1
|
Kim SH, Lee KM, Yoo JY, Chae KJ, Kim GH, Cho JY, The NM, Kim EG, Choi JK, Choe KH, Lee H, Yang B. Mucus plugging, disease severity and sputum myeloperoxidase concentration in bronchiectasis. ERJ Open Res 2025; 11:00279-2024. [PMID: 40391060 PMCID: PMC12086857 DOI: 10.1183/23120541.00279-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/16/2024] [Indexed: 05/21/2025] Open
Abstract
Background Bronchiectasis is characterised by impaired mucociliary clearance, leading to persistent mucus accumulation, known as mucus plugging (MP). Myeloperoxidase (MPO) is a key molecule that activates neutrophilic inflammation during bronchiectasis, leading to disease progression. However, whether sputum MPO concentration is an independent factor associated with MP remains unclear. Methods We retrospectively evaluated 78 patients with bronchiectasis at Chungbuk National University Hospital (Cheongju, Republic of Korea) between June 2022 and April 2023. According to the extent of MP on chest computed tomography, participants were divided into high MP (MP in >9 bronchopulmonary segments) and low MP (MP in ≤9 bronchopulmonary segments) groups. We evaluated whether sputum MPO concentration is independently associated with MP using multivariable adjusted logistic regression analyses. Results There were 59 (75.6%) and 19 (24.4%) participants in the low and high MP groups, respectively. Compared with the low MP group, the high MP group had significantly higher disease severity, as measured by the modified Reiff score (p<0.001) and FACED score (p=0.003). Sputum MPO concentration was significantly correlated with the extent of MP (ρ=0.313, p=0.009). In addition, sputum MPO concentration was independently associated with a high extent of MP even after adjusting for potential confounders (adjusted OR 1.60 (95% CI 1.06-2.42)). Conclusions Sputum MPO concentration was associated with a high degree of MP, suggesting a potential role of MPO in the pathogenesis of mucus plugging.
Collapse
Affiliation(s)
- Sun-Hyung Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
- S-H. Kim and K.M. Lee contributed equally
| | - Ki Man Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
- S-H. Kim and K.M. Lee contributed equally
| | - Jin Young Yoo
- Department of Radiology, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Kum Ju Chae
- Department of Radiology, Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Geun-Hyeong Kim
- The Medical Artificial Intelligence Center, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Jun Yeun Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Nguyen Minh The
- Department of Tuberculosis and Lung Disease, Military Hospital 175, Ho Chi Minh City, Vietnam
| | - Eung-Gook Kim
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Joong-Kook Choi
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Kang Hyeon Choe
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Hyun Lee
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
- H. Lee and B. Yang contributed equally
| | - Bumhee Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
- H. Lee and B. Yang contributed equally
| |
Collapse
|
2
|
Keith JD, Murphree-Terry M, Bollar G, Oden AM, Doty IH, Birket SE. Ivacaftor ameliorates mucus burden, bacterial load, and inflammation in acute but not chronic P. aeruginosa infection in hG551D rats. Respir Res 2024; 25:397. [PMID: 39497082 PMCID: PMC11536857 DOI: 10.1186/s12931-024-03029-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Newly approved highly effective modulation therapies (HEMT) have been life-changing for people with CF. Although these drugs have resulted in significant improvements in lung function and exacerbation rate, bacterial populations in the lung have not been eradicated. The mechanisms behind the continued colonization are not completely clear. METHODS We used a humanized rat to assess the effects of ivacaftor therapy on infection outcomes. Rats harbor an insert expressing humanized CFTR cDNA, including the G551D mutation. hG551D rats were treated with ivacaftor either during or before infection with P. aeruginosa. The response to infection was assessed by bacterial burden in the lung and mucus burden in the lung. RESULTS We found that hG551D rats treated with ivacaftor had reduced bacteria present in the lung in the acute phase of the infection but were not different than vehicle control in the chronic phase of the infection. Similarly, the percentage of neutrophils in the airways were reduced at the acute, but not chronic, timepoints. Overall weight data indicated that the hG551D rats had significantly better weight recovery during the course of infection when treated with ivacaftor. Potentiation of the G551D mutation with ivacaftor resultant in short-circuit current measurements equal to WT, even during the chronic phase of the infection. Despite the persistent infection, hG551D rats treated with ivacaftor had fewer airways with mucus plugs during the chronic infection. CONCLUSIONS The data indicate that the hG551D rats have better outcomes during infection when treated with ivacaftor compared to the vehicle group. Rats have increased weight gain, increased CFTR protein function, and decreased mucus accumulation, despite the persistence of infection and inflammation. These data suggest that ivacaftor improves tolerance of infection, rather than eradication, in this rat model.
Collapse
Affiliation(s)
- Johnathan D Keith
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
| | - Mikayla Murphree-Terry
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gretchen Bollar
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashley M Oden
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ian H Doty
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Susan E Birket
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, USA.
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Cui G, Moustafa DA, Zhao S, Cegla AV, Lyles JT, Goldberg JB, Chandler JD, McCarty NA. Chronic hyperglycemia aggravates lung function in a Scnn1b-Tg murine model. Am J Physiol Lung Cell Mol Physiol 2024; 327:L473-L486. [PMID: 39010826 PMCID: PMC11482466 DOI: 10.1152/ajplung.00279.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 05/02/2024] [Accepted: 06/29/2024] [Indexed: 07/17/2024] Open
Abstract
Cystic fibrosis-related diabetes (CFRD), the most common comorbidity in cystic fibrosis (CF), leads to increased mortality by accelerating the decline in lung function. Scnn1b-Tg transgenic mice overexpressing the epithelial sodium channel β subunit exhibit spontaneous CF-like lung disease, including airway mucus obstruction and chronic inflammation. Here, we established a chronic CFRD-like model using Scnn1b-Tg mice made diabetic by injection of streptozotocin (STZ). In Ussing chamber recordings of the trachea, Scnn1b-Tg mice exhibited larger amiloride-sensitive currents and forskolin-activated currents, without a difference in adenosine triphosphate (ATP)-activated currents compared with wild-type (WT) littermates. Both diabetic WT (WT-D) and diabetic Scnn1b-Tg (Scnn1b-Tg-D) mice on the same genetic background exhibited substantially elevated blood glucose at 8 wk; glucose levels also were elevated in bronchoalveolar lavage fluid (BALF). Bulk lung RNA-seq data showed significant differences between WT-D and Scnn1b-Tg-D mice. Neutrophil counts in BALF were substantially increased in Scnn1b-Tg-D lungs compared with controls (Scnn1b-Tg-con) and compared with WT-D lungs. Lung histology data showed enhanced parenchymal destruction, alveolar wall thickening, and neutrophilic infiltration in Scnn1b-Tg-D mice compared with WT-D mice, consistent with the development of a spontaneous lung infection. We intranasally administered Pseudomonas aeruginosa to induce lung infection in these mice for 24 h, which led to severe lung leukocytic infiltration and an increase in pro-inflammatory cytokine levels in the BALF. In summary, we established a chronic CFRD-like lung mouse model using the Scnn1b-Tg mice. The model can be used for future studies toward understanding the mechanisms underlying the lung pathophysiology associated with CFRD and developing novel therapeutics.NEW & NOTEWORTHY We established a chronic CFRD-like mouse model using the Scnn1b-Tg transgenic mice overexpressing the epithelial sodium channel β subunit made diabetic by injection of streptozotocin. The results underscore the urgent need to develop novel therapeutics for CF lung disease.
Collapse
Affiliation(s)
- Guiying Cui
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Dina A Moustafa
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Analia Vazquez Cegla
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, United States
| | - James T Lyles
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Joanna B Goldberg
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Joshua D Chandler
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, United States
| | - Nael A McCarty
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia, United States
| |
Collapse
|
4
|
Pouwels SD, Ter Haar EAMD, Heijink IH, Hylkema MN, Koster TD, Kuks PJM, Maassen S, Slebos DJ, Vasse GF, de Vries M, Woldhuis RR, Brandsma CA. Highlights from the 11th Bronchitis International Symposium: "Heterogeneity of Lung Disease in a Changing Environment," Groningen, The Netherlands, 2024. Respiration 2024; 103:765-776. [PMID: 39348815 DOI: 10.1159/000541655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024] Open
Abstract
This meeting report provides an overview of the highlights of the Bronchitis XI international symposium, held in June 2024 in Groningen, The Netherlands. The theme of this year's symposium was "heterogeneity of lung disease in a changing environment," and the symposium contained five different sessions focused on (i) heterogeneity of chronic lung disease, (ii) environmental changes with impact on lung disease, (iii) the aging lung, (iv) bronchitis, and (v) innovative therapy. The highlights from each of these sessions will be discussed separately, providing an overview of latest studies, new data, and enthralling discussions.
Collapse
Affiliation(s)
- Simon D Pouwels
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Else A M D Ter Haar
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Machteld N Hylkema
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - T David Koster
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Pauline J M Kuks
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Sjors Maassen
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Dirk-Jan Slebos
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Gwenda F Vasse
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Maaike de Vries
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Epidemiology, University of Groningen, Groningen, The Netherlands
| | - Roy R Woldhuis
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
5
|
Corrao F, Kelly-Aubert M, Sermet-Gaudelus I, Semeraro M. Unmet challenges in cystic fibrosis treatment with modulators. Expert Rev Respir Med 2024; 18:145-157. [PMID: 38755109 DOI: 10.1080/17476348.2024.2357210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION 'Highly effective' modulator therapies (HEMTs) have radically changed the Cystic Fibrosis (CF) therapeutic landscape. AREAS COVERED A comprehensive search strategy was undertaken to assess impact of HEMT in life of pwCF, treatment challenges in specific populations such as very young children, and current knowledge gaps. EXPERT OPINION HEMTs are prescribed for pwCF with definite genotypes. The heterogeneity of variants complicates treatment possibilities and around 10% of pwCF worldwide remains ineligible. Genotype-specific treatments are prompting theratyping and personalized medicine strategies. Improvement in lung function and quality of life increase survival rates, shifting CF from a pediatric to an adult disease. This implies new studies addressing long-term efficacy, side effects, emergence of adult co-morbidities and possible drug-drug interactions. More sensitive and predictive biomarkers for both efficacy and toxicity are warranted. As HEMTs cross the placenta and are found in breast milk, studies addressing the potential consequences of treatment during pregnancy and breastfeeding are urgently needed. Finally, although the treatment and expected outcomes of CF have improved dramatically in high- and middle-income countries, lack of access in low-income countries to these life-changing medicines highlights inequity of care worldwide.
Collapse
Affiliation(s)
- Federica Corrao
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
- INSERM, Institut Necker Enfants Malades, Paris, France
| | | | - Isabelle Sermet-Gaudelus
- INSERM, Institut Necker Enfants Malades, Paris, France
- Centre de Référence Maladies Rares Mucoviscidose et maladies apparentées. Site constitutif, Université de Paris, Paris, France
- European Reference Lung Center, Frankfurt, Germany
- Université Paris Cité, Paris, France
| | - Michaela Semeraro
- Université Paris Cité, Paris, France
- Centre Investigation Clinique, Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
6
|
Gaudin C, Ghinnagow R, Lemaire F, Villeret B, Sermet-Gaudelus I, Sallenave JM. Abnormal functional lymphoid tolerance and enhanced myeloid exocytosis are characteristics of resting and stimulated PBMCs in cystic fibrosis patients. Front Immunol 2024; 15:1360716. [PMID: 38469306 PMCID: PMC10925672 DOI: 10.3389/fimmu.2024.1360716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 01/30/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Cystic Fibrosis (CF) is the commonest genetically inherited disease (1 in 4,500 newborns) and 70% of people with CF (pwCF) harbour the F508Del mutation, resulting in misfolding and incorrect addressing of the channel CFTR to the epithelial membrane and subsequent dysregulation of fluid homeostasis. Although studies have underscored the importance and over-activation of myeloid cells, and in particular neutrophils in the lungs of people with CF (pwCF), relatively less emphasis has been put on the potential immunological bias in CF blood cells, at homeostasis or following stimulation/infection. Methods Here, we revisited, in an exhaustive fashion, in pwCF with mild disease (median age of 15, median % FEV1 predicted = 87), whether their PBMCs, unprimed or primed with a 'non specific' stimulus (PMA+ionomycin mix) and a 'specific' one (live P.a =PAO1 strain), were differentially activated, compared to healthy controls (HC) PBMCs. Results 1) we analysed the lymphocytic and myeloid populations present in CF and Control PBMCs (T cells, NKT, Tgd, ILCs) and their production of the signature cytokines IFN-g, IL-13, IL-17, IL-22. 2) By q-PCR, ELISA and Luminex analysis we showed that CF PBMCs have increased background cytokines and mediators production and a partial functional tolerance phenotype, when restimulated. 3) we showed that CF PBMCs low-density neutrophils release higher levels of granule components (S100A8/A9, lactoferrin, MMP-3, MMP-7, MMP-8, MMP-9, NE), demonstrating enhanced exocytosis of potentially harmful mediators. Discussion In conclusion, we demonstrated that functional lymphoid tolerance and enhanced myeloid protease activity are key features of cystic fibrosis PBMCs.
Collapse
Affiliation(s)
- Clémence Gaudin
- Laboratoire d’Excellence Inflamex, Institut National de la Santé et de la Recherche Medicale, Physiopathologie et Épidémiologie des Maladies Respiratoires, Université Paris-Cité, Paris, France
| | - Reem Ghinnagow
- Laboratoire d’Excellence Inflamex, Institut National de la Santé et de la Recherche Medicale, Physiopathologie et Épidémiologie des Maladies Respiratoires, Université Paris-Cité, Paris, France
| | - Flora Lemaire
- Laboratoire d’Excellence Inflamex, Institut National de la Santé et de la Recherche Medicale, Physiopathologie et Épidémiologie des Maladies Respiratoires, Université Paris-Cité, Paris, France
| | - Bérengère Villeret
- Laboratoire d’Excellence Inflamex, Institut National de la Santé et de la Recherche Medicale, Physiopathologie et Épidémiologie des Maladies Respiratoires, Université Paris-Cité, Paris, France
| | - Isabelle Sermet-Gaudelus
- INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Université Paris-Cité, Paris, France
- ERN-LUNG CF Network, Frankfurt, Germany
- Centre de Ressources et de Compétence de la Mucoviscidose Pédiatrique, Hôpital Mignot, Paris, France
| | - Jean-Michel Sallenave
- Laboratoire d’Excellence Inflamex, Institut National de la Santé et de la Recherche Medicale, Physiopathologie et Épidémiologie des Maladies Respiratoires, Université Paris-Cité, Paris, France
| |
Collapse
|
7
|
De M, Serpa G, Zuiker E, Hisert KB, Liles WC, Manicone AM, Hemann EA, Long ME. MEK1/2 inhibition decreases pro-inflammatory responses in macrophages from people with cystic fibrosis and mitigates severity of illness in experimental murine methicillin-resistant Staphylococcus aureus infection. Front Cell Infect Microbiol 2024; 14:1275940. [PMID: 38352056 PMCID: PMC10861668 DOI: 10.3389/fcimb.2024.1275940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Chronic pulmonary bacterial infections and associated inflammation remain a cause of morbidity and mortality in people with cystic fibrosis (PwCF) despite new modulator therapies. Therapies targeting host factors that dampen detrimental inflammation without suppressing immune responses critical for controlling infections remain limited, while the development of lung infections caused by antimicrobial resistant bacteria is an increasing global problem, and a significant challenge in CF. Pharmacological compounds targeting the mammalian MAPK proteins MEK1 and MEK2, referred to as MEK1/2 inhibitor compounds, have potential combined anti-microbial and anti-inflammatory effects. Here we examined the immunomodulatory properties of MEK1/2 inhibitor compounds PD0325901, trametinib, and CI-1040 on CF innate immune cells. Human CF macrophage and neutrophil phagocytic functions were assessed by quantifying phagocytosis of serum opsonized pHrodo red E. coli, Staphylococcus aureus, and zymosan bioparticles. MEK1/2 inhibitor compounds reduced CF macrophage pro-inflammatory cytokine production without impairing CF macrophage or neutrophil phagocytic abilities. Wild-type C57BL6/J and Cftr tm1kth (F508del homozygous) mice were used to evaluate the in vivo therapeutic potential of PD0325901 compared to vehicle treatment in an intranasal methicillin-resistant Staphylococcus aureus (MRSA) infection with the community-acquired MRSA strain USA300. In both wild-type and CF mice, PD0325901 reduced inflammation associated body mass loss. Wild-type mice treated with PD0325901 had significant reduction in neutrophil-mediated inflammation compared to vehicle treatment groups, with preserved clearance of bacteria in lung, liver, or spleen 1 day after infection in either wild-type or CF mouse models. In summary, this study provides the first data evaluating the therapeutic potential of MEK1/2 inhibitor to modulate CF immune cells and demonstrates that MEK1/2 inhibitors diminish pro-inflammatory responses without impairing host defense mechanisms required for acute pathogen clearance.
Collapse
Affiliation(s)
- Mithu De
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Gregory Serpa
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Eryn Zuiker
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | | | - W. Conrad Liles
- Department of Medicine, Division of Infectious Diseases, University of Washington, Seattle, WA, United States
- Center for Lung Biology, University of Washington, Seattle, WA, United States
| | - Anne M. Manicone
- Center for Lung Biology, University of Washington, Seattle, WA, United States
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, United States
| | - Emily A. Hemann
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Matthew E. Long
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
8
|
Wang S, Niroula S, Hoffman A, Khorrami M, Khorrami M, Yuan F, Gasser GN, Choi S, Liu B, Li J, Metersky ML, Vincent M, Crum CP, Boucher RC, Karmouty-Quintana H, Huang HJ, Sheshadri A, Dickey BF, Parekh KR, Engelhardt JF, McKeon FD, Xian W. Inflammatory Activity of Epithelial Stem Cell Variants from Cystic Fibrosis Lungs Is Not Resolved by CFTR Modulators. Am J Respir Crit Care Med 2023; 208:930-943. [PMID: 37695863 PMCID: PMC10870857 DOI: 10.1164/rccm.202305-0818oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/11/2023] [Indexed: 09/13/2023] Open
Abstract
Rationale: CFTR (cystic fibrosis transmembrane conductance regulator) modulator drugs restore function to mutant channels in patients with cystic fibrosis (CF) and lead to improvements in body mass index and lung function. Although it is anticipated that early childhood treatment with CFTR modulators will significantly delay or even prevent the onset of advanced lung disease, lung neutrophils and inflammatory cytokines remain high in patients with CF with established lung disease despite modulator therapy, underscoring the need to identify and ultimately target the sources of this inflammation in CF lungs. Objectives: To determine whether CF lungs, like chronic obstructive pulmonary disease (COPD) lungs, harbor potentially pathogenic stem cell "variants" distinct from the normal p63/Krt5 lung stem cells devoted to alveolar fates, to identify specific variants that might contribute to the inflammatory state of CF lungs, and to assess the impact of CFTR genetic complementation or CFTR modulators on the inflammatory variants identified herein. Methods: Stem cell cloning technology developed to resolve pathogenic stem cell heterogeneity in COPD and idiopathic pulmonary fibrosis lungs was applied to end-stage lungs of patients with CF (three homozygous CFTR:F508D, one CFTR F508D/L1254X; FEV1, 14-30%) undergoing therapeutic lung transplantation. Single-cell-derived clones corresponding to the six stem cell clusters resolved by single-cell RNA sequencing of these libraries were assessed by RNA sequencing and xenografting to monitor inflammation, fibrosis, and mucin secretion. The impact of CFTR activity on these variants after CFTR gene complementation or exposure to CFTR modulators was assessed by molecular and functional studies. Measurements and Main Results: End-stage CF lungs display a stem cell heterogeneity marked by five predominant variants in addition to the normal lung stem cell, of which three are proinflammatory both at the level of gene expression and their ability to drive neutrophilic inflammation in xenografts in immunodeficient mice. The proinflammatory functions of these three variants were unallayed by genetic or pharmacological restoration of CFTR activity. Conclusions: The emergence of three proinflammatory stem cell variants in CF lungs may contribute to the persistence of lung inflammation in patients with CF with advanced disease undergoing CFTR modulator therapy.
Collapse
Affiliation(s)
- Shan Wang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Suchan Niroula
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Ashley Hoffman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Melika Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Melina Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Feng Yuan
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Grace N. Gasser
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Soon Choi
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Bovey Liu
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | - Mark L. Metersky
- Center for Bronchiectasis Care, Pulmonary, Critical Care, and Sleep Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | | | - Christopher P. Crum
- Women’s and Perinatal Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard C. Boucher
- Cystic Fibrosis and Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Howard J. Huang
- Department of Medicine, Houston Methodist Hospital, Houston, Texas; and
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Burton F. Dickey
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kalpaj R. Parekh
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
- Division of Thoracic Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology
- Gene Therapy Center for Cystic Fibrosis and Other Genetic Diseases, and
| | - Frank D. McKeon
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Wa Xian
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| |
Collapse
|
9
|
Grandy S, Scur M, Dolan K, Nickerson R, Cheng Z. Using model systems to unravel host-Pseudomonas aeruginosa interactions. Environ Microbiol 2023; 25:1765-1784. [PMID: 37290773 DOI: 10.1111/1462-2920.16440] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/29/2023] [Indexed: 06/10/2023]
Abstract
Using model systems in infection biology has led to the discoveries of many pathogen-encoded virulence factors and critical host immune factors to fight pathogenic infections. Studies of the remarkable Pseudomonas aeruginosa bacterium that infects and causes disease in hosts as divergent as humans and plants afford unique opportunities to shed new light on virulence strategies and host defence mechanisms. One of the rationales for using model systems as a discovery tool to characterise bacterial factors driving human infection outcomes is that many P. aeruginosa virulence factors are required for pathogenesis in diverse different hosts. On the other side, many host signalling components, such as the evolutionarily conserved mitogen-activated protein kinases, are involved in immune signalling in a diverse range of hosts. Some model organisms that have less complex immune systems also allow dissection of the direct impacts of innate immunity on host defence without the interference of adaptive immunity. In this review, we start with discussing the occurrence of P. aeruginosa in the environment and the ability of this bacterium to cause disease in various hosts as a natural opportunistic pathogen. We then summarise the use of some model systems to study host defence and P. aeruginosa virulence.
Collapse
Affiliation(s)
- Shannen Grandy
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michal Scur
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kathleen Dolan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Rhea Nickerson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
10
|
Hisert KB, Birket SE, Clancy JP, Downey DG, Engelhardt JF, Fajac I, Gray RD, Lachowicz-Scroggins ME, Mayer-Hamblett N, Thibodeau P, Tuggle KL, Wainwright CE, De Boeck K. Understanding and addressing the needs of people with cystic fibrosis in the era of CFTR modulator therapy. THE LANCET. RESPIRATORY MEDICINE 2023; 11:916-931. [PMID: 37699420 DOI: 10.1016/s2213-2600(23)00324-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/07/2023] [Accepted: 08/20/2023] [Indexed: 09/14/2023]
Abstract
Cystic fibrosis is a multiorgan disease caused by impaired function of the cystic fibrosis transmembrane conductance regulator (CFTR). Since the introduction of the CFTR modulator combination elexacaftor-tezacaftor-ivacaftor (ETI), which acts directly on mutant CFTR to enhance its activity, most people with cystic fibrosis (pwCF) have seen pronounced reductions in symptoms, and studies project marked increases in life expectancy for pwCF who are eligible for ETI. However, modulator therapy has not cured cystic fibrosis and the success of CFTR modulators has resulted in immediate questions about the new state of cystic fibrosis disease and clinical challenges in the care of pwCF. In this Series paper, we summarise key questions about cystic fibrosis disease in the era of modulator therapy, highlighting state-of-the-art research and clinical practices, knowledge gaps, new challenges faced by pwCF and the potential for future health-care challenges, and the pressing need for additional therapies to treat the underlying genetic or molecular causes of cystic fibrosis.
Collapse
Affiliation(s)
| | - Susan E Birket
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Damian G Downey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Isabelle Fajac
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Robert D Gray
- Institution of Regeneration and Repair, Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | | | - Nicole Mayer-Hamblett
- Department of Pediatrics, Department of Biostatistics, Seattle Children's Research Institute, University of Washington, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
11
|
Graeber SY, Mall MA. The future of cystic fibrosis treatment: from disease mechanisms to novel therapeutic approaches. Lancet 2023; 402:1185-1198. [PMID: 37699417 DOI: 10.1016/s0140-6736(23)01608-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
With the 2019 breakthrough in the development of highly effective modulator therapy providing unprecedented clinical benefits for over 90% of patients with cystic fibrosis who are genetically eligible for treatment, this rare disease has become a front runner of transformative molecular therapy. This success is based on fundamental research, which led to the identification of the disease-causing CFTR gene and our subsequent understanding of the disease mechanisms underlying the pathogenesis of cystic fibrosis, working together with a continuously evolving clinical research and drug development pipeline. In this Series paper, we focus on advances since 2018, and remaining knowledge gaps in our understanding of the molecular mechanisms of CFTR dysfunction in the airway epithelium and their links to mucus dysfunction, impaired host defences, airway infection, and chronic inflammation of the lungs of people with cystic fibrosis. We review progress in (and the remaining obstacles to) pharmacological approaches to rescue CFTR function, and novel strategies for improved symptomatic therapies for cystic fibrosis, including how these might be applicable to common lung diseases, such as bronchiectasis and chronic obstructive pulmonary disease. Finally, we discuss the promise of genetic therapies and gene editing approaches to restore CFTR function in the lungs of all patients with cystic fibrosis independent of their CFTR genotype, and the unprecedented opportunities to transform cystic fibrosis from a fatal disease to a treatable and potentially curable one.
Collapse
Affiliation(s)
- Simon Y Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Cystic Fibrosis Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research, associated partner site, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Cystic Fibrosis Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research, associated partner site, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Meyerholz DK, Leidinger MR, Adam Goeken J, Businga TR, Vizuett S, Akers A, Evans I, Zhang Y, Engelhardt JF. Immunohistochemical detection of MUC5AC and MUC5B mucins in ferrets. BMC Res Notes 2023; 16:111. [PMID: 37349833 PMCID: PMC10286488 DOI: 10.1186/s13104-023-06388-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVE Cystic fibrosis (CF) is a genetic condition that causes abnormal mucus secretions in affected organs. MUC5AC and MUC5B are gel-forming mucins and frequent targets for investigations in CF tissues. Our objective was to qualify MUC5AC and MUC5B immunohistochemical techniques to provide a useful tool to identify, localize and interpret mucin expression in ferret tissues. RESULTS MUC5AC and MUC5B mucins were detected most commonly in large airways and least in small airways, consistent with reported goblet cell density in airway surface epithelia. We evaluated whether staining method affected the detection of goblet cell mucins in serial sections of bronchial surface epithelia. Significant differences between stains were not observed suggesting common co-expression MUC5AC and MUC5B proteins in goblet cells of airway surface epithelia. Gallbladder and stomach tissues are reported to have differential mucin enrichment, so we tested these tissues in wildtype ferrets. Stomach tissues were enriched in MUC5AC and gallbladder tissues enriched in MUC5B, mucin enrichment similar to human tissues. Mucin immunostaining techniques were further qualified for specificity using lung tissue from recently generated MUC5AC-/- and MUC5B-/- ferrets. Qualified techniques for MUC5AC and MUC5B immunohistochemistry will be useful tools for mucin tissue studies in CF and other ferret models.
Collapse
Affiliation(s)
- David K. Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Mariah R. Leidinger
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - J. Adam Goeken
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Thomas R. Businga
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Sebastian Vizuett
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Allison Akers
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Idil Evans
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Yan Zhang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| |
Collapse
|
13
|
Tang Y, Fakhari S, Huntemann ED, Feng Z, Wu P, Feng WY, Lei J, Yuan F, Excoffon KJ, Wang K, Limberis MP, Kolbeck R, Yan Z, Engelhardt JF. Immunosuppression reduces rAAV2.5T neutralizing antibodies that limit efficacy following repeat dosing to ferret lungs. Mol Ther Methods Clin Dev 2023; 29:70-80. [PMID: 36950451 PMCID: PMC10025970 DOI: 10.1016/j.omtm.2023.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
The efficacy of redosing the recombinant adeno-associated virus (rAAV) vector rAAV2.5T to ferret lung is limited by AAV neutralizing antibody (NAb) responses. While immunosuppression strategies have allowed for systemic rAAV repeat dosing, their utility for rAAV lung-directed gene therapy is largely unexplored. To this end, we evaluated two immunosuppression (IS) strategies to improve repeat dosing of rAAV2.5T to ferret lungs: (1) a combination of three IS drugs (Tri-IS) with broad coverage against cellular and humoral responses (methylprednisolone [MP], azathioprine, and cyclosporine) and (2) MP alone, which is typically used in systemic rAAV applications. Repeat dosing utilized AAV2.5T-SP183-fCFTRΔR (recombinant ferret CFTR transgene), followed 28 days later by AAV2.5T-SP183-gLuc (for quantification of transgene expression). Both the Tri-IS and MP strategies significantly improved transgene expression following repeat dosing and reduced AAV2.5T NAb responses in the bronchioalveolar lavage fluid (BALF) and plasma, while AAV2.5T binding antibody subtypes and cellular immune responses by ELISpot were largely unchanged by IS. One exception was the reduction in plasma AAV2.5T binding immunoglobulin G (IgG) in both IS groups. Only the Tri-IS strategy significantly suppressed splenocyte expression of IFNA (interferon α [IFN-α]) and IL4. Our studies suggest that IS strategies may be useful in clinical application of rAAV targeting lung genetic diseases such as cystic fibrosis.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shahab Fakhari
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Eric D. Huntemann
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zehua Feng
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Peipei Wu
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - William Y. Feng
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Junying Lei
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Feng Yuan
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Kai Wang
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | | | | | - Ziying Yan
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - John F. Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
14
|
Mikami Y, Grubb BR, Rogers TD, Dang H, Asakura T, Kota P, Gilmore RC, Okuda K, Morton LC, Sun L, Chen G, Wykoff JA, Ehre C, Vilar J, van Heusden C, Livraghi-Butrico A, Gentzsch M, Button B, Stutts MJ, Randell SH, O’Neal WK, Boucher RC. Chronic airway epithelial hypoxia exacerbates injury in muco-obstructive lung disease through mucus hyperconcentration. Sci Transl Med 2023; 15:eabo7728. [PMID: 37285404 PMCID: PMC10664029 DOI: 10.1126/scitranslmed.abo7728] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/17/2023] [Indexed: 06/09/2023]
Abstract
Unlike solid organs, human airway epithelia derive their oxygen from inspired air rather than the vasculature. Many pulmonary diseases are associated with intraluminal airway obstruction caused by aspirated foreign bodies, virus infection, tumors, or mucus plugs intrinsic to airway disease, including cystic fibrosis (CF). Consistent with requirements for luminal O2, airway epithelia surrounding mucus plugs in chronic obstructive pulmonary disease (COPD) lungs are hypoxic. Despite these observations, the effects of chronic hypoxia (CH) on airway epithelial host defense functions relevant to pulmonary disease have not been investigated. Molecular characterization of resected human lungs from individuals with a spectrum of muco-obstructive lung diseases (MOLDs) or COVID-19 identified molecular features of chronic hypoxia, including increased EGLN3 expression, in epithelia lining mucus-obstructed airways. In vitro experiments using cultured chronically hypoxic airway epithelia revealed conversion to a glycolytic metabolic state with maintenance of cellular architecture. Chronically hypoxic airway epithelia unexpectedly exhibited increased MUC5B mucin production and increased transepithelial Na+ and fluid absorption mediated by HIF1α/HIF2α-dependent up-regulation of β and γENaC (epithelial Na+ channel) subunit expression. The combination of increased Na+ absorption and MUC5B production generated hyperconcentrated mucus predicted to perpetuate obstruction. Single-cell and bulk RNA sequencing analyses of chronically hypoxic cultured airway epithelia revealed transcriptional changes involved in airway wall remodeling, destruction, and angiogenesis. These results were confirmed by RNA-in situ hybridization studies of lungs from individuals with MOLD. Our data suggest that chronic airway epithelial hypoxia may be central to the pathogenesis of persistent mucus accumulation in MOLDs and associated airway wall damage.
Collapse
Affiliation(s)
- Yu Mikami
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Barbara R. Grubb
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Troy D. Rogers
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Takanori Asakura
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Pradeep Kota
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rodney C. Gilmore
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lisa C. Morton
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ling Sun
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Gang Chen
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jason A. Wykoff
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Camille Ehre
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Juan Vilar
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Catharina van Heusden
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Martina Gentzsch
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brian Button
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - M. Jackson Stutts
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Scott H. Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
15
|
Cook DP, Thomas CM, Wu AY, Rusznak M, Zhang J, Zhou W, Cephus JY, Gibson-Corley KN, Polosukhin VV, Norlander AE, Newcomb DC, Stoltz DA, Peebles RS. Cystic Fibrosis Reprograms Airway Epithelial IL-33 Release and Licenses IL-33-Dependent Inflammation. Am J Respir Crit Care Med 2023; 207:1486-1497. [PMID: 36952660 PMCID: PMC10263140 DOI: 10.1164/rccm.202211-2096oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/23/2023] [Indexed: 03/25/2023] Open
Abstract
Rationale: Type 2 inflammation has been described in people with cystic fibrosis (CF). Whether loss of CFTR (cystic fibrosis transmembrane conductance regulator) function contributes directly to a type 2 inflammatory response has not been fully defined. Objectives: The potent alarmin IL-33 has emerged as a critical regulator of type 2 inflammation. We tested the hypothesis that CFTR deficiency increases IL-33 expression and/or release and deletion of IL-33 reduces allergen-induced inflammation in the CF lung. Methods: Human airway epithelial cells (AECs) grown from non-CF and CF cell lines and Cftr+/+ and Cftr-/- mice were used in this study. Pulmonary inflammation in Cftr+/+ and Cftr-/- mice with and without IL-33 or ST2 (IL-1 receptor-like 1) germline deletion was determined by histological analysis, BAL, and cytokine analysis. Measurements and Main Results: After allergen challenge, both CF human AECs and Cftr-/- mice had increased IL-33 expression compared with control AECs and Cftr+/+ mice, respectively. DUOX1 (dual oxidase 1) expression was increased in CF human AECs and Cftr-/- mouse lungs compared with control AECs and lungs from Cftr+/+ mice and was necessary for the increased IL-33 release in Cftr-/- mice compared with Cftr+/+ mice. IL-33 stimulation of Cftr-/- CD4+ T cells resulted in increased type 2 cytokine production compared with Cftr+/+ CD4+ T cells. Deletion of IL-33 or ST2 decreased both type 2 inflammation and neutrophil recruitment in Cftr-/- mice compared with Cftr+/+ mice. Conclusions: Absence of CFTR reprograms airway epithelial IL-33 release and licenses IL-33-dependent inflammation. Modulation of the IL-33/ST2 axis represents a novel therapeutic target in CF type 2-high and neutrophilic inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Dawn C. Newcomb
- Department of Internal Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David A. Stoltz
- Department of Internal Medicine and
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; and
| | - R. Stokes Peebles
- Department of Internal Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
16
|
Kerschner JL, Paranjapye A, Schacht M, Meckler F, Huang F, Bebek G, Van Wettere AJ, Regouski M, Perisse IV, White KL, Polejaeva IA, Leir SH, Harris A. Transcriptomic analysis of lung development in wildtype and CFTR -/- sheep suggests an early inflammatory signature in the CF distal lung. Funct Integr Genomics 2023; 23:135. [PMID: 37085733 PMCID: PMC10121546 DOI: 10.1007/s10142-023-01050-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/22/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023]
Abstract
The precise molecular events initiating human lung disease are often poorly characterized. Investigating prenatal events that may underlie lung disease in later life is challenging in man, but insights from the well-characterized sheep model of lung development are valuable. Here, we determine the transcriptomic signature of lung development in wild-type sheep (WT) and use a sheep model of cystic fibrosis (CF) to characterize disease associated changes in gene expression through the pseudoglandular, canalicular, saccular, and alveolar stages of lung growth and differentiation. Using gene ontology process enrichment analysis of differentially expressed genes at each developmental time point, we define changes in biological processes (BP) in proximal and distal lung from WT or CF animals. We also compare divergent BP in WT and CF animals at each time point. Next, we establish the developmental profile of key genes encoding components of ion transport and innate immunity that are pivotal in CF lung disease and validate transcriptomic data by RT-qPCR. Consistent with the known pro-inflammatory phenotype of the CF lung after birth, we observe upregulation of inflammatory response processes in the CF sheep distal lung during the saccular stage of prenatal development. These data suggest early commencement of therapeutic regimens may be beneficial.
Collapse
Affiliation(s)
- Jenny L Kerschner
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Makayla Schacht
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Frederick Meckler
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Felix Huang
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Gurkan Bebek
- Center for Proteomics and Bioinformatics, Cleveland, OH, USA
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Arnaud J Van Wettere
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UH, USA
| | - Misha Regouski
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UH, USA
| | - Iuri Viotti Perisse
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UH, USA
| | - Kenneth L White
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UH, USA
| | - Irina A Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UH, USA
| | - Shih-Hsing Leir
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
17
|
Rodriguez-Piñeiro AM, Jaudas F, Klymiuk N, Bähr A, Hansson GC, Ermund A. Proteome of airway surface liquid and mucus in newborn wildtype and cystic fibrosis piglets. Respir Res 2023; 24:83. [PMID: 36927357 PMCID: PMC10022022 DOI: 10.1186/s12931-023-02381-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/04/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND The respiratory tract is protected from inhaled particles and microbes by mucociliary clearance, mediated by the mucus and the cilia creating a flow to move the mucus cephalad. Submucosal glands secrete linear MUC5B mucin polymers and because they pass through the gland duct before reaching the airway surface, bundled strands of 1000-5000 parallel molecules exit the glands. In contrast, the surface goblet cells secrete both MUC5AC and MUC5B. METHODS We used mass-spectrometry based proteomic analysis of unstimulated and carbachol stimulated newborn wild-type (WT) and cystic fibrosis transmembrane conductance regulator (CFTR) null (CF) piglet airways to study proteins in the airway surface liquid and mucus, to investigate if levels of MUC5AC and MUC5B were affected by carbachol stimulation and whether the proteins clustered according to function. RESULTS Proteins in the first four extracted fractions clustered together and the fifth fraction contained the mucus cluster, mucins and other proteins known to associate with mucins, whereas the traditional airway surface liquid proteins clustered to fraction 1-4 and were absent from the mucus fraction. Carbachol stimulation resulted in increased MUC5AC and MUC5B. CONCLUSIONS These results indicate a distinct separation between proteins in the washable surface liquid and the mucus fraction. In fractions 1-4 from newborn CF piglets an additional cluster containing acute phase proteins was observed, suggesting an early inflammatory response in CF piglets. Alternatively, increased levels of these proteins could indicate altered lung development in the CF piglets. This observation suggests that CF airway disease is present at birth and thus, treatment should commence directly after diagnosis.
Collapse
Affiliation(s)
- Ana M Rodriguez-Piñeiro
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Florian Jaudas
- Center for Innovative Animal Models, Ludwig-Maximilians-University, Munich, Germany
| | - Nikolai Klymiuk
- Center for Innovative Animal Models, Ludwig-Maximilians-University, Munich, Germany
| | - Andrea Bähr
- Center for Innovative Animal Models, Ludwig-Maximilians-University, Munich, Germany
| | - Gunnar C Hansson
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Ermund
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
18
|
O’Connor JB, Wagner BD, Harris JK, Frank DN, Clabots DE, Laguna TA. Detection and identification of fungi in the lower airway of children with and without cystic fibrosis. Front Microbiol 2023; 14:1119703. [PMID: 36846802 PMCID: PMC9948248 DOI: 10.3389/fmicb.2023.1119703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Airway infection and inflammation lead to the progression of obstructive lung disease in persons with cystic fibrosis (PWCF). However, cystic fibrosis (CF) fungal communities, known drivers of CF pathophysiology, remain poorly understood due to the shortcomings of traditional fungal culture. Our objective was to apply a novel small subunit rRNA gene (SSU-rRNA) sequencing approach to characterize the lower airway mycobiome in children with and without CF. Methods Bronchoalveolar lavage fluid (BALF) samples and relevant clinical data were collected from pediatric PWCF and disease control (DC) subjects. Total fungal load (TFL) was measured using quantitative PCR, and SSU-rRNA sequencing was used for mycobiome characterization. Results were compared across groups, and Morisita-Horn clustering was performed. Results 161 (84%) of the BALF samples collected had sufficient load for SSU-rRNA sequencing, with amplification being more common in PWCF. BALF from PWCF had increased TFL and increased neutrophilic inflammation compared to DC subjects. PWCF exhibited increased abundance of Aspergillus and Candida, while Malassezia, Cladosporium, and Pleosporales were prevalent in both groups. CF and DC samples showed no clear differences in clustering when compared to each other or to negative controls. SSU-rRNA sequencing was used to profile the mycobiome in pediatric PWCF and DC subjects. Notable differences were observed between the groups, including the abundance of Aspergillus and Candida. Discussion Fungal DNA detected in the airway could represent a combination of pathogenic fungi and environmental exposure (e.g., dust) to fungus indicative of a common background signature. Next steps will require comparisons to airway bacterial communities.
Collapse
Affiliation(s)
- John B. O’Connor
- Division of Pulmonary and Sleep Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States,*Correspondence: John B. O’Connor, ✉
| | - Brandie D. Wagner
- University of Colorado School of Medicine, Aurora, CO, United States,Colorado School of Public Health, University of Colorado Denver, Aurora, CO, United States
| | - J. Kirk Harris
- University of Colorado School of Medicine, Aurora, CO, United States
| | - Daniel N. Frank
- University of Colorado School of Medicine, Aurora, CO, United States
| | - Diana E. Clabots
- University of Colorado School of Medicine, Aurora, CO, United States,Department of Internal Medicine, Palmetto General Hospital, Hialeah, FL, United States
| | - Theresa A. Laguna
- Division of Pulmonary and Sleep Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
19
|
De M, Hisert KB, Liles WC, Manicone AM, Hemann EA, Long ME. MEK1/2 inhibition decreases pro-inflammatory responses in macrophages from people with cystic fibrosis and mitigates severity of illness in experimental murine methicillin-resistant Staphylococcus aureus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525092. [PMID: 36712028 PMCID: PMC9882267 DOI: 10.1101/2023.01.22.525092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Chronic pulmonary bacterial infections and associated inflammation remain a cause of morbidity and mortality in people with cystic fibrosis (PwCF) despite new modulator therapies. Therapies targeting host factors that dampen detrimental inflammation without suppressing immune responses critical for controlling infections remain limited, while the acquisition of antibiotic resistance bacterial infections is an increasing global problem, and a significant challenge in CF. Pharmacological compounds targeting the mammalian MAPK proteins MEK1 and MEK2, referred to as MEK1/2 inhibitor compounds, have potential combined anti-microbial and anti-inflammatory effects. Here we examined the immunomodulatory properties of MEK1/2 inhibitor compounds PD0325901, trametinib, and CI-1040 on CF innate immune cells. Human CF macrophage and neutrophil phagocytic functions were assessed by quantifying phagocytosis of serum opsonized pHrodo red E. coli , Staphylococcus aureus , and zymosan bioparticles. MEK1/2 inhibitor compounds reduced CF macrophage pro-inflammatory cytokine production without impairing CF macrophage or neutrophil phagocytic abilities. Wild-type C57BL6/J and Cftr tm1kth (F508del homozygous) mice were used to evaluate the in vivo therapeutic potential of PD0325901 compared to vehicle treatment in an intranasal methicillin-resistant Staphylococcus aureus (MRSA) infection with the community-acquired MRSA strain USA300. In both wild-type and CF mice, PD0325901 reduced infection related weight loss compared to vehicle treatment groups but did not impair clearance of bacteria in lung, liver, or spleen 1 day after infection. In summary, this study provides the first data evaluating the therapeutic potential of MEK1/2 inhibitor to modulate CF immune cells, and demonstrates that MEK1/2 inhibitors dampen pro-inflammatory responses without impairing host defense mechanisms mediating pathogen clearance.
Collapse
|
20
|
Sagel SD, Kupfer O, Wagner BD, Davis SD, Dell SD, Ferkol TW, Hoppe JE, Rosenfeld M, Sullivan KM, Tiddens HAWM, Knowles MR, Leigh MW. Airway Inflammation in Children with Primary Ciliary Dyskinesia. Ann Am Thorac Soc 2023; 20:67-74. [PMID: 35984413 PMCID: PMC9819265 DOI: 10.1513/annalsats.202204-314oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
Rationale: The role of airway inflammation in disease pathogenesis in children with primary ciliary dyskinesia (PCD) is poorly understood. Objectives: We investigated relationships between sputum inflammation measurements, age, lung function, bronchiectasis, airway infection, and ultrastructural defects in children with PCD. Methods: Spontaneously expectorated sputum was collected from clinically stable children and adolescents with PCD ages 6 years and older participating in a multicenter, observational study. Sputum protease and inflammatory cytokine concentrations were correlated with age, lung function, and chest computed tomography measures of structural lung disease, whereas differences in concentrations were compared between ultrastructural defect categories and between those with and without detectable bacterial infection. Results: Sputum from 77 children with PCD (39 females [51%]; mean [standard deviation] age, 13.9 [4.9] yr; mean [standard deviation] forced expiratory volume in 1 second [FEV1]% predicted, 80.8 [20.5]) was analyzed. Sputum inflammatory marker measurements, including neutrophil elastase activity, IL-1β (interleukin-1β), IL-8, and TNF-α (tumor necrosis factor α) concentrations, correlated positively with age, percentage of bronchiectasis, and percentage of total structural lung disease on computed tomography, and negatively with lung function. Correlations between neutrophil elastase concentrations and FEV1% predicted and percentage of bronchiectasis were -0.32 (95% confidence interval, -0.51 to -0.10) and 0.46 (0.14 to 0.69), respectively. Sputum neutrophil elastase, IL-1β, and TNF-α concentrations were higher in those with detectable bacterial pathogens. Participants with absent inner dynein arm and microtubular disorganization had similar inflammatory profiles compared with participants with outer dynein arm defects. Conclusions: In this multicenter pediatric PCD cohort, elevated concentrations of sputum proteases and cytokines were associated with impaired lung function and structural damage as determined by chest computed tomography, suggesting that sputum inflammatory measurements could serve as biomarkers in PCD.
Collapse
Affiliation(s)
- Scott D. Sagel
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Oren Kupfer
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brandie D. Wagner
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| | | | - Sharon D. Dell
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Thomas W. Ferkol
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Jordana E. Hoppe
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Margaret Rosenfeld
- Department of Pediatrics, Children’s Hospital and Regional Medical Center, Seattle, Washington; and
| | - Kelli M. Sullivan
- Department of Medicine, Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Harm A. W. M. Tiddens
- Department of Pediatric Pulmonology and Allergology, Erasmus MC‐Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Michael R. Knowles
- Department of Medicine, Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | | |
Collapse
|
21
|
A phase I study assessing the safety and tolerability of allogeneic mesenchymal stem cell infusion in adults with cystic fibrosis. J Cyst Fibros 2022:S1569-1993(22)01421-7. [PMID: 36549988 DOI: 10.1016/j.jcf.2022.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Mesenchymal stem cells are of particular interest in cystic fibrosis (CF) as a potential therapeutic. Data from pre-clinical studies suggest that allogeneic bone marrow-derived human mesenchymal stem cells (hMSCs) may provide a new therapeutic treatment for CF lung disease by attenuating pulmonary inflammation while decreasing bacterial growth and enhancing antibiotic efficacy. METHODS Fifteen adults with CF were enrolled in a phase 1 dose-escalation trial of a single intravenous infusion of hMSCs derived from bone marrow aspirates obtained from a single pre-clinically validated healthy volunteer donor. The study employed a 3+3 dose escalation design with subjects receiving a single, intravenous dose of either 1×106, 3×106, or 5×106 hMSCs/kg. Subjects were monitored inpatient for 24 hours and by outpatient visits and telephone calls for 12 months after the infusion. Safety and tolerability were evaluated by monitoring symptoms, patient reported outcome questionnaires, adverse events (AEs), physical exam findings, spirometry, and analyses of safety laboratories. Preliminary evidence for potential efficacy using inflammatory markers in the blood and sputum were also evaluated. RESULTS No dose-limiting toxicities, deaths or life-threatening adverse events were observed. Most AEs and serious adverse events (SAEs) were consistent with underlying CF. Vital signs, physical exam findings, spirometry and safety laboratory results showed no significant change from baseline. No trends over time were seen in serum or sputum inflammatory markers nor with clinical spirometry. CONCLUSION Allogeneic hMSC intravenous infusions were safe and well-tolerated in this phase 1 study and warrant additional clinical testing as a potential therapeutic for CF lung disease.
Collapse
|
22
|
Öz HH, Cheng EC, Di Pietro C, Tebaldi T, Biancon G, Zeiss C, Zhang PX, Huang PH, Esquibies SS, Britto CJ, Schupp JC, Murray TS, Halene S, Krause DS, Egan ME, Bruscia EM. Recruited monocytes/macrophages drive pulmonary neutrophilic inflammation and irreversible lung tissue remodeling in cystic fibrosis. Cell Rep 2022; 41:111797. [PMID: 36516754 PMCID: PMC9833830 DOI: 10.1016/j.celrep.2022.111797] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 09/30/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022] Open
Abstract
Persistent neutrophil-dominated lung inflammation contributes to lung damage in cystic fibrosis (CF). However, the mechanisms that drive persistent lung neutrophilia and tissue deterioration in CF are not well characterized. Starting from the observation that, in patients with CF, c-c motif chemokine receptor 2 (CCR2)+ monocytes/macrophages are abundant in the lungs, we investigate the interplay between monocytes/macrophages and neutrophils in perpetuating lung tissue damage in CF. Here we show that CCR2+ monocytes in murine CF lungs drive pathogenic transforming growth factor β (TGF-β) signaling and sustain a pro-inflammatory environment by facilitating neutrophil recruitment. Targeting CCR2 to lower the numbers of monocytes in CF lungs ameliorates neutrophil inflammation and pathogenic TGF-β signaling and prevents lung tissue damage. This study identifies CCR2+ monocytes as a neglected contributor to the pathogenesis of CF lung disease and as a therapeutic target for patients with CF, for whom lung hyperinflammation and tissue damage remain an issue despite recent advances in CF transmembrane conductance regulator (CFTR)-specific therapeutic agents.
Collapse
Affiliation(s)
- Hasan H Öz
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Ee-Chun Cheng
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | | | - Toma Tebaldi
- Department of Hematology, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA; Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Giulia Biancon
- Department of Hematology, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Caroline Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ping-Xia Zhang
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Pamela H Huang
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Sofia S Esquibies
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Clemente J Britto
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jonas C Schupp
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), Hannover, Germany
| | - Thomas S Murray
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Department of Hematology, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Diane S Krause
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Marie E Egan
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Emanuela M Bruscia
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
23
|
Caverly LJ, Riquelme SA, Hisert KB. The Impact of Highly Effective Modulator Therapy on Cystic Fibrosis Microbiology and Inflammation. Clin Chest Med 2022; 43:647-665. [PMID: 36344072 PMCID: PMC10224747 DOI: 10.1016/j.ccm.2022.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Highly effective cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulator therapy (HEMT) corrects the underlying molecular defect causing CF disease. HEMT decreases symptom burden and improves clinical metrics and quality of life for most people with CF (PwCF) and eligible cftr mutations. Improvements in measures of pulmonary health suggest that restoration of function of defective CFTR anion channels by HEMT not only enhances airway mucociliary clearance, but also reduces chronic pulmonary infection and inflammation. This article reviews the evidence for how HEMT influences the dynamic and interdependent processes of infection and inflammation in the CF airway, and what questions remain unanswered.
Collapse
Affiliation(s)
- Lindsay J Caverly
- Department of Pediatrics, University of Michigan Medical School, L2221 UH South, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5212, USA
| | - Sebastián A Riquelme
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, Columbia University Medical Center, 650West 168th Street, New York, NY 10032, USA
| | - Katherine B Hisert
- Department of Medicine, National Jewish Health, Smith A550, 1400 Jackson Street, Denver, CO 80205, USA.
| |
Collapse
|
24
|
Hill DB, Button B, Rubinstein M, Boucher RC. Physiology and pathophysiology of human airway mucus. Physiol Rev 2022; 102:1757-1836. [PMID: 35001665 PMCID: PMC9665957 DOI: 10.1152/physrev.00004.2021] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 01/27/2023] Open
Abstract
The mucus clearance system is the dominant mechanical host defense system of the human lung. Mucus is cleared from the lung by cilia and airflow, including both two-phase gas-liquid pumping and cough-dependent mechanisms, and mucus transport rates are heavily dependent on mucus concentration. Importantly, mucus transport rates are accurately predicted by the gel-on-brush model of the mucociliary apparatus from the relative osmotic moduli of the mucus and periciliary-glycocalyceal (PCL-G) layers. The fluid available to hydrate mucus is generated by transepithelial fluid transport. Feedback interactions between mucus concentrations and cilia beating, via purinergic signaling, coordinate Na+ absorptive vs Cl- secretory rates to maintain mucus hydration in health. In disease, mucus becomes hyperconcentrated (dehydrated). Multiple mechanisms derange the ion transport pathways that normally hydrate mucus in muco-obstructive lung diseases, e.g., cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), non-CF bronchiectasis (NCFB), and primary ciliary dyskinesia (PCD). A key step in muco-obstructive disease pathogenesis is the osmotic compression of the mucus layer onto the airway surface with the formation of adherent mucus plaques and plugs, particularly in distal airways. Mucus plaques create locally hypoxic conditions and produce airflow obstruction, inflammation, infection, and, ultimately, airway wall damage. Therapies to clear adherent mucus with hydrating and mucolytic agents are rational, and strategies to develop these agents are reviewed.
Collapse
Affiliation(s)
- David B Hill
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina
| | - Brian Button
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael Rubinstein
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Mechanical Engineering and Materials Science, Biomedical Engineering, Physics, and Chemistry, Duke University, Durham, North Carolina
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
25
|
Ash JJ, Hilkin BM, Gansemer ND, Hoffman EA, Zabner J, Stoltz DA, Abou Alaiwa MH. Tromethamine improves mucociliary clearance in cystic fibrosis pigs. Physiol Rep 2022; 10:e15340. [PMID: 36073059 PMCID: PMC9453173 DOI: 10.14814/phy2.15340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023] Open
Abstract
In cystic fibrosis (CF), the loss of cystic fibrosis transmembrane conductance regulator (CFTR) mediated Cl- and HCO3 - secretion across the epithelium acidifies the airway surface liquid (ASL). Acidic ASL alters two key host defense mechanisms: Rapid ASL bacterial killing and mucociliary transport (MCT). Aerosolized tromethamine (Tham) increases ASL pH and restores the ability of ASL to rapidly kill bacteria in CF pigs. In CF pigs, clearance of insufflated microdisks is interrupted due to abnormal mucus causing microdisks to abruptly recoil. Aerosolizing a reducing agent to break disulfide bonds that link mucins improves MCT. Here, we are interested in restoring MCT in CF by aerosolizing Tham, a buffer with a pH of 8.4. Because Tham is hypertonic to serum, we use an acidified formulation as a control. We measure MCT by tracking the caudal movement of individual tantalum microdisks with serial chest computed tomography scans. Alkaline Tham improves microdisk clearance to within the range of that seen in non-CF pigs. It also partially reverses MCT defects, including reduced microdisk recoil and elapse time until they start moving after methacholine stimulation in CF pig airways. The effect is not due to hypertonicity, as it is not seen with acidified Tham or hypertonic saline. This finding indicates acidic ASL impairs CF MCT and suggests that alkalinization of ASL pH with inhaled Tham may improve CF airway disease.
Collapse
Affiliation(s)
- Jamison J. Ash
- Department of Internal MedicinePappajohn Biomedical InstituteRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Brieanna M. Hilkin
- Department of Internal MedicinePappajohn Biomedical InstituteRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Nicholas D. Gansemer
- Department of Internal MedicinePappajohn Biomedical InstituteRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Eric A. Hoffman
- Department of RadiologyRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Roy J Carver, Department of Biomedical EngineeringUniversity of IowaIowa CityIowaUSA
| | - Joseph Zabner
- Department of Internal MedicinePappajohn Biomedical InstituteRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - David A. Stoltz
- Department of Internal MedicinePappajohn Biomedical InstituteRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Roy J Carver, Department of Biomedical EngineeringUniversity of IowaIowa CityIowaUSA
- Department of Molecular Physiology and BiophysicsRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Mahmoud H. Abou Alaiwa
- Department of Internal MedicinePappajohn Biomedical InstituteRoy J and Lucille A Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Roy J Carver, Department of Biomedical EngineeringUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
26
|
Bengtson CD, Kim MD, Salathe M. Is CF airway inflammation still relevant in the era of highly effective modulators? J Cyst Fibros 2022; 21:901-903. [PMID: 36028422 DOI: 10.1016/j.jcf.2022.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Charles D Bengtson
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael D Kim
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Matthias Salathe
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
27
|
Kim N, Kwak G, Rodriguez J, Livraghi-Butrico A, Zuo X, Simon V, Han E, Shenoy SK, Pandey N, Mazur M, Birket SE, Kim A, Rowe SM, Boucher R, Hanes J, Suk JS. Inhaled gene therapy of preclinical muco-obstructive lung diseases by nanoparticles capable of breaching the airway mucus barrier. Thorax 2022; 77:812-820. [PMID: 34697091 PMCID: PMC9129924 DOI: 10.1136/thoraxjnl-2020-215185] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/27/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Inhaled gene therapy of muco-obstructive lung diseases requires a strategy to achieve therapeutically relevant gene transfer to airway epithelium covered by particularly dehydrated and condensed mucus gel layer. Here, we introduce a synthetic DNA-loaded mucus-penetrating particle (DNA-MPP) capable of providing safe, widespread and robust transgene expression in in vivo and in vitro models of muco-obstructive lung diseases. METHODS We investigated the ability of DNA-MPP to mediate reporter and/or therapeutic transgene expression in lung airways of a transgenic mouse model of muco-obstructive lung diseases (ie, Scnn1b-Tg) and in air-liquid interface cultures of primary human bronchial epithelial cells harvested from an individual with cystic fibrosis. A plasmid designed to silence epithelial sodium channel (ENaC) hyperactivity, which causes airway surface dehydration and mucus stasis, was intratracheally administered via DNA-MPP to evaluate therapeutic effects in vivo with or without pretreatment with hypertonic saline, a clinically used mucus-rehydrating agent. RESULTS DNA-MPP exhibited marked greater reporter transgene expression compared with a mucus-impermeable formulation in in vivo and in vitro models of muco-obstructive lung diseases. DNA-MPP carrying ENaC-silencing plasmids provided efficient downregulation of ENaC and reduction of mucus burden in the lungs of Scnn1b-Tg mice, and synergistic impacts on both gene transfer efficacy and therapeutic effects were achieved when DNA-MPP was adjuvanted with hypertonic saline. DISCUSSION DNA-MPP constitutes one of the rare gene delivery systems providing therapeutically meaningful gene transfer efficacy in highly relevant in vivo and in vitro models of muco-obstructive lung diseases due to its unique ability to efficiently penetrate airway mucus.
Collapse
Affiliation(s)
- Namho Kim
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Whiting School of Engineering, Baltimore, Maryland, USA
| | - Gijung Kwak
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jason Rodriguez
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alessandra Livraghi-Butrico
- Marisco Lung Institute and Cystic Fibrosis Research Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Xinyuan Zuo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Whiting School of Engineering, Baltimore, Maryland, USA
| | - Valentina Simon
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Eric Han
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Siddharth Kaup Shenoy
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marina Mazur
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama School of Medicine, Birmingham, Alabama, USA
| | - Susan E Birket
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama School of Medicine, Birmingham, Alabama, USA
- Department of Medicine, The University of Alabama, Birmingham, Alabama, USA
| | - Anthony Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Steven M Rowe
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama School of Medicine, Birmingham, Alabama, USA
- Department of Medicine, The University of Alabama, Birmingham, Alabama, USA
| | - Richard Boucher
- Marisco Lung Institute and Cystic Fibrosis Research Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Justin Hanes
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Whiting School of Engineering, Baltimore, Maryland, USA
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Environmental and Health Sciences, Oncology, Neurosurgery, and Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jung Soo Suk
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Whiting School of Engineering, Baltimore, Maryland, USA
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Lepissier A, Addy C, Hayes K, Noel S, Bui S, Burgel PR, Dupont L, Eickmeier O, Fayon M, Leal T, Lopes C, Downey DG, Sermet-Gaudelus I. Inflammation biomarkers in sputum for clinical trials in cystic fibrosis: current understanding and gaps in knowledge. J Cyst Fibros 2022; 21:691-706. [PMID: 34772643 DOI: 10.1016/j.jcf.2021.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022]
Abstract
RATIONALE Sputum biomarkers hold promise as a direct measure of inflammation within the cystic fibrosis (CF) lung, but variability in study design and sampling methodology have limited their use. A full evaluation of the reliability, validity and clinical relevance of individual biomarkers is required to optimise their use within CF clinical research. OBJECTIVES A biomarker Special Interest Working Group was established within the European Cystic Fibrosis Society-Clinical Trials Network Standardisation Committee, to perform a review of the evidence regarding sputum biomarkers in CF. METHODS From the 139 included articles, we identified 71 sputum biomarkers to undergo evaluation of their clinimetric properties, responsiveness, discriminant, concurrent and convergent validity. RESULTS Current evidence confirms the potential of sputum biomarkers as outcome measures in clinical trials. Inconsistency in responsiveness, concurrent and convergent validity require further research into these markers and processing standardisation before translation into wider use. Of the 71 biomarkers identified, Neutrophil Elastase (NE), IL-8, TNF-α and IL-1β, demonstrated validity and responsiveness to be currently considered for use in clinical trials. Other biomarkers show future promise, including IL-6, calprotectin, HMGB-1 and YKL-40. CONCLUSION A concerted international effort across the cystic fibrosis community is needed to promote high quality biomarker trial design, establish large population-based biomarker studies, and work together to create standards for collection, storage and analysis of sputum biomarkers.
Collapse
Affiliation(s)
- Agathe Lepissier
- Paediatric Center for Cystic Fibrosis, Centre de Référence Maladies Rares, Mucoviscidose et Maladies Apparentées, Hôpital Necker Enfants Malades 149 rue de Sévres, Paris 75743, France; INSERM U1151, Institut Necker Enfants Malades, 160 rue de Vaugirard, Paris 75743, France; European Reference Network (ERN Lung)
| | - Charlotte Addy
- Northern Ireland Clinical Research Facility, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL; All Wales Adult Cystic Fibrosis Centre, University Hopsital Llandough, Penlan Road, CF64 2XX
| | - Kate Hayes
- Northern Ireland Clinical Research Facility, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Sabrina Noel
- INSERM U1151, Institut Necker Enfants Malades, 160 rue de Vaugirard, Paris 75743, France
| | - Stéphanie Bui
- Université de Bordeaux (INSERM U1045), CHU de Bordeaux, (CIC1401), F-33000 Bordeaux, France
| | - Pierre-Régis Burgel
- European Reference Network (ERN Lung); National Reference Cystic Fibrosis Center and Department of Respiratory Medicine, Cochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, 75014, France; Institut Cochin, INSERM U1016 and Université de Paris; Paris 75014, France
| | - Lieven Dupont
- University Hospital Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | - Olaf Eickmeier
- Facharzt für Kinder- und Jugendmedizin, Universitätsklinikum Frankfurt a.M., Johann Wolfgang-Goethe-Universität, Allergologie, Pneumologie & Mukoviszidose, Theodor-Stern-Kai 7, 60590 Frankfurt/Main
| | - Michael Fayon
- Université de Bordeaux (INSERM U1045), CHU de Bordeaux, (CIC1401), F-33000 Bordeaux, France
| | - Teresinha Leal
- Louvain Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Carlos Lopes
- Departamento do Tórax, Hospital de Santa Maria, Lisbon
| | - Damian G Downey
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Isabelle Sermet-Gaudelus
- Paediatric Center for Cystic Fibrosis, Centre de Référence Maladies Rares, Mucoviscidose et Maladies Apparentées, Hôpital Necker Enfants Malades 149 rue de Sévres, Paris 75743, France; INSERM U1151, Institut Necker Enfants Malades, 160 rue de Vaugirard, Paris 75743, France; European Reference Network (ERN Lung); Service de Pneumologie et Allergologie Pédiatriques, Centre de Ressources et de Compétence de la Mucoviscidose, Hôpital Necker Enfants Malades 149 rue de Sévres, INSERM U1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris 75743, France.
| |
Collapse
|
29
|
Grubb BR, Livraghi-Butrico A. Animal models of cystic fibrosis in the era of highly effective modulator therapies. Curr Opin Pharmacol 2022; 64:102235. [DOI: 10.1016/j.coph.2022.102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022]
|
30
|
Keith JD, Henderson AG, Fernandez-Petty CM, Davis JM, Oden AM, Birket SE. Muc5b Contributes to Mucus Abnormality in Rat Models of Cystic Fibrosis. Front Physiol 2022; 13:884166. [PMID: 35574458 PMCID: PMC9096080 DOI: 10.3389/fphys.2022.884166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) airway disease is characterized by excessive and accumulative mucus in the airways. Mucociliary clearance becomes defective as mucus secretions become hyperconcentrated and viscosity increases. The CFTR-knockout (KO) rat has been previously shown to progressively develop delayed mucociliary transport, secondary to increased viscoelasticity of airway secretions. The humanized-G551D CFTR rat model has demonstrated that abnormal mucociliary clearance and hyperviscosity is reversed by ivacaftor treatment. In this study, we sought to identify the components of mucus that changes as the rat ages to contribute to these abnormalities. We found that Muc5b concentrations, and to a lesser extent Muc5ac, in the airway were increased in the KO rat compared to WT, and that Muc5b concentration was directly related to the viscosity of the mucus. Additionally, we found that methacholine administration to the airway exacerbates these characteristics of disease in the KO, but not WT rat trachea. Lastly we determined that at 6 months of age, CF rats had mucus that was adherent to the airway epithelium, a process that is reversed by ivacaftor therapy in the hG551D rat. Overall, these data indicate that accumulation of Muc5b initiates the muco-obstructive process in the CF lung prior to infection.
Collapse
Affiliation(s)
- Johnathan D Keith
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Alexander G Henderson
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Courtney M Fernandez-Petty
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joy M Davis
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ashley M Oden
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Susan E Birket
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
31
|
Variation in CFTR-dependent ‘β-sweating’ among healthy adults. PLoS One 2022; 17:e0265432. [PMID: 35312728 PMCID: PMC8936459 DOI: 10.1371/journal.pone.0265432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 03/01/2022] [Indexed: 11/25/2022] Open
Abstract
The genetic disease cystic fibrosis (CF) results when mutations in the gene for the anion channel CFTR reduce CFTR’s activity below a critical level. CFTR activity = N·PO·γ (number of channels x open probability x channel conductance). Small molecules are now available that partially restore CFTR function with dramatic improvements in health of CF subjects. Continued evaluation of these and other compounds in development will be aided by accurate assessments of CFTR function. However, measuring CFTR activity in vivo is challenging and estimates vary widely. The most accurate known measure of CFTR activity in vivo is the ‘β/M’ ratio of sweat rates, which is produced by stimulation with a β-adrenergic agonist cocktail referenced to the same individual’s methacholine-stimulated sweat rate. The most meaningful metric of CFTR activity is to express it as a percent of normal function, so it is critical to establish β/M carefully in a population of healthy control subjects. Here, we analyze β/M from a sample of 50 healthy adults in which sweat rates to cholinergic and β-adrenergic agonists were measured repeatedly (3 times) in multiple, (~50) identified sweat glands from each individual (giving ~20,000 measurements). The results show an approximately 7-fold range, 26–187% of the WT average set to 100%. These provide a benchmark against which other measures of CFTR activity can be compared. Factors contributing to β/M variation in healthy controls are discussed.
Collapse
|
32
|
He N, Liu X, Vegter AR, Evans TIA, Gray JS, Guo J, Moll SR, Guo LJ, Luo M, Ma N, Sun X, Liang B, Yan Z, Feng Z, Qi L, Joshi AS, Shahin W, Yi Y, Gibson-Corley KN, Hoffman EA, Wang K, Mueller C, Engelhardt JF, Rosen BH. Ferret models of alpha-1 antitrypsin deficiency develop lung and liver disease. JCI Insight 2022; 7:e143004. [PMID: 35104244 PMCID: PMC8983124 DOI: 10.1172/jci.insight.143004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/28/2022] [Indexed: 11/19/2022] Open
Abstract
Alpha-1 antitrypsin deficiency (AATD) is the most common genetic cause and risk factor for chronic obstructive pulmonary disease, but the field lacks a large-animal model that allows for longitudinal assessment of pulmonary function. We hypothesized that ferrets would model human AATD-related lung and hepatic disease. AAT-knockout (AAT-KO) and PiZZ (E342K, the most common mutation in humans) ferrets were generated and compared with matched controls using custom-designed flexiVent modules to perform pulmonary function tests, quantitative computed tomography (QCT), bronchoalveolar lavage (BAL) proteomics, and alveolar morphometry. Complete loss of AAT (AAT-KO) led to increased pulmonary compliance and expiratory airflow limitation, consistent with obstructive lung disease. QCT and morphometry confirmed emphysema and airspace enlargement, respectively. Pathway analysis of BAL proteomics data revealed inflammatory lung disease and impaired cellular migration. The PiZ mutation resulted in altered AAT protein folding in the liver, hepatic injury, and reduced plasma concentrations of AAT, and PiZZ ferrets developed obstructive lung disease. In summary, AAT-KO and PiZZ ferrets model the progressive obstructive pulmonary disease seen in AAT-deficient patients and may serve as a platform for preclinical testing of therapeutics including gene therapy.
Collapse
Affiliation(s)
- Nan He
- Department of Anatomy and Cell Biology
| | | | | | | | | | | | | | | | | | | | | | - Bo Liang
- Department of Anatomy and Cell Biology
| | | | | | - Lisi Qi
- Department of Anatomy and Cell Biology
| | | | | | - Yaling Yi
- Department of Anatomy and Cell Biology
| | | | | | - Kai Wang
- Department of Biostatistics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Christian Mueller
- Department of Pediatrics, University of Massachusetts Medical Center, Worcester, Massachusetts, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Bradley H. Rosen
- Department of Anatomy and Cell Biology
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
33
|
Ekkelenkamp MB, Díez-Aguilar M, Tunney MM, Elborn JS, Fluit AC, Cantón R. Establishing antimicrobial susceptibility testing methods and clinical breakpoints for inhaled antibiotic therapy. Open Forum Infect Dis 2022; 9:ofac082. [PMID: 35265731 PMCID: PMC8900927 DOI: 10.1093/ofid/ofac082] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/10/2022] [Indexed: 12/05/2022] Open
Abstract
Inhaled antibiotics are a common and valuable therapy for patients suffering from chronic lung infection, with this particularly well demonstrated for patients with cystic fibrosis. However, in vitro tests to predict patient response to inhaled antibiotic therapy are currently lacking. There are indications that antimicrobial susceptibility testing (AST) may have a role in guidance of therapy, but which tests would correlate best still needs to be researched in clinical studies or animal models. Applying the principles of European Committee on Antimicrobial Susceptibility Testing methodology, the analysis of relevant and reliable data correlating different AST tests to patients’ outcomes may yield clinical breakpoints for susceptibility, but these data are currently unavailable. At present, we believe that it is unlikely that standard determination of minimum inhibitory concentration will prove the best predictor.
Collapse
Affiliation(s)
- Miquel B Ekkelenkamp
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - María Díez-Aguilar
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Servicio de Microbiología y Parasitología, Hospital Universitario La Princesa, Madrid, Spain
| | - Michael M Tunney
- Queen’s University Belfast, Department of Pulmonology, Belfast, United Kingdom
| | - J Stuart Elborn
- Queen’s University Belfast, Department of Pulmonology, Belfast, United Kingdom
| | - Ad C Fluit
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - Rafael Cantón
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
34
|
Novel Immunomodulatory Therapies for Respiratory Pathologies. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC8238403 DOI: 10.1016/b978-0-12-820472-6.00073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
35
|
Lukasiak A, Zajac M. The Distribution and Role of the CFTR Protein in the Intracellular Compartments. MEMBRANES 2021; 11:membranes11110804. [PMID: 34832033 PMCID: PMC8618639 DOI: 10.3390/membranes11110804] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis is a hereditary disease that mainly affects secretory organs in humans. It is caused by mutations in the gene encoding CFTR with the most common phenylalanine deletion at position 508. CFTR is an anion channel mainly conducting Cl− across the apical membranes of many different epithelial cells, the impairment of which causes dysregulation of epithelial fluid secretion and thickening of the mucus. This, in turn, leads to the dysfunction of organs such as the lungs, pancreas, kidney and liver. The CFTR protein is mainly localized in the plasma membrane; however, there is a growing body of evidence that it is also present in the intracellular organelles such as the endosomes, lysosomes, phagosomes and mitochondria. Dysfunction of the CFTR protein affects not only the ion transport across the epithelial tissues, but also has an impact on the proper functioning of the intracellular compartments. The review aims to provide a summary of the present state of knowledge regarding CFTR localization and function in intracellular compartments, the physiological role of this localization and the consequences of protein dysfunction at cellular, epithelial and organ levels. An in-depth understanding of intracellular processes involved in CFTR impairment may reveal novel opportunities in pharmacological agents of cystic fibrosis.
Collapse
|
36
|
Hanssens LS, Duchateau J, Casimir GJ. CFTR Protein: Not Just a Chloride Channel? Cells 2021; 10:2844. [PMID: 34831067 PMCID: PMC8616376 DOI: 10.3390/cells10112844] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/17/2022] Open
Abstract
Cystic fibrosis (CF) is a recessive genetic disease caused by mutations in a gene encoding a protein called Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). The CFTR protein is known to acts as a chloride (Cl-) channel expressed in the exocrine glands of several body systems where it also regulates other ion channels, including the epithelial sodium (Na+) channel (ENaC) that plays a key role in salt absorption. This function is crucial to the osmotic balance of the mucus and its viscosity. However, the pathophysiology of CF is more challenging than a mere dysregulation of epithelial ion transport, mainly resulting in impaired mucociliary clearance (MCC) with consecutive bronchiectasis and in exocrine pancreatic insufficiency. This review shows that the CFTR protein is not just a chloride channel. For a long time, research in CF has focused on abnormal Cl- and Na+ transport. Yet, the CFTR protein also regulates numerous other pathways, such as the transport of HCO3-, glutathione and thiocyanate, immune cells, and the metabolism of lipids. It influences the pH homeostasis of airway surface liquid and thus the MCC as well as innate immunity leading to chronic infection and inflammation, all of which are considered as key pathophysiological characteristics of CF.
Collapse
Affiliation(s)
- Laurence S. Hanssens
- Department of Pediatric Pulmonology and Cystic Fibrosis Clinic, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Avenue J.J. Crocq 15, 1020 Brussels, Belgium;
| | - Jean Duchateau
- Laboratoire Académique de Pédiatrie, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Avenue J.J. Crocq 15, 1020 Brussels, Belgium;
| | - Georges J. Casimir
- Department of Pediatric Pulmonology and Cystic Fibrosis Clinic, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Avenue J.J. Crocq 15, 1020 Brussels, Belgium;
- Laboratoire Académique de Pédiatrie, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Avenue J.J. Crocq 15, 1020 Brussels, Belgium;
| |
Collapse
|
37
|
O’Connor JB, Mottlowitz MM, Wagner BD, Boyne KL, Stevens MJ, Robertson CE, Harris JK, Laguna TA. Divergence of bacterial communities in the lower airways of CF patients in early childhood. PLoS One 2021; 16:e0257838. [PMID: 34613995 PMCID: PMC8494354 DOI: 10.1371/journal.pone.0257838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
Rationale Chronic airway infection and inflammation resulting in progressive, obstructive lung disease is the leading cause of morbidity and mortality in cystic fibrosis. Understanding the lower airway microbiota across the ages can provide valuable insight and potential therapeutic targets. Objectives To characterize and compare the lower airway microbiota in cystic fibrosis and disease control subjects across the pediatric age spectrum. Methods Bronchoalveolar lavage fluid samples from 191 subjects (63 with cystic fibrosis) aged 0 to 21 years were collected along with relevant clinical data. We measured total bacterial load using quantitative polymerase chain reaction and performed 16S rRNA gene sequencing to characterize bacterial communities with species-level sensitivity for select genera. Clinical comparisons were investigated. Measurements and main results Cystic fibrosis samples had higher total bacterial load and lower microbial diversity, with a divergence from disease controls around 2–5 years of age, as well as higher neutrophilic inflammation relative to bacterial burden. Cystic fibrosis samples had increased abundance of traditional cystic fibrosis pathogens and decreased abundance of the Streptococcus mitis species group in older subjects. Interestingly, increased diversity in the heterogeneous disease controls was independent of diagnosis and indication. Sequencing was more sensitive than culture, and antibiotic exposure was more common in disease controls, which showed a negative relationship with load and neutrophilic inflammation. Conclusions Analysis of lower airway samples from people with cystic fibrosis and disease controls across the ages revealed key differences in airway microbiota and inflammation. The divergence in subjects during early childhood may represent a window of opportunity for intervention and additional study.
Collapse
Affiliation(s)
- John B. O’Connor
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Madison M. Mottlowitz
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Brandie D. Wagner
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Kathleen L. Boyne
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Mark J. Stevens
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Charles E. Robertson
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Jonathan K. Harris
- Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Theresa A. Laguna
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
38
|
Combined agonists act synergistically to increase mucociliary clearance in a cystic fibrosis airway model. Sci Rep 2021; 11:18828. [PMID: 34552115 PMCID: PMC8458446 DOI: 10.1038/s41598-021-98122-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/03/2021] [Indexed: 11/30/2022] Open
Abstract
Mucus clearance, a primary innate defense mechanism of airways, is defective in patients with cystic fibrosis (CF) and CF animals. In previous work, the combination of a low dose of the cholinergic agonist, carbachol with forskolin or a β adrenergic agonist, isoproterenol synergistically increased mucociliary clearance velocity (MCCV) in ferret tracheas. Importantly, the present study shows that synergistic MCCV can also be produced in CF ferrets, with increases ~ 55% of WT. Synergistic MCCV was also produced in pigs. The combined agonists increased MCCV by increasing surface fluid via multiple mechanisms: increased fluid secretion from submucosal glands, increased anion secretion across surface epithelia and decreased Na+ absorption. To avoid bronchoconstriction, the cAMP agonist was applied 30 min before carbachol. This approach to increasing mucus clearance warrants testing for safety and efficacy in humans as a potential therapeutic for muco-obstructive diseases.
Collapse
|
39
|
Lu S, Kolls JK. Early Antibiotics in Cystic Fibrosis: Lessons from the CF Pig Model. Am J Respir Crit Care Med 2021; 204:626-627. [PMID: 34343466 PMCID: PMC8521697 DOI: 10.1164/rccm.202106-1383ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Shiping Lu
- Tulane University School of Medicine, 12255, Medicine, New Orleans, Louisiana, United States
| | - Jay K Kolls
- Tulane University School of Medicine, 12255, Medicine, New Orleans, Louisiana, United States;
| |
Collapse
|
40
|
Voynow JA, Shinbashi M. Neutrophil Elastase and Chronic Lung Disease. Biomolecules 2021; 11:biom11081065. [PMID: 34439732 PMCID: PMC8394930 DOI: 10.3390/biom11081065] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022] Open
Abstract
Neutrophil elastase (NE) is a major inflammatory protease released by neutrophils and is present in the airways of patients with cystic fibrosis (CF), chronic obstructive pulmonary disease, non-CF bronchiectasis, and bronchopulmonary dysplasia. Although NE facilitates leukocyte transmigration to the site of infection and is required for clearance of Gram-negative bacteria, it also activates inflammation when released into the airway milieu in chronic inflammatory airway diseases. NE exposure induces airway remodeling with increased mucin expression and secretion and impaired ciliary motility. NE interrupts epithelial repair by promoting cellular apoptosis and senescence and it activates inflammation directly by increasing cytokine expression and release, and indirectly by triggering extracellular trap release and exosome release, which magnify protease activity and inflammation in the airway. NE inhibits innate immune function by digesting opsonins and opsonin receptors, degrading innate immune proteins such as lactoferrin, and inhibiting macrophage phagocytosis. Importantly, NE-directed therapies have not yet been effective in preventing the pathologic sequelae of NE exposure, but new therapies are being developed that offer both direct antiprotease activity and multifunctional anti-inflammatory properties.
Collapse
Affiliation(s)
- Judith A. Voynow
- Division of Pediatric Pulmonology, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, VA 23298, USA
- Correspondence:
| | - Meagan Shinbashi
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| |
Collapse
|
41
|
Mandal P, Lyons JD, Burd EM, Koval M, Mocarski ES, Coopersmith CM. Integrated evaluation of lung disease in single animals. PLoS One 2021; 16:e0246270. [PMID: 34237078 PMCID: PMC8266100 DOI: 10.1371/journal.pone.0246270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 01/15/2021] [Indexed: 11/18/2022] Open
Abstract
During infectious disease, pathogen load drives inflammation and immune response that together contribute to tissue injury often resulting in organ dysfunction. Pulmonary failure in SARS-CoV2-infected hospitalized COVID-19 patients is one such prominent example. Intervention strategies require characterization of the host-pathogen interaction by accurately assessing all of the above-mentioned disease parameters. To study infection in intact mammals, mice are often used as essential genetic models. Due to humane concerns, there is a constant unmet demand to develop studies that reduce the number of mice utilized while generating objective data. Here, we describe an integrated method of evaluating lung inflammation in mice infected with Pseudomonas aeruginosa or murine gammaherpesvirus (MHV)-68. This method conserves animal resources while permitting evaluation of disease mechanisms in both infection settings. Lungs from a single euthanized mouse were used for two purposes-biological assays to determine inflammation and infection load, as well as histology to evaluate tissue architecture. For this concurrent assessment of multiple parameters from a single euthanized mouse, we limit in-situ formalin fixation to the right lung of the cadaver. The unfixed left lung is collected immediately and divided into several segments for biological assays including determination of pathogen titer, assessment of infection-driven cytokine levels and appearance of cell death markers. In situ fixed right lung was then processed for histological determination of tissue injury and confirmation of infection-driven cell death patterns. This method reduces overall animal use and minimizes inter-animal variability that results from sacrificing different animals for different types of assays. The technique can be applied to any lung disease study in mice or other mammals.
Collapse
Affiliation(s)
- Pratyusha Mandal
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - John D. Lyons
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Eileen M. Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine and Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Edward S. Mocarski
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Craig M. Coopersmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
42
|
Bouzek DC, Abou Alaiwa MH, Adam RJ, Pezzulo AA, Reznikov LR, Cook DP, Aguilar Pescozo MI, Ten Eyck P, Wu C, Gross TJ, Hornick DB, Hoffman EA, Meyerholz DK, Stoltz DA. Early Lung Disease Exhibits Bacterial-Dependent and -Independent Abnormalities in Cystic Fibrosis Pigs. Am J Respir Crit Care Med 2021; 204:692-702. [PMID: 34170795 DOI: 10.1164/rccm.202102-0451oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE While it is clear that cystic fibrosis airway disease begins at a very young age, the early and subsequent steps in disease pathogenesis and the relative contribution of infection, mucus, and inflammation are not well understood. OBJECTIVES As one approach to assessing the early contribution of infection, we tested the hypothesis that early and continuous antibiotics would decrease the airway bacterial burden. We thought that, if it does, it might reveal aspects of the disease that are more or less sensitive to decreasing infection. METHODS Three groups of pigs were studied from birth until ~3 weeks of age: 1) wild-type, 2) cystic fibrosis, and 3) cystic fibrosis pigs treated continuously with broad-spectrum antibiotics from birth until study completion. Disease was assessed with chest computed tomography, histopathology, microbiology, and bronchoalveolar lavage. MEASUREMENTS AND MAIN RESULTS Disease was present by 3 weeks of age in cystic fibrosis pigs. Continuous antibiotics from birth improved chest computed tomography imaging abnormalities and airway mucus accumulation, but not airway inflammation in the cystic fibrosis pig model. However, reducing bacterial infection did not improve two disease features already present at birth in cystic fibrosis pigs, air trapping and submucosal gland duct plugging. In the cystic fibrosis sinuses, antibiotics did not prevent the development of infection, disease, or the number of bacteria but did alter the bacterial species. CONCLUSIONS These findings suggest that cystic fibrosis airway disease begins immediately following birth, and that early and continuous antibiotics impact some, but not all, aspects of CF lung disease development.
Collapse
Affiliation(s)
- Drake C Bouzek
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Mahmoud H Abou Alaiwa
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Internal Medicine, Iowa City, Iowa, United States
| | - Ryan J Adam
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Alejandro A Pezzulo
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Internal Medicine, Iowa City, Iowa, United States
| | - Leah R Reznikov
- University of Florida, 3463, Physiological Sciences, Gainesville, Florida, United States
| | - Daniel P Cook
- Vanderbilt University Medical Center, 12328, Department of Medicine, Nashville, Tennessee, United States
| | - Maria I Aguilar Pescozo
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Patrick Ten Eyck
- The University of Iowa, 4083, Institute for Clinical and Translational Science, Iowa City, Iowa, United States
| | - Chaorong Wu
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Thomas J Gross
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Douglas B Hornick
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Eric A Hoffman
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Radiology, Iowa City, Iowa, United States
| | - David K Meyerholz
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Pathology, Iowa City, Iowa, United States
| | - David A Stoltz
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States;
| |
Collapse
|
43
|
Shteinberg M, Haq IJ, Polineni D, Davies JC. Cystic fibrosis. Lancet 2021; 397:2195-2211. [PMID: 34090606 DOI: 10.1016/s0140-6736(20)32542-3] [Citation(s) in RCA: 391] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/03/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis is a monogenic disease considered to affect at least 100 000 people worldwide. Mutations in CFTR, the gene encoding the epithelial ion channel that normally transports chloride and bicarbonate, lead to impaired mucus hydration and clearance. Classical cystic fibrosis is thus characterised by chronic pulmonary infection and inflammation, pancreatic exocrine insufficiency, male infertility, and might include several comorbidities such as cystic fibrosis-related diabetes or cystic fibrosis liver disease. This autosomal recessive disease is diagnosed in many regions following newborn screening, whereas in other regions, diagnosis is based on a group of recognised multiorgan clinical manifestations, raised sweat chloride concentrations, or CFTR mutations. Disease that is less easily diagnosed, and in some cases affecting only one organ, can be seen in the context of gene variants leading to residual protein function. Management strategies, including augmenting mucociliary clearance and aggressively treating infections, have gradually improved life expectancy for people with cystic fibrosis. However, restoration of CFTR function via new small molecule modulator drugs is transforming the disease for many patients. Clinical trial pipelines are actively exploring many other approaches, which will be increasingly needed as survival improves and as the population of adults with cystic fibrosis increases. Here, we present the current understanding of CFTR mutations, protein function, and disease pathophysiology, consider strengths and limitations of current management strategies, and look to the future of multidisciplinary care for those with cystic fibrosis.
Collapse
Affiliation(s)
- Michal Shteinberg
- Pulmonology Institute and CF Center, Carmel Medical Center, Haifa, Israel; Rappaport Faculty of Medicine, The Technion-Israel Institute of Technology, Haifa, Israel
| | - Iram J Haq
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Jane C Davies
- National Heart and Lung Institute, Imperial College London, London, UK; Royal Brompton and Harefield, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
44
|
Green M, Lindgren N, Henderson A, Keith JD, Oden AM, Birket SE. Ivacaftor partially corrects airway inflammation in a humanized G551D rat. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1093-L1100. [PMID: 33825507 PMCID: PMC8285630 DOI: 10.1152/ajplung.00082.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 01/09/2023] Open
Abstract
Animal models have been highly informative for understanding the pathogenesis and progression of cystic fibrosis (CF) lung disease. In particular, the CF rat models recently developed have addressed mechanistic causes of the airway mucus defect characteristic of CF, and how these may change when cystic fibrosis transmembrane conductance regulator (CFTR) activity is restored using new modulator therapies. We hypothesized that inflammatory changes to the airway would develop spontaneously and progressively, and that these changes would be resolved with modulator therapy. To test this, we used a humanized-CFTR rat expressing the G551D variant that responds to the CFTR modulator ivacaftor. Markers typically found in the CF lung were assessed, including neutrophil influx, small airway histopathology, and inflammatory cytokine concentration. Young hG551D rats did not express inflammatory cytokines at baseline but did upregulate these in response to inflammatory trigger. As the hG551D rats aged, histopathology worsened, accompanied by neutrophil influx into the airway and increasing concentrations of TNF-α, IL-1α, and IL-6 in the airways. Ivacaftor administration reduced concentrations of these cytokines when administered to the rats at baseline but was less effective in the rats that had also received inflammatory stimulus. Therefore, we conclude that administration of ivacaftor resulted in an incomplete resolution of inflammation when rats received an external trigger, suggesting that CFTR activation may not be enough to resolve inflammation in the lungs of patients with CF.
Collapse
Affiliation(s)
- Morgan Green
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Natalie Lindgren
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Alexander Henderson
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Johnathan D Keith
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ashley M Oden
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Susan E Birket
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
45
|
Westhölter D, Beckert H, Straßburg S, Welsner M, Sutharsan S, Taube C, Reuter S. Pseudomonas aeruginosa infection, but not mono or dual-combination CFTR modulator therapy affects circulating regulatory T cells in an adult population with cystic fibrosis. J Cyst Fibros 2021; 20:1072-1079. [PMID: 34030985 DOI: 10.1016/j.jcf.2021.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/20/2021] [Accepted: 05/01/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Chronic infection and an exaggerated inflammatory response are key drivers of the pathogenesis of cystic fibrosis (CF), especially CF lung disease. An imbalance of pro- and anti-inflammatory mediators, including dysregulated Th2/Th17 cells and impairment of regulatory T cells (Tregs), maintain CF inflammation. CF transmembrane conductance regulator (CFTR) modulator therapy might influence these immune cell abnormalities. METHODS Peripheral blood mononuclear cells and serum samples were collected from 108 patients with CF (PWCF) and 40 patients with non-CF bronchiectasis. Samples were analysed for peripheral blood lymphocytes subsets (Tregs; Th1-, Th1/17-, Th17- and Th2-effector cells) and systemic T helper cell-associated cytokines (interleukin [IL]-5, IL-13, IL-2, IL-6, IL-9, IL-10, IL-17A, IL-17F, IL-4, IL-22, interferon-γ, tumour necrosis factor-α) using flow cytometry. RESULTS 51% of PWCF received CFTR modulators (ivacaftor, ivacaftor/ lumacaftor or tezacaftor/ ivacaftor). There were no differences in proportions of analysed T cell subsets or cytokines between PWCF who were versus were not receiving CFTR modulators. Additional analysis revealed lower percentages of Tregs in PWCF and chronic pulmonary Pseudomonas aeruginosa infection; this difference was also present in PWCF treated with CFTR modulators. Patients with non-CF bronchiectasis tended to have higher percentages of Th2- and Th17-cells and higher levels of peripheral cytokines versus PWCF. CONCLUSIONS Chronic P. aeruginosa lung infection appears to impair Tregs in PWCF (independent of CFTR modulator therapy) but not those with non-CF bronchiectasis. Moreover, our data showed no statistically significant differences in major subsets of peripheral lymphocytes and cytokines among PWCF who were versus were not receiving CFTR modulators.
Collapse
Affiliation(s)
- Dirk Westhölter
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany.
| | - Hendrik Beckert
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Svenja Straßburg
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany; Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Matthias Welsner
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany; Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Sivagurunathan Sutharsan
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany; Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| |
Collapse
|
46
|
van Heeckeren AM, Sutton MT, Fletcher DR, Hodges CA, Caplan AI, Bonfield TL. Enhancing Cystic Fibrosis Immune Regulation. Front Pharmacol 2021; 12:573065. [PMID: 34054509 PMCID: PMC8155373 DOI: 10.3389/fphar.2021.573065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023] Open
Abstract
In cystic fibrosis (CF), sustained infection and exuberant inflammation results in debilitating and often fatal lung disease. Advancement in CF therapeutics has provided successful treatment regimens for a variety of clinical consequences in CF; however effective means to treat the pulmonary infection and inflammation continues to be problematic. Even with the successful development of small molecule cystic fibrosis transmembrane conductance regulator (CFTR) correctors and potentiators, there is only a modest effect on established infection and inflammation in CF patients. In the pursuit of therapeutics to treat inflammation, the conundrum to address is how to overcome the inflammatory response without jeopardizing the required immunity to manage pathogens and prevent infection. The key therapeutic would have the capacity to dull the inflammatory response, while sustaining the ability to manage infections. Advances in cell-based therapy have opened up the avenue for dynamic and versatile immune interventions that may support this requirement. Cell based therapy has the capacity to augment the patient’s own ability to manage their inflammatory status while at the same time sustaining anti-pathogen immunity. The studies highlighted in this manuscript outline the potential use of cell-based therapy for CF. The data demonstrate that 1) total bone marrow aspirates containing Cftr sufficient hematopoietic and mesenchymal stem cells (hMSCs) provide Cftr deficient mice >50% improvement in survival and improved management of infection and inflammation; 2) myeloid cells can provide sufficient Cftr to provide pre-clinical anti-inflammatory and antimicrobial benefit; 3) hMSCs provide significant improvement in survival and management of infection and inflammation in CF; 4) the combined interaction between macrophages and hMSCs can potentially enhance anti-inflammatory and antimicrobial support through manipulating PPARγ. These data support the development of optimized cell-based therapeutics to enhance CF patient’s own immune repertoire and capacity to maintain the balance between inflammation and pathogen management.
Collapse
Affiliation(s)
- Anna M van Heeckeren
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Morgan T Sutton
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,St. Jude Children's Research Hospital Graduate School of Biomedical Sciences, Memphis, TN, United States
| | - David R Fletcher
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Craig A Hodges
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Arnold I Caplan
- Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Tracey L Bonfield
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
47
|
McKelvey MC, Brown R, Ryan S, Mall MA, Weldon S, Taggart CC. Proteases, Mucus, and Mucosal Immunity in Chronic Lung Disease. Int J Mol Sci 2021; 22:5018. [PMID: 34065111 PMCID: PMC8125985 DOI: 10.3390/ijms22095018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Dysregulated protease activity has long been implicated in the pathogenesis of chronic lung diseases and especially in conditions that display mucus obstruction, such as chronic obstructive pulmonary disease, cystic fibrosis, and non-cystic fibrosis bronchiectasis. However, our appreciation of the roles of proteases in various aspects of such diseases continues to grow. Patients with muco-obstructive lung disease experience progressive spirals of inflammation, mucostasis, airway infection and lung function decline. Some therapies exist for the treatment of these symptoms, but they are unable to halt disease progression and patients may benefit from novel adjunct therapies. In this review, we highlight how proteases act as multifunctional enzymes that are vital for normal airway homeostasis but, when their activity becomes immoderate, also directly contribute to airway dysfunction, and impair the processes that could resolve disease. We focus on how proteases regulate the state of mucus at the airway surface, impair mucociliary clearance and ultimately, promote mucostasis. We discuss how, in parallel, proteases are able to promote an inflammatory environment in the airways by mediating proinflammatory signalling, compromising host defence mechanisms and perpetuating their own proteolytic activity causing structural lung damage. Finally, we discuss some possible reasons for the clinical inefficacy of protease inhibitors to date and propose that, especially in a combination therapy approach, proteases represent attractive therapeutic targets for muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Ryan Brown
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Sinéad Ryan
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| |
Collapse
|
48
|
Bayfield KJ, Douglas TA, Rosenow T, Davies JC, Elborn SJ, Mall M, Paproki A, Ratjen F, Sly PD, Smyth AR, Stick S, Wainwright CE, Robinson PD. Time to get serious about the detection and monitoring of early lung disease in cystic fibrosis. Thorax 2021; 76:1255-1265. [PMID: 33927017 DOI: 10.1136/thoraxjnl-2020-216085] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/24/2021] [Accepted: 03/10/2021] [Indexed: 12/26/2022]
Abstract
Structural and functional defects within the lungs of children with cystic fibrosis (CF) are detectable soon after birth and progress throughout preschool years often without overt clinical signs or symptoms. By school age, most children have structural changes such as bronchiectasis or gas trapping/hypoperfusion and lung function abnormalities that persist into later life. Despite improved survival, gains in forced expiratory volume in one second (FEV1) achieved across successive birth cohorts during childhood have plateaued, and rates of FEV1 decline in adolescence and adulthood have not slowed. This suggests that interventions aimed at preventing lung disease should be targeted to mild disease and commence in early life. Spirometry-based classifications of 'normal' (FEV1≥90% predicted) and 'mild lung disease' (FEV1 70%-89% predicted) are inappropriate, given the failure of spirometry to detect significant structural or functional abnormalities shown by more sensitive imaging and lung function techniques. The state and readiness of two imaging (CT and MRI) and two functional (multiple breath washout and oscillometry) tools for the detection and monitoring of early lung disease in children and adults with CF are discussed in this article.Prospective research programmes and technological advances in these techniques mean that well-designed interventional trials in early lung disease, particularly in young children and infants, are possible. Age appropriate, randomised controlled trials are critical to determine the safety, efficacy and best use of new therapies in young children. Regulatory bodies continue to approve medications in young children based on safety data alone and extrapolation of efficacy results from older age groups. Harnessing the complementary information from structural and functional tools, with measures of inflammation and infection, will significantly advance our understanding of early CF lung disease pathophysiology and responses to therapy. Defining clinical utility for these novel techniques will require effective collaboration across multiple disciplines to address important remaining research questions. Future impact on existing management burden for patients with CF and their family must be considered, assessed and minimised.To address the possible role of these techniques in early lung disease, a meeting of international leaders and experts in the field was convened in August 2019 at the Australiasian Cystic Fibrosis Conference. The meeting entitiled 'Shaping imaging and functional testing for early disease detection of lung disease in Cystic Fibrosis', was attended by representatives across the range of disciplines involved in modern CF care. This document summarises the proceedings, key priorities and important research questions highlighted.
Collapse
Affiliation(s)
- Katie J Bayfield
- Department of Respiratory Medicine, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Tonia A Douglas
- Department of Respiratory and Sleep Medicine, Queensland Children's Hospital, South Brisbane, Queensland, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Tim Rosenow
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Perth, Western Australia, Australia
| | - Jane C Davies
- National Heart and Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Stuart J Elborn
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Marcus Mall
- Department of Pediatric Pulmonology, Immunology, and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Department of Translational Pulmonology, German Center for Lung Research, Berlin, Germany
| | - Anthony Paproki
- The Australian e-Health Research Centre, CSIRO, Brisbane, Queensland, Australia
| | - Felix Ratjen
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada.,University of Toronto, Toronto, Ontario, Canada
| | - Peter D Sly
- Children's Health and Environment Program, Child Health Research Centre, The University of Queenland, Herston, Queensland, Australia
| | - Alan R Smyth
- Division of Child Health, Obstetrics & Gynaecology. School of Medicine, University of Nottingham, Nottingham, Nottinghamshire, UK
| | - Stephen Stick
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| | - Claire E Wainwright
- Department of Respiratory and Sleep Medicine, Queensland Children's Hospital, South Brisbane, Queensland, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Paul D Robinson
- Department of Respiratory Medicine, Children's Hospital at Westmead, Westmead, New South Wales, Australia .,Airway Physiology and Imaging Group, Woolcock Institute of Medical Research, Glebe, New South Wales, Australia.,The Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
49
|
Pai AC, Parekh KR, Engelhardt JF, Lynch TJ. Ferret respiratory disease models for the study of lung stem cells. LUNG STEM CELLS IN DEVELOPMENT, HEALTH AND DISEASE 2021. [DOI: 10.1183/2312508x.10010320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
50
|
Gibson-Corley KN, Engelhardt JF. Animal Models and Their Role in Understanding the Pathophysiology of Cystic Fibrosis-Associated Gastrointestinal Lesions. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:51-67. [PMID: 33497264 DOI: 10.1146/annurev-pathol-022420-105133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The life expectancy of cystic fibrosis (CF) patients has greatly increased over the past decade, and researchers and clinicians must now navigate complex disease manifestations that were not a concern prior to the development of modern therapies. Explosive growth in the number of CF animal models has also occurred over this time span, clarifying CF disease pathophysiology and creating opportunities to understand more complex disease processes associated with an aging CF population. This review focuses on the CF-associated pathologies of the gastrointestinal system and how animal models have increased our understanding of this complex multisystemic disease. Although CF is primarily recognized as a pulmonary disease, gastrointestinal pathology occurs very commonly and can affect the quality of life for these patients. Furthermore, we discuss how next-generation genetic engineering of larger animal models will impact the field's understanding of CF disease pathophysiology and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Katherine N Gibson-Corley
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA.,Current affiliation: Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee 37232, USA;
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA;
| |
Collapse
|