leukocyte populations were not different between CF and non-CF pigs. CONCLUSIONS We discovered an activated immune response that was specific to the pancreas of newborn CF pigs; inflammation was not systemic. The presence of both innate and adaptive immune cells suggests that the disease process is complex and extensive.
Collapse
Affiliation(s)
- Maisam Abu-El-Haija
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Marek Sinkora
- Department of Immunology and Gnotogiology, Institute of Microbiology, Academy of Sciences of the Czech Republic, v.v.i., Novy Hradek, Czech Republic
| | - David K. Meyerholz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Michael J. Welsh
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA,Department of Howard Hughes Medical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Paul B. McCray, Jr.
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - John Butler
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Aliye Uc
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA,*Aliye Uc, MD, 2865 JPP Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242 (USA), Tel. +1 319 384 6032, E-Mail
| |
Collapse
|
347
|
Grubb BR, O'Neal WK, Ostrowski LE, Kreda SM, Button B, Boucher RC. Transgenic hCFTR expression fails to correct β-ENaC mouse lung disease. Am J Physiol Lung Cell Mol Physiol 2011; 302:L238-47. [PMID: 22003093 DOI: 10.1152/ajplung.00083.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The relationships between airway epithelial Cl(-) secretion-Na(+) absorption balance, airway surface liquid (ASL) homeostasis, and lung disease were investigated in selected transgenic mice. 1) To determine if transgenic overexpression of wild-type (WT) human CFTR (hCFTR) accelerated Cl(-) secretion and regulated Na(+) absorption in murine airways, we utilized a Clara cell secretory protein (CCSP)-specific promoter to generate mice expressing airway-specific hCFTR. Ussing chamber studies revealed significantly (∼2.5-fold) elevated basal Cl(-) secretory currents in CCSP-hCFTR transgenic mouse airways. Endogenous murine airway Na(+) absorption was not regulated by hCFTR, and these mice exhibited no lung disease. 2) We tested whether hCFTR, transgenically expressed on a transgenic mouse background overexpressing the β-subunit of the epithelial Na(+) channel (β-ENaC), restored ion transport balance and ASL volume homeostasis and ameliorated lung disease. Both transgenes were active in CCSP-hCFTR/β-ENaC transgenic mouse airways, which exhibited an elevated basal Cl(-) secretion and Na(+) hyperabsorption. However, the airway disease characteristic of β-ENaC mice persisted. Confocal studies of ASL volume homeostasis in cultured tracheal cells revealed ASL autoregulation to a height of ∼6 μm in WT and CCSP-hCFTR cultures, whereas ASL was reduced to <4 μm in β-ENaC and CCSP-hCFTR/β-ENaC cultures. We conclude that 1) hCFTR overexpression increases basal Cl(-) secretion but does not regulate Na(+) transport in WT mice and 2) transgenic hCFTR produces increased Cl(-) secretion, but not regulation of Na(+) channels, in β-ENaC mouse airways and does not ameliorate β-ENaC mouse lung disease.
Collapse
Affiliation(s)
- B R Grubb
- Cystic Fibrosis/Pulmonary Research and Treatment Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7248, USA.
| | | | | | | | | | | |
Collapse
|
348
|
Abstract
The thyroid hormones thyroxine (T(4)) and triiodothyronine (T(3)) play key roles in regulating development, growth and metabolism in pre- and postnatal life. Iodide (I(-)) is an essential component of the thyroid hormones and is accumulated avidly by the thyroid gland. The rarity of elemental iodine and I(-) in the environment challenges the thyroid to orchestrate a remarkable series of transport processes that ultimately ensure sufficient levels for hormone synthesis. In addition to actively extracting circulating I(-), thyroid follicular epithelial cells must also translocate I(-) into a central intrafollicular compartment, where thyroglobulin is iodinated to form the protein precursor to T(4) and T(3). In the last decade, several bodies of evidence render questionable the notion that I(-) exits thyrocytes solely via the Cl(-)/I(-) exchanger Pendrin (SLC26A4), therefore necessitating reconsideration of several other candidate I(-) conduits: the Cl(-)/H(+) antiporter, CLC-5, the cystic fibrosis transmembrane conductance regulator (CFTR) and the sodium monocarboxylic acid transporter (SMCT1).
Collapse
Affiliation(s)
- Peying Fong
- Department of Anatomy and Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA.
| |
Collapse
|
349
|
Whyte JJ, Prather RS. Genetic modifications of pigs for medicine and agriculture. Mol Reprod Dev 2011; 78:879-91. [PMID: 21671302 PMCID: PMC3522184 DOI: 10.1002/mrd.21333] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 05/09/2011] [Indexed: 12/18/2022]
Abstract
Genetically modified swine hold great promise in the fields of agriculture and medicine. Currently, these swine are being used to optimize production of quality meat, to improve our understanding of the biology of disease resistance, and to reduced waste. In the field of biomedicine, swine are anatomically and physiologically analogous to humans. Alterations of key swine genes in disease pathways provide model animals to improve our understanding of the causes and potential treatments of many human genetic disorders. The completed sequencing of the swine genome will significantly enhance the specificity of genetic modifications, and allow for more accurate representations of human disease based on syntenic genes between the two species. Improvements in both methods of gene alteration and efficiency of model animal production are key to enabling routine use of these swine models in medicine and agriculture.
Collapse
Affiliation(s)
- Jeffrey J. Whyte
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, U.S.A
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, U.S.A
- Division of Animal Science, University of Missouri, Columbia, MO, U.S.A
| | - Randall S. Prather
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, U.S.A
- Division of Animal Science, University of Missouri, Columbia, MO, U.S.A
| |
Collapse
|
350
|
Li Y, Ganta S, Fong P. Endogenous surface expression of ΔF508-CFTR mediates cAMP-stimulated Cl(-) current in CFTR(ΔF508/ΔF508) pig thyroid epithelial cells. Exp Physiol 2011; 97:115-24. [PMID: 21948195 DOI: 10.1113/expphysiol.2011.060756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is both an anion channel and a regulator of other transport proteins. Mutations in the CFTR gene underlie the human disease, cystic fibrosis. The most common CFTR mutation, ΔF508, produces a misfolded protein which traffics improperly. The availability of transgenic CFTR(ΔF508/ΔF508) pigs allows measurement of the impact of ΔF508 in native tissue. Thyroid epithelia respond to cAMP-elevating agents by increasing anion transport, a process reliant on functional CFTR. To assess whether endogenous levels of ΔF508-CFTR mediate thyroid transport, primary thyroid epithelial cultures (pThECs) were grown from newborn CFTR(+/+) (wild-type) and CFTR(ΔF508/ΔF508) (ΔF) pig thyroids and the stimulated, secretory components of short-circuit current (I(sc)) compared. Surface biotinylation studies assessed the surface presentation of ΔF508-CFTR. Baseline I(sc) levels of both wild-type and ΔF pThECs consisted of an amiloride-sensitive component. In ΔF pThECs, this mirrored previous measurements in CFTR(-/-) (knockout) pThECs. Surprisingly, elevation of cAMP transiently increased I(sc) to peak levels ∼65% of those achieved by wild-type. In contrast, knockout pThECs were indifferent to cAMP activation. In ΔF pThECs, total ΔF508-CFTR expression was ∼9% that of wild-type, consistent with misfolding and enhanced degradation. Surface biotinylation studies indicated that ∼4% of the total ΔF508 resided at the surface and did not increase with cAMP elevation. The present findings show that low endogenous levels of pig ΔF508-CFTR can mediate substantial anion transport by thyroid epithelia. These data suggest that both wild-type and ΔF508-CFTR regulate additional thyroid transporters, and together co-ordinate the overall I(sc) response.
Collapse
Affiliation(s)
- Yonghai Li
- Department of Anatomy and Physiology, Kansas State University College of Veterinary Medicine, 1600 Denison Avenue, Manhattan, KS 66506, USA
| | | | | |
Collapse
|