1
|
Jinson S, Shanthikumar S, Werder RB. Evaluating the Use of iPSC-Derived Models in Understanding the Pathogenesis of Childhood Interstitial Lung Disease. Pediatr Pulmonol 2025; 60:e71143. [PMID: 40432281 PMCID: PMC12117302 DOI: 10.1002/ppul.71143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025]
Abstract
RATIONALE Genetic testing has significantly improved the diagnosis of childhood interstitial lung diseases (chILD), which have long challenged clinicians due to their heterogeneity and poor characterization. It is now imperative to study variants of unknown significance (VUS) to identify pathogenic mutations to optimize diagnosis and screening in patients. Furthermore, the limited treatment options for patients with chILD worsen patient outcomes. Induced pluripotent stem cell (iPSC)-derived models could be a tool to understand the effect of novel VUS and discover new therapeutic interventions. OBJECTIVE This review seeks to evaluate the fidelity of iPSC-derived models to recapitulate the pathogenic processes of chILD and test therapeutic interventions. METHODS This paper performs a systematic search over three databases to identify iPSC-derived models studying disease-causing mutations in pediatric patients with chILD and Hermansky-Pudlak Syndrome. RESULTS Of the 1452 papers initially reviewed, eight papers met the inclusion criteria using iPSC-derived models to study genetic variants. The diseases covered included common manifestations of chILDs such as surfactant protein deficiencies and hereditary pulmonary alveolar proteinosis, as well as fibrotic disorders like Hermansky-Pudlak Syndrome. These models recapitulated patient histology and key pathogenic features reported in the literature, delivering mechanistic insights into these conditions. Some papers also explored the efficacy of novel treatments, such as gene therapy. CONCLUSIONS iPSC-derived models can mimic aspects of human lung responses to provide a platform for disease modeling and therapeutic testing in chILD. There are opportunities to develop more complex multi-cellular models and for the study of a wider range of variants using these tools.
Collapse
Affiliation(s)
- Swetha Jinson
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW MelbourneMelbourneVictoriaAustralia
| | - Shivanthan Shanthikumar
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Respiratory and Sleep MedicineRoyal Children's HospitalMelbourneVictoriaAustralia
| | - Rhiannon B. Werder
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
2
|
Lee E, Ataya A, McCarthy C, Godart E, Cosenza J, King A, Robinson B, Wang T. The healthcare burden of pulmonary alveolar proteinosis (PAP). Orphanet J Rare Dis 2025; 20:73. [PMID: 39953603 PMCID: PMC11829527 DOI: 10.1186/s13023-024-03478-2] [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: 09/05/2024] [Accepted: 11/21/2024] [Indexed: 02/17/2025] Open
Abstract
INTRODUCTION Pulmonary alveolar proteinosis (PAP) is a rare lung syndrome characterized by the accumulation of surfactant in the alveoli. Using a longitudinal claims database, we compared measures of clinical and economic burden between a sample of diagnosed PAP patients and non-PAP matched controls. METHODS PAP patients were identified leveraging IPM.ai's longitudinal U.S. claims database spanning January 1, 2009, through May 1, 2022. PAP patients were selected based on the presence of ICD-10: J84.01 or ICD-9: 516.0 in their claims history and were indexed for observation. An age, gender, and geographically matched control cohort was created (ratio of 1:4) for comparison. A third cohort, consisting of likely undiagnosed PAP patients, was identified using a machine learning model. The PAP and control cohorts were tracked longitudinally, depending on individual index dates, from January 1, 2018, through May 1, 2023. Inclusion criteria required evidence of continual claims activity 12 months prior to and after the index date, which reduced the total number of diagnosed PAP and control patients in the analysis. Demographics, comorbidities, procedures, medication use, annual healthcare resource utilization (HCRU), and costs were calculated for eligible PAP and control patients and were compared 12 months prior to, and 12 months after each patient's index date. RESULTS After inclusion criteria were applied, 2312 confirmed PAP patients and 9247 matched controls were included in the analysis. Compared with matched controls, PAP patients had significantly higher rates of diagnosed conditions at baseline as defined by the Charlson Comorbidity Index (CCI). During the follow-up period, PAP patients had higher rates of diagnosed conditions, procedures, medication use, and cost-of-care compared with controls. PAP patients also had higher rates of emergency room visits (35% vs. 14%; P < 0.001), outpatient visits (87% vs. 56%; P < 0.001), inpatient visits (20% vs. 5%; P < 0.001) and had longer lengths of stay for inpatient hospitalizations (2.8 days vs. 0.56 days; P < 0.001), respectively. CONCLUSION This study represents the largest dataset of PAP patients and matched controls to be analyzed to date. Findings indicate that PAP patients have higher rates of diagnosed conditions, procedures, medication use, HCRU, and costs compared with non-PAP patients.
Collapse
Affiliation(s)
- Elinor Lee
- UCLA Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, 650 Charles E. Young Drive South, 43-229 CHS, Los Angeles, CA, 90095-1690, USA.
| | - Ali Ataya
- Division of Pulmonary and Critical Care Medicine, University of Florida, Gainesville, FL, USA
| | - Cormac McCarthy
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Erica Godart
- UCLA Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, 650 Charles E. Young Drive South, 43-229 CHS, Los Angeles, CA, 90095-1690, USA
| | - John Cosenza
- IPM.Ai, a Real Chemistry Company, New York, NY, USA
| | - Alysse King
- IPM.Ai, a Real Chemistry Company, New York, NY, USA
| | | | - Tisha Wang
- UCLA Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, 650 Charles E. Young Drive South, 43-229 CHS, Los Angeles, CA, 90095-1690, USA
| |
Collapse
|
3
|
Giunta-Stibb H, Hackett B. Interstitial lung disease in the newborn. J Perinatol 2025; 45:13-23. [PMID: 38956315 DOI: 10.1038/s41372-024-02036-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/30/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
Although relatively rare, interstitial lung diseases may present with respiratory distress in the newborn period. Most commonly these include developmental and growth disorders, disorders of surfactant synthesis and homeostasis, pulmonary interstitial glycogenosis, and neuroendocrine cell hyperplasia of infancy. Although the diagnosis of these disorders is sometimes made based on clinical presentation and imaging, due to the significant overlap between disorders and phenotypic variability, lung biopsy or, increasingly genetic testing is needed for diagnosis. These diseases may result in significant morbidity and mortality. Effective medical treatment options are in some cases limited and/or invasive. The genetic basis for some of these disorders has been identified, and with increased utilization of exome and whole genome sequencing even before lung biopsy, further insights into their genetic etiologies should become available.
Collapse
Affiliation(s)
- Hannah Giunta-Stibb
- Divisions of Neonatology and Pulmonology, Department of Pediatrics, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA.
| | - Brian Hackett
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
4
|
Tay CK, Kumar A, Hsu AAL, Lee P. Whole lung and sequential bronchoscopic lavage for pulmonary alveolar proteinosis. Curr Opin Pulm Med 2025; 31:41-52. [PMID: 39569652 DOI: 10.1097/mcp.0000000000001138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
PURPOSE OF REVIEW Pulmonary alveolar proteinosis (PAP) is a diffuse lung disease that results from the accumulation of lipoproteinaceous material in the alveoli due to abnormal surfactant homeostasis. Since its introduction in the 1960s, whole lung lavage (WLL) has been the primary treatment for PAP. This review focuses on WLL, including its technique modifications, and sequential bronchoscopic lavage. RECENT FINDINGS Autoimmune PAP, which accounts for the majority of cases, occurs when antigranulocyte-macrophage colony-stimulating factor (GM-CSF) autoantibodies lead to the deficiency of bioavailable GM-CSF. At present, there are no international guidelines or consensus statements for PAP treatment. Traditionally, therapeutic decisions are made based on the severity and type of PAP. Despite emerging data on GM-CSF-based therapies, WLL remains a central component in the therapeutic strategy for PAP. SUMMARY Although the technique of WLL has evolved over time, there is still no universally adopted, standardized protocol. However, key periprocedural aspects - such as preprocedural planning, patient evaluation, anesthetic technique, lavage protocol, and postprocedural care - remain essential to ensuring the safety and success of WLL.
Collapse
Affiliation(s)
- Chee Kiang Tay
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital
| | - Anupam Kumar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota Medical School
| | - Anne Ann Ling Hsu
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital
| | - Pyng Lee
- Division of Respiratory and Critical Care Medicine, National University Hospital, Singapore
| |
Collapse
|
5
|
Mishra-Sopori V, Khosla I, Khan S, Kataria D, Prabhudesai P, Chandane P, Sehgal K, Bodhanwala M, Pandrowala A, Hiwarkar P. Restitutio ad integrum: Rescuing the Alveolar Macrophage Function with HSCT in Pulmonary Alveolar Proteinosis Due to CSF2Rα Deficiency. J Clin Immunol 2024; 45:52. [PMID: 39621143 DOI: 10.1007/s10875-024-01844-0] [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: 01/23/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024]
Abstract
Hereditary pulmonary alveolar proteinosis (hPAP) is a rare lung-related primary immunodeficiency. In hPAP, variants of genes encoding the heterodimeric GM-CSF receptor alpha or beta-chains (CSF2Rα, CSF2Rβ) lead to perturbations in GM-CSF signalling. These perturbations impair the scavenging function of pulmonary alveolar macrophages leading to accumulation of surfactant proteins and lipids within the alveoli. The replacement of defective pulmonary alveolar macrophages can be achieved with allogeneic hematopoietic stem cell transplantation. However, previous reports highlight undesirable pulmonary outcomes associated with this therapeutic approach. We report a 4-year-old developmentally normal girl born of second-degree consanguineous marriage diagnosed with severe form of CSFRα-deficient PAP. She required recurrent whole lung lavage and hence was treated with allogeneic hematopoietic stem cell transplantation. A reduced toxicity treosulfan-based myeloablative regimen with alemtuzumab serotherapy was used for conditioning. Ciclosporin, mycophenolate mofetil and FAM (fluticasone inhaler, azithromycin, montelukast) were used to prevent graft-versus-host disease and immune-related complications of lung. Her post-transplant course was uneventful with full donor chimerism and complete resolution of symptoms. We demonstrate for the first time in a case of severe CSF2Rα-deficient PAP, the successful use of hematopoietic stem cell transplantation as a primary curative treatment, restoring normal lungs both anatomically and functionally. The case report provides evidence for considering allogeneic hematopoietic stem cell transplant in severe forms of CSF2R-deficient PAP.
Collapse
Affiliation(s)
- Varsha Mishra-Sopori
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, India, 400012
| | - Indu Khosla
- Department of Pediatrics, Lilavati Hospital and Research Center, Mumbai, India
| | - Sanaa Khan
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, India, 400012
| | - Darshan Kataria
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, India, 400012
| | - Pralhad Prabhudesai
- Department of Pulmonology, Lilavati Hospital and Research Center, Mumbai, India
| | - Parmarth Chandane
- Department of Pulmonology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | | | - Minnie Bodhanwala
- Department of Pediatrics, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Ambreen Pandrowala
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, India, 400012
| | - Prashant Hiwarkar
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, India, 400012.
| |
Collapse
|
6
|
Klubdaeng A, Tovichien P. Clinical approach for pulmonary alveolar proteinosis in children. World J Clin Cases 2024; 12:6339-6345. [PMID: 39464322 PMCID: PMC11438685 DOI: 10.12998/wjcc.v12.i30.6339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
In this editorial, we discuss the clinical implications of the article by Zhang et al. Pulmonary alveolar proteinosis (PAP) is a rare lung disease characterized by excessive surfactant accumulation in the alveoli. It is classified into four categories: Primary, secondary, congenital, and unclassified forms. Primary PAP is caused by the disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor signaling, which is necessary for the clearance of surfactant by alveolar macrophages. It is further divided into autoimmune PAP, caused by anti-GM-CSF antibodies blocking alveolar macrophage activation, and hereditary PAP, resulting from mutations in genes encoding GM-CSF receptors. Secondary PAP develops due to conditions affecting the number or function of alveolar macrophages, such as infections, immunodeficiency, hematological disorders, or exposure to inhaled toxins. Congenital PAP is linked to mutations in genes involved in surfactant protein production. Notably, the causes of PAP differ between children and adults. Diagnostic features include a characteristic "crazy-paving" pattern on high-resolution computed tomography, accompanied by diffuse ground-glass opacities and interlobular septal thickening. The presence of PAP can be identified by the milky appearance of bronchoalveolar lavage fluid and histological evaluation. However, these methods cannot definitively determine the cause of PAP. Whole lung lavage remains the standard treatment, often combined with specific therapies based on the underlying cause.
Collapse
Affiliation(s)
- Anuvat Klubdaeng
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Prakarn Tovichien
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
7
|
Borie R, Berteloot L, Kannengiesser C, Griese M, Cazes A, Crestani B, Hadchouel A, Debray MP. Rare genetic interstitial lung diseases: a pictorial essay. Eur Respir Rev 2024; 33:240101. [PMID: 39537246 PMCID: PMC11558537 DOI: 10.1183/16000617.0101-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024] Open
Abstract
The main monogenic causes of pulmonary fibrosis in adults are mutations in telomere-related genes. These mutations may be associated with extrapulmonary signs (hepatic, haematological and dermatological) and typically present radiologically as usual interstitial pneumonia or unclassifiable fibrosis. In children, the monogenic causes of pulmonary fibrosis are dominated by mutations in surfactant-related genes. These mutations are not associated with extrapulmonary signs and often manifest radiologically as unclassifiable fibrosis with cysts that can lead to chest wall deformities in adults. This review discusses these mutations, along with most of the monogenic causes of interstitial lung disease, including interferon-related genes, mutations in genes causing cystic lung disease, Hermansky-Pudlak syndrome, pulmonary alveolar proteinosis, lysinuric protein intolerance and lysosomal storage disorders, and their pulmonary and extrapulmonary manifestations.
Collapse
Affiliation(s)
- Raphael Borie
- Université Paris Cité, Inserm, PHERE, Paris, France
- Hôpital Bichat, APHP, Service de Pneumologie A, Centre constitutif du centre de référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | - Laureline Berteloot
- Service d'Imagerie Pédiatrique, Hôpital universitaire Necker-Enfants malades, Paris, France
- INSERM U1163, Paris, France
| | | | - Matthias Griese
- Department of Pediatric Pneumology, Dr von Hauner Children's Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Aurelie Cazes
- Département d'Anatomo-Pathologie, Hôpital Bichat, AP-HP, Paris, France
| | - Bruno Crestani
- Université Paris Cité, Inserm, PHERE, Paris, France
- Hôpital Bichat, APHP, Service de Pneumologie A, Centre constitutif du centre de référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | - Alice Hadchouel
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Université Paris Cité, Paris, France
| | - Marie Pierre Debray
- Service de Radiologie, Hopital Bichat, APHP, Université Paris Cité, Paris, France
| |
Collapse
|
8
|
Lettieri S, Bonella F, Marando VA, Franciosi AN, Corsico AG, Campo I. Pathogenesis-driven treatment of primary pulmonary alveolar proteinosis. Eur Respir Rev 2024; 33:240064. [PMID: 39142709 PMCID: PMC11322829 DOI: 10.1183/16000617.0064-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/28/2024] [Indexed: 08/16/2024] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is a syndrome that results from the accumulation of lipoproteinaceous material in the alveolar space. According to the underlying pathogenetic mechanisms, three different forms have been identified, namely primary, secondary and congenital. Primary PAP is caused by disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling due to the presence of neutralising autoantibodies (autoimmune PAP) or GM-CSF receptor genetic defects (hereditary PAP), which results in dysfunctional alveolar macrophages with reduced phagocytic clearance of particles, cholesterol and surfactant. The serum level of GM-CSF autoantibody is the only disease-specific biomarker of autoimmune PAP, although it does not correlate with disease severity. In PAP patients with normal serum GM-CSF autoantibody levels, elevated serum GM-CSF levels is highly suspicious for hereditary PAP. Several biomarkers have been correlated with disease severity, although they are not specific for PAP. These include lactate dehydrogenase, cytokeratin 19 fragment 21.1, carcinoembryonic antigen, neuron-specific enolase, surfactant proteins, Krebs von Lungen 6, chitinase-3-like protein 1 and monocyte chemotactic proteins. Finally, increased awareness of the disease mechanisms has led to the development of pathogenesis-based treatments, such as GM-CSF augmentation and cholesterol-targeting therapies.
Collapse
Affiliation(s)
- Sara Lettieri
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Francesco Bonella
- Center for interstitial and rare lung diseases, Ruhrlandklinik, University of Essen, Essen, Germany
| | | | | | - Angelo Guido Corsico
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Ilaria Campo
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
9
|
Arumugam P, Carey BC, Wikenheiser-Brokamp KA, Krischer J, Wessendarp M, Shima K, Chalk C, Stock J, Ma Y, Black D, Imbrogno M, Collins M, Kalenda Yombo DJ, Sakthivel H, Suzuki T, Lutzko C, Cancelas JA, Adams M, Hoskins E, Lowe-Daniels D, Reeves L, Kaiser A, Trapnell BC. A toxicology study of Csf2ra complementation and pulmonary macrophage transplantation therapy of hereditary PAP in mice. Mol Ther Methods Clin Dev 2024; 32:101213. [PMID: 38596536 PMCID: PMC11001781 DOI: 10.1016/j.omtm.2024.101213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 02/13/2024] [Indexed: 04/11/2024]
Abstract
Pulmonary macrophage transplantation (PMT) is a gene and cell transplantation approach in development as therapy for hereditary pulmonary alveolar proteinosis (hPAP), a surfactant accumulation disorder caused by mutations in CSF2RA/B (and murine homologs). We conducted a toxicology study of PMT of Csf2ra gene-corrected macrophages (mGM-Rα+Mϕs) or saline-control intervention in Csf2raKO or wild-type (WT) mice including single ascending dose and repeat ascending dose studies evaluating safety, tolerability, pharmacokinetics, and pharmacodynamics. Lentiviral-mediated Csf2ra cDNA transfer restored GM-CSF signaling in mGM-Rα+Mϕs. Following PMT, mGM-Rα+Mϕs engrafted, remained within the lungs, and did not undergo uncontrolled proliferation or result in bronchospasm, pulmonary function abnormalities, pulmonary or systemic inflammation, anti-transgene product antibodies, or pulmonary fibrosis. Aggressive male fighting caused a similarly low rate of serious adverse events in saline- and PMT-treated mice. Transient, minor pulmonary neutrophilia and exacerbation of pre-existing hPAP-related lymphocytosis were observed 14 days after PMT of the safety margin dose but not the target dose (5,000,000 or 500,000 mGM-Rα+Mϕs, respectively) and only in Csf2raKO mice but not in WT mice. PMT reduced lung disease severity in Csf2raKO mice. Results indicate PMT of mGM-Rα+Mϕs was safe, well tolerated, and therapeutically efficacious in Csf2raKO mice, and established a no adverse effect level and 10-fold safety margin.
Collapse
Affiliation(s)
- Paritha Arumugam
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Brenna C Carey
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
- Division of Pathology & Laboratory Medicine, CCHMC, Cincinnati, OH, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeffrey Krischer
- Departments of Pediatrics and Internal Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Matthew Wessendarp
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
| | - Kenjiro Shima
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
| | - Claudia Chalk
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
| | - Jennifer Stock
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
| | - Yan Ma
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
| | - Diane Black
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
| | - Michelle Imbrogno
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, UCMC, Cincinnati, OH, USA
| | - Margaret Collins
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, UCMC, Cincinnati, OH, USA
| | - Dan Justin Kalenda Yombo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, UCMC, Cincinnati, OH, USA
| | - Haripriya Sakthivel
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Takuji Suzuki
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
| | - Carolyn Lutzko
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
- Cell Manipulations Laboratory, CCHMC, Cincinnati, OH, USA
| | - Jose A Cancelas
- Division of Experimental Hematology, CCHMC, Cincinnati, OH, USA
| | - Michelle Adams
- Office for Clinical and Translational Research, CCHMC, Cincinnati, OH, USA
| | - Elizabeth Hoskins
- Office for Clinical and Translational Research, CCHMC, Cincinnati, OH, USA
| | | | - Lilith Reeves
- Translational Core Laboratory, CCHMC, Cincinnati, OH, USA
| | - Anne Kaiser
- Office of Research Compliance & Regulatory Affairs, CCHMC, Cincinnati, OH, USA
| | - Bruce C Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA
- Division of Pulmonary Biology, Perinatal Institute, CCHMC, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, UCMC, Cincinnati, OH, USA
- Division of Pulmonary Medicine, CCHMC, Cincinnati, OH, USA
| |
Collapse
|
10
|
O’Callaghan M, McCarthy C. Pulmonary macrophage transplant therapy in parenchymal lung diseases. Mol Ther Methods Clin Dev 2024; 32:101180. [PMID: 38249935 PMCID: PMC10797184 DOI: 10.1016/j.omtm.2023.101180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Affiliation(s)
- Marissa O’Callaghan
- Department of Respiratory Medicine, St. Vincent’s University Hospital, Dublin 4, Ireland
- School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Cormac McCarthy
- Department of Respiratory Medicine, St. Vincent’s University Hospital, Dublin 4, Ireland
- School of Medicine, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
11
|
Danhaive O, Galambos C, Lakshminrusimha S, Abman SH. Pulmonary Hypertension in Developmental Lung Diseases. Clin Perinatol 2024; 51:217-235. [PMID: 38325943 DOI: 10.1016/j.clp.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Diverse genetic developmental lung diseases can present in the neonatal period with hypoxemic respiratory failure, often associated with with pulmonary hypertension. Intractable hypoxemia and lack of sustained response to medical management should increase the suspicion of a developmental lung disorder. Genetic diagnosis and lung biopsy are helpful in establishing the diagnosis. Early diagnosis can result in optimizing management and redirecting care if needed. This article reviews normal lung development, various developmental lung disorders that can result from genetic abnormalities at each stage of lung development, their clinical presentation, management, prognosis, and differential diagnoses.
Collapse
Affiliation(s)
- Olivier Danhaive
- Division of Neonatology, Saint-Luc University Hospital, UCLouvain, Avenue Hippocrate 10, B-1200 Brussels, Belgium; Department of Pediatrics, University of California San Francisco, 530 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - Csaba Galambos
- Department of Pathology and Laboratory Medicine, University of Colorado Anschutz School of Medicine, 13001 East 17th Place, Aurora, CO 80045, USA
| | - Satyan Lakshminrusimha
- Department of Pediatrics, University of California, UC Davis Children's Hospital, 2516 Stockton Boulevard, Sacramento CA 95817, USA
| | - Steven H Abman
- Department of Pediatrics, The Pediatric Heart Lung Center, University of Colorado Anschutz Medical Campus, Mail Stop B395, 13123 East 16th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
12
|
Rodriguez Gonzalez C, Schevel H, Hansen G, Schwerk N, Lachmann N. Pulmonary Alveolar Proteinosis and new therapeutic concepts. KLINISCHE PADIATRIE 2024; 236:73-79. [PMID: 38286410 PMCID: PMC10883756 DOI: 10.1055/a-2233-1243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/15/2023] [Indexed: 01/31/2024]
Abstract
Pulmonary alveolar proteinosis (PAP) is an umbrella term used to refer to a pulmonary syndrome which is characterized by excessive accumulation of surfactant in the lungs of affected individuals. In general, PAP is a rare lung disease affecting children and adults, although its prevalence and incidence is variable among different countries. Even though PAP is a rare disease, it is a prime example on how modern medicine can lead to new therapeutic concepts, changing ways and techniques of (genetic) diagnosis which ultimately led into personalized treatments, all dedicated to improve the function of the impaired lung and thus life expectancy and quality of life in PAP patients. In fact, new technologies, such as new sequencing technologies, gene therapy approaches, new kind and sources of stem cells and completely new insights into the ontogeny of immune cells such as macrophages have increased our understanding in the onset and progression of PAP, which have paved the way for novel therapeutic concepts for PAP and beyond. As of today, classical monocyte-derived macrophages are known as important immune mediator and immune sentinels within the innate immunity. Furthermore, macrophages (known as tissue resident macrophages (TRMs)) can also be found in various tissues, introducing e. g. alveolar macrophages in the broncho-alveolar space as crucial cellular determinants in the onset of PAP and other lung disorders. Given recent insights into the onset of alveolar macrophages and knowledge about factors which impede their function, has led to the development of new therapies, which are applied in the context of PAP, with promising implications also for other diseases in which macrophages play an important role. Thus, we here summarize the latest insights into the various forms of PAP and introduce new pre-clinical work which is currently conducted in the framework of PAP, introducing new therapies for children and adults who still suffer from this severe, potentially life-threatening disease.
Collapse
Affiliation(s)
- Claudio Rodriguez Gonzalez
- Department for Pediatric Pneumology, Allergology and Neonatology,
Hannover Medical School, Hannover, Germany
| | - Hannah Schevel
- Department for Pediatric Pneumology, Allergology and Neonatology,
Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Department for Pediatric Pneumology, Allergology and Neonatology,
Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Biomedical Research in Endstage
and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625
Hannover, Germany.
| | - Nicolaus Schwerk
- Department for Pediatric Pneumology, Allergology and Neonatology,
Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Biomedical Research in Endstage
and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Nico Lachmann
- Department for Pediatric Pneumology, Allergology and Neonatology,
Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Biomedical Research in Endstage
and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625
Hannover, Germany.
- Fraunhofer Institute for Toxicology and Experimental Medicine,
Hannover, Germany
| |
Collapse
|
13
|
Neehus AL, Carey B, Landekic M, Panikulam P, Deutsch G, Ogishi M, Arango-Franco CA, Philippot Q, Modaresi M, Mohammadzadeh I, Corcini Berndt M, Rinchai D, Le Voyer T, Rosain J, Momenilandi M, Martin-Fernandez M, Khan T, Bohlen J, Han JE, Deslys A, Bernard M, Gajardo-Carrasco T, Soudée C, Le Floc'h C, Migaud M, Seeleuthner Y, Jang MS, Nikolouli E, Seyedpour S, Begueret H, Emile JF, Le Guen P, Tavazzi G, Colombo CNJ, Marzani FC, Angelini M, Trespidi F, Ghirardello S, Alipour N, Molitor A, Carapito R, Mazloomrezaei M, Rokni-Zadeh H, Changi-Ashtiani M, Brouzes C, Vargas P, Borghesi A, Lachmann N, Bahram S, Crestani B, Fayon M, Galode F, Pahari S, Schlesinger LS, Marr N, Bogunovic D, Boisson-Dupuis S, Béziat V, Abel L, Borie R, Young LR, Deterding R, Shahrooei M, Rezaei N, Parvaneh N, Craven D, Gros P, Malo D, Sepulveda FE, Nogee LM, Aladjidi N, Trapnell BC, Casanova JL, Bustamante J. Human inherited CCR2 deficiency underlies progressive polycystic lung disease. Cell 2024; 187:390-408.e23. [PMID: 38157855 PMCID: PMC10842692 DOI: 10.1016/j.cell.2023.11.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/26/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024]
Abstract
We describe a human lung disease caused by autosomal recessive, complete deficiency of the monocyte chemokine receptor C-C motif chemokine receptor 2 (CCR2). Nine children from five independent kindreds have pulmonary alveolar proteinosis (PAP), progressive polycystic lung disease, and recurrent infections, including bacillus Calmette Guérin (BCG) disease. The CCR2 variants are homozygous in six patients and compound heterozygous in three, and all are loss-of-expression and loss-of-function. They abolish CCR2-agonist chemokine C-C motif ligand 2 (CCL-2)-stimulated Ca2+ signaling in and migration of monocytic cells. All patients have high blood CCL-2 levels, providing a diagnostic test for screening children with unexplained lung or mycobacterial disease. Blood myeloid and lymphoid subsets and interferon (IFN)-γ- and granulocyte-macrophage colony-stimulating factor (GM-CSF)-mediated immunity are unaffected. CCR2-deficient monocytes and alveolar macrophage-like cells have normal gene expression profiles and functions. By contrast, alveolar macrophage counts are about half. Human complete CCR2 deficiency is a genetic etiology of PAP, polycystic lung disease, and recurrent infections caused by impaired CCL2-dependent monocyte migration to the lungs and infected tissues.
Collapse
Affiliation(s)
- Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France.
| | - Brenna Carey
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Marija Landekic
- Department of Medicine, McGill University, Montreal, QC H3G 0B1, Canada
| | - Patricia Panikulam
- Molecular Basis of Altered Immune Homeostasis, INSERM U1163, Paris Cité University, Imagine Institute, Paris 75015, France
| | - Gail Deutsch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Masato Ogishi
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Carlos A Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France; Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Mohammadreza Modaresi
- Pediatric Pulmonary and Sleep Medicine Department, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran; Pediatric Pulmonary Disease and Sleep Medicine Research Center, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Science, Tehran, Iran
| | - Iraj Mohammadzadeh
- Non-communicable Pediatric Diseases Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Melissa Corcini Berndt
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Darawan Rinchai
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris 75015, France
| | - Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Marta Martin-Fernandez
- Center for Inborn Errors of Immunity, Icahn School, New York, NY 10029, USA; Precision Immunology Institute, Icahn School, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School, New York, NY 10029, USA; Department of Pediatrics, Icahn School, New York, NY 10029, USA; Department of Microbiology, Icahn School, New York, NY 10029, USA
| | - Taushif Khan
- The Jackson Laboratory, Farmington, CT 06032, USA
| | - Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Ji Eun Han
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Alexandre Deslys
- Leukomotion Laboratory, Paris Cité University, INSERM UMR-S1151, CNRS UMR-S8253, Necker Hospital for Sick Children, Paris 75015, France
| | - Mathilde Bernard
- Leukomotion Laboratory, Paris Cité University, INSERM UMR-S1151, CNRS UMR-S8253, Necker Hospital for Sick Children, Paris 75015, France; Curie Institute, PSL Research University, CNRS, UMR144, Paris 75248, France; Pierre-Gilles de Gennes Institute, PSL Research University, Paris 75005, France
| | - Tania Gajardo-Carrasco
- Molecular Basis of Altered Immune Homeostasis, INSERM U1163, Paris Cité University, Imagine Institute, Paris 75015, France
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Corentin Le Floc'h
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France
| | - Mi-Sun Jang
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover 30625, Germany
| | - Eirini Nikolouli
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover 30625, Germany
| | - Simin Seyedpour
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran; Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hugues Begueret
- Department of Pathology, Haut-Lévèque Hospital, CHU Bordeaux, Pessac 33604, France
| | | | - Pierre Le Guen
- Pulmonology Service, Bichat Hospital, AP-HP and Paris Cité University, INSERM U1152, PHERE, Paris 75018, France
| | - Guido Tavazzi
- Department of Surgical, Pediatric, and Diagnostic Sciences, University of Pavia, Pavia 27100, Italy; Anesthesia and Intensive Care, San Matteo Research Hospital, Pavia 27100, Italy
| | - Costanza Natalia Julia Colombo
- Anesthesia and Intensive Care, San Matteo Research Hospital, Pavia 27100, Italy; Experimental Medicine, University of Pavia, Pavia 27100, Italy
| | | | - Micol Angelini
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia 27100, Italy
| | - Francesca Trespidi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia 27100, Italy
| | - Stefano Ghirardello
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia 27100, Italy
| | - Nasrin Alipour
- Molecular Immuno-Rheumatology Laboratory, INSERM UMR_S1109, GENOMAX Platform, Faculty of Medicine, OMICARE University Hospital Federation, Immunology and Hematology Research Center, Research Center in Biomedicine of Strasbourg (CRBS), Federation of Translational Medicine of Strasbourg (FMTS), University of Strasbourg, Strasbourg 67081, France; Interdisciplinary Thematic Institute (ITI) of Precision Medicine of Strasbourg, University of Strasbourg, Strasbourg 67081, France
| | - Anne Molitor
- Molecular Immuno-Rheumatology Laboratory, INSERM UMR_S1109, GENOMAX Platform, Faculty of Medicine, OMICARE University Hospital Federation, Immunology and Hematology Research Center, Research Center in Biomedicine of Strasbourg (CRBS), Federation of Translational Medicine of Strasbourg (FMTS), University of Strasbourg, Strasbourg 67081, France; Interdisciplinary Thematic Institute (ITI) of Precision Medicine of Strasbourg, University of Strasbourg, Strasbourg 67081, France
| | - Raphael Carapito
- Molecular Immuno-Rheumatology Laboratory, INSERM UMR_S1109, GENOMAX Platform, Faculty of Medicine, OMICARE University Hospital Federation, Immunology and Hematology Research Center, Research Center in Biomedicine of Strasbourg (CRBS), Federation of Translational Medicine of Strasbourg (FMTS), University of Strasbourg, Strasbourg 67081, France; Interdisciplinary Thematic Institute (ITI) of Precision Medicine of Strasbourg, University of Strasbourg, Strasbourg 67081, France; Immunology Laboratory, Biology Technical Platform, Biology Pole, New Civil Hospital, Strasbourg 67091, France
| | | | - Hassan Rokni-Zadeh
- Department of Medical Biotechnology, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Majid Changi-Ashtiani
- School of Mathematics, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Chantal Brouzes
- Laboratory of Onco-Hematology, Necker Hospital for Sick Children, Paris 75015, France
| | - Pablo Vargas
- Leukomotion Laboratory, Paris Cité University, INSERM UMR-S1151, CNRS UMR-S8253, Necker Hospital for Sick Children, Paris 75015, France; Curie Institute, PSL Research University, CNRS, UMR144, Paris 75248, France; Pierre-Gilles de Gennes Institute, PSL Research University, Paris 75005, France
| | - Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia 27100, Italy; School of Life Sciences, Swiss Federal Institute of Technology, Lausanne 1015, Switzerland
| | - Nico Lachmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover 30625, Germany; REBIRTH - Research Center for Translational Regenerative Medicine, Hannover 30625, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover 30625, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover 30625, Germany
| | - Seiamak Bahram
- Molecular Immuno-Rheumatology Laboratory, INSERM UMR_S1109, GENOMAX Platform, Faculty of Medicine, OMICARE University Hospital Federation, Immunology and Hematology Research Center, Research Center in Biomedicine of Strasbourg (CRBS), Federation of Translational Medicine of Strasbourg (FMTS), University of Strasbourg, Strasbourg 67081, France; Interdisciplinary Thematic Institute (ITI) of Precision Medicine of Strasbourg, University of Strasbourg, Strasbourg 67081, France; Immunology Laboratory, Biology Technical Platform, Biology Pole, New Civil Hospital, Strasbourg 67091, France
| | - Bruno Crestani
- Pulmonology Service, Bichat Hospital, AP-HP and Paris Cité University, INSERM U1152, PHERE, Paris 75018, France
| | - Michael Fayon
- Department of Pediatrics, Bordeaux Hospital, University of Bordeaux, 33000 Bordeaux, France; Cardiothoracic Research Center, U1045 INSERM, 33000 Bordeaux, France
| | - François Galode
- Department of Pediatrics, Bordeaux Hospital, University of Bordeaux, 33000 Bordeaux, France; Cardiothoracic Research Center, U1045 INSERM, 33000 Bordeaux, France
| | - Susanta Pahari
- Host-Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Larry S Schlesinger
- Host-Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Nico Marr
- Department of Human Immunology, Sidra Medicine, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar; Institute of Translational Immunology, Brandenburg Medical School, Brandenburg 14770, Germany
| | - Dusan Bogunovic
- Center for Inborn Errors of Immunity, Icahn School, New York, NY 10029, USA; Precision Immunology Institute, Icahn School, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School, New York, NY 10029, USA; Department of Pediatrics, Icahn School, New York, NY 10029, USA; Department of Microbiology, Icahn School, New York, NY 10029, USA
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA
| | - Raphael Borie
- Pulmonology Service, Bichat Hospital, AP-HP and Paris Cité University, INSERM U1152, PHERE, Paris 75018, France
| | - Lisa R Young
- Division of Pulmonary and Sleep Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Robin Deterding
- Pediatric Pulmonary Medicine, Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Mohammad Shahrooei
- Dr. Shahrooei Laboratory, 22 Bahman St., Ashrafi Esfahani Blvd, Tehran, Iran; Clinical and Diagnostic Immunology, KU Leuven, Leuven 3000, Belgium
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity to Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Parvaneh
- Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran
| | - Daniel Craven
- Division of Pediatric Pulmonology, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Philippe Gros
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada; Department of Biochemistry, McGill University, Montreal, QC H3A 2B4, Canada
| | - Danielle Malo
- Department of Medicine, McGill University, Montreal, QC H3G 0B1, Canada; Department of Human Genetics, McGill University, Montreal, QC H3G 0B1, Canada
| | - Fernando E Sepulveda
- Molecular Basis of Altered Immune Homeostasis, INSERM U1163, Paris Cité University, Imagine Institute, Paris 75015, France
| | - Lawrence M Nogee
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nathalie Aladjidi
- Pediatric Oncology Hematology Unit, Clinical Investigation Center (CIC), Multi-theme-CIC (CICP), University Hospital Bordeaux, Bordeaux 33000, France
| | - Bruce C Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Departments of Medicine and Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, New York, NY 10065, USA; Department of Pediatrics, Necker Hospital for Sick Children, Paris 75015, France.
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France; Paris Cité University, Imagine Institute, Paris 75015, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris 75015, France.
| |
Collapse
|
14
|
Bohlen J, Zhou Q, Philippot Q, Ogishi M, Rinchai D, Nieminen T, Seyedpour S, Parvaneh N, Rezaei N, Yazdanpanah N, Momenilandi M, Conil C, Neehus AL, Schmidt C, Arango-Franco CA, Voyer TL, Khan T, Yang R, Puchan J, Erazo L, Roiuk M, Vatovec T, Janda Z, Bagarić I, Materna M, Gervais A, Li H, Rosain J, Peel JN, Seeleuthner Y, Han JE, L'Honneur AS, Moncada-Vélez M, Martin-Fernandez M, Horesh ME, Kochetkov T, Schmidt M, AlShehri MA, Salo E, Saxen H, ElGhazali G, Yatim A, Soudée C, Sallusto F, Ensser A, Marr N, Zhang P, Bogunovic D, Cobat A, Shahrooei M, Béziat V, Abel L, Wang X, Boisson-Dupuis S, Teleman AA, Bustamante J, Zhang Q, Casanova JL. Human MCTS1-dependent translation of JAK2 is essential for IFN-γ immunity to mycobacteria. Cell 2023; 186:5114-5134.e27. [PMID: 37875108 PMCID: PMC10841658 DOI: 10.1016/j.cell.2023.09.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 08/11/2023] [Accepted: 09/22/2023] [Indexed: 10/26/2023]
Abstract
Human inherited disorders of interferon-gamma (IFN-γ) immunity underlie severe mycobacterial diseases. We report X-linked recessive MCTS1 deficiency in men with mycobacterial disease from kindreds of different ancestries (from China, Finland, Iran, and Saudi Arabia). Complete deficiency of this translation re-initiation factor impairs the translation of a subset of proteins, including the kinase JAK2 in all cell types tested, including T lymphocytes and phagocytes. JAK2 expression is sufficiently low to impair cellular responses to interleukin-23 (IL-23) and partially IL-12, but not other JAK2-dependent cytokines. Defective responses to IL-23 preferentially impair the production of IFN-γ by innate-like adaptive mucosal-associated invariant T cells (MAIT) and γδ T lymphocytes upon mycobacterial challenge. Surprisingly, the lack of MCTS1-dependent translation re-initiation and ribosome recycling seems to be otherwise physiologically redundant in these patients. These findings suggest that X-linked recessive human MCTS1 deficiency underlies isolated mycobacterial disease by impairing JAK2 translation in innate-like adaptive T lymphocytes, thereby impairing the IL-23-dependent induction of IFN-γ.
Collapse
Affiliation(s)
- Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany.
| | - Qinhua Zhou
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA; Children's Hospital of Fudan University, 201102 Shanghai, China
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Tea Nieminen
- New Children's Hospital, 00290 Helsinki, Finland
| | - Simin Seyedpour
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, P94V+8MF Tehran, Iran; Nanomedicine Research Association (NRA), P94V+8MF Tehran, Iran
| | - Nima Parvaneh
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, P94V+8MF Tehran, Iran; Department of Pediatrics, Tehran University of Medical Sciences, P94V+8MF Tehran, Iran; Children's Medical Center, P94V+8MF Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, P94V+8MF Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 1419733151 Tehran, Iran
| | - Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, P94V+8MF Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 1419733151 Tehran, Iran
| | - Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Clément Conil
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Carltin Schmidt
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA; Faculty of Medicine, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Carlos A Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Taushif Khan
- College of Health and Life Sciences, Hamad Bin Khalifa University, 8C8M+6Q Doha, Qatar; Department of Immunology, Sidra Medicine, 8C8M+6Q Doha, Qatar; The Jackson Laboratory, Farmington, CT, USA
| | - Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Julia Puchan
- Institute of Microbiology, ETH Zürich, 8049 Zürich, Switzerland
| | - Lucia Erazo
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Mykola Roiuk
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | - Taja Vatovec
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; Heidelberg University, 69120 Heidelberg, Germany
| | - Zarah Janda
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; Heidelberg University, 69120 Heidelberg, Germany
| | - Ivan Bagarić
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; Heidelberg University, 69120 Heidelberg, Germany
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Hailun Li
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Jessica N Peel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Ji Eun Han
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | | | - Marcela Moncada-Vélez
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Marta Martin-Fernandez
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School, New York, NY 10029, USA; Department of Pediatrics, Icahn School, New York, NY 10029, USA; Department of Microbiology, Icahn School, New York, NY 10029, USA
| | - Michael E Horesh
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School, New York, NY 10029, USA; Department of Pediatrics, Icahn School, New York, NY 10029, USA; Department of Microbiology, Icahn School, New York, NY 10029, USA
| | - Tatiana Kochetkov
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Monika Schmidt
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Mohammed A AlShehri
- King Fahad Medical City, Children's Specialized Hospital, 12231 Riyadh, Saudi Arabia
| | - Eeva Salo
- New Children's Hospital, 00290 Helsinki, Finland
| | - Harri Saxen
- New Children's Hospital, 00290 Helsinki, Finland
| | - Gehad ElGhazali
- Sheikh Khalifa Medical City- Union71, Purehealth, Abu Dhabi, United Arab Emirates, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Federica Sallusto
- Institute of Microbiology, ETH Zürich, 8049 Zürich, Switzerland; Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Armin Ensser
- University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Nico Marr
- College of Health and Life Sciences, Hamad Bin Khalifa University, 8C8M+6Q Doha, Qatar; Department of Immunology, Sidra Medicine, 8C8M+6Q Doha, Qatar
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Dusan Bogunovic
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School, New York, NY 10029, USA; Department of Pediatrics, Icahn School, New York, NY 10029, USA; Department of Microbiology, Icahn School, New York, NY 10029, USA
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Mohammad Shahrooei
- Clinical and Diagnostic Immunology, KU Leuven, 3000 Leuven, Belgium; Dr. Shahrooei Laboratory, 22 Bahman St., Ashrafi Esfahani Blvd, Tehran, Iran
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Xiaochuan Wang
- Children's Hospital of Fudan University, 201102 Shanghai, China
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Aurelio A Teleman
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA; Study Center for Primary Immunodeficiencies, AP-HP, Necker Hospital for Sick Children, 75015 Paris, France.
| | - Qian Zhang
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, INSERM UMR1163, Necker hospital for sick children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, New York, NY 10032, USA; Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, 75015 Paris, France.
| |
Collapse
|
15
|
Dharra R, Kumar Sharma A, Datta S. Emerging aspects of cytokine storm in COVID-19: The role of proinflammatory cytokines and therapeutic prospects. Cytokine 2023; 169:156287. [PMID: 37402337 PMCID: PMC10291296 DOI: 10.1016/j.cyto.2023.156287] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/24/2023] [Indexed: 07/06/2023]
Abstract
COVID-19 has claimed millions of lives during the last 3 years since initial cases were reported in Wuhan, China, in 2019. Patients with COVID-19 suffer from severe pneumonia, high fever, acute respiratory distress syndrome (ARDS), and multiple-organ dysfunction, which may also result in fatality in extreme cases. Cytokine storm (CS) is hyperactivation of the immune system, wherein the dysregulated production of proinflammatory cytokines could result in excessive immune cell infiltrations in the pulmonary tissues, resulting in tissue damage. The immune cell infiltration could also occur in other tissues and organs and result in multiple organs' dysfunction. The key cytokines implicated in the onset of disease severity include TNF-α, IFN-γ, IL-6, IL-1β, GM-CSF, and G-CSF. Controlling the CS is critical in treating COVID-19 disease. Therefore, different strategies are employed to mitigate the effects of CS. These include using monoclonal antibodies directed against soluble cytokines or the cytokine receptors, combination therapies, mesenchymal stem cell therapy, therapeutic plasma exchange, and some non-conventional treatment methods to improve patient immunity. The current review describes the role/s of critical cytokines in COVID-19-mediated CS and the respective treatment modalities.
Collapse
Affiliation(s)
- Renu Dharra
- CSIR-Institute of Microbial Technology, Sector 39 A, Chandigarh 160036, India
| | - Anil Kumar Sharma
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Sonal Datta
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| |
Collapse
|
16
|
Na YR, Kim SW, Seok SH. A new era of macrophage-based cell therapy. Exp Mol Med 2023; 55:1945-1954. [PMID: 37653035 PMCID: PMC10545778 DOI: 10.1038/s12276-023-01068-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 09/02/2023] Open
Abstract
Macrophages are essential innate immune cells found throughout the body that have protective and pathogenic functions in many diseases. When activated, macrophages can mediate the phagocytosis of dangerous cells or materials and participate in effective tissue regeneration by providing growth factors and anti-inflammatory molecules. Ex vivo-generated macrophages have thus been used in clinical trials as cell-based therapies, and based on their intrinsic characteristics, they outperformed stem cells within specific target diseases. In addition to the old methods of generating naïve or M2 primed macrophages, the recently developed chimeric antigen receptor-macrophages revealed the potential of genetically engineered macrophages for cell therapy. Here, we review the current developmental status of macrophage-based cell therapy. The findings of important clinical and preclinical trials are updated, and patent status is investigated. Additionally, we discuss the limitations and future directions of macrophage-based cell therapy, which will help broaden the potential utility and clinical applications of macrophages.
Collapse
Affiliation(s)
- Yi Rang Na
- Translational Immunology Laboratory, Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sang Wha Kim
- Macrophage Laboratory, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Seung Hyeok Seok
- Macrophage Laboratory, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, 110-799, South Korea.
- Department of Biomedical Sciences and Seoul National University College of Medicine, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
17
|
McLachlan G, Alton EWFW, Boyd AC, Clarke NK, Davies JC, Gill DR, Griesenbach U, Hickmott JW, Hyde SC, Miah KM, Molina CJ. Progress in Respiratory Gene Therapy. Hum Gene Ther 2022; 33:893-912. [PMID: 36074947 PMCID: PMC7615302 DOI: 10.1089/hum.2022.172] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The prospect of gene therapy for inherited and acquired respiratory disease has energized the research community since the 1980s, with cystic fibrosis, as a monogenic disorder, driving early efforts to develop effective strategies. The fact that there are still no approved gene therapy products for the lung, despite many early phase clinical trials, illustrates the scale of the challenge: In the 1990s, first-generation non-viral and viral vector systems demonstrated proof-of-concept but low efficacy. Since then, there has been steady progress toward improved vectors with the capacity to overcome at least some of the formidable barriers presented by the lung. In addition, the inclusion of features such as codon optimization and promoters providing long-term expression have improved the expression characteristics of therapeutic transgenes. Early approaches were based on gene addition, where a new DNA copy of a gene is introduced to complement a genetic mutation: however, the advent of RNA-based products that can directly express a therapeutic protein or manipulate gene expression, together with the expanding range of tools for gene editing, has stimulated the development of alternative approaches. This review discusses the range of vector systems being evaluated for lung delivery; the variety of cargoes they deliver, including DNA, antisense oligonucleotides, messenger RNA (mRNA), small interfering RNA (siRNA), and peptide nucleic acids; and exemplifies progress in selected respiratory disease indications.
Collapse
Affiliation(s)
- Gerry McLachlan
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
| | - Eric W F W Alton
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - A Christopher Boyd
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Nora K Clarke
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jane C Davies
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Deborah R Gill
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Uta Griesenbach
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jack W Hickmott
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen C Hyde
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Kamran M Miah
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Claudia Juarez Molina
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Mathew JL, Kumar K, Singhal KK, Mathew PJ, Sodhi KS, Bhatia A, Bal A, Gupta N, Menon P, Attri SV, Singh M. Protein estimation in whole lung lavage fluid in hereditary pulmonary alveolar proteinosis due to a novel GM-CSF receptor mutation. Pediatr Pulmonol 2022; 57:1802-1805. [PMID: 35438839 DOI: 10.1002/ppul.25932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/15/2022] [Accepted: 04/17/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Joseph L Mathew
- Pediatric Pulmonology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ketan Kumar
- Pediatric Pulmonology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kamal K Singhal
- Pediatric Pulmonology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Preethy J Mathew
- Department of Anesthesia and Intensive Care, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kushaljit S Sodhi
- Department of Radiodiagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anmol Bhatia
- Department of Radiodiagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amanjit Bal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nalini Gupta
- Department of Cytology and Gynaecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prema Menon
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Savita V Attri
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Meenu Singh
- Pediatric Pulmonology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
19
|
McCarthy C, Carey BC, Trapnell BC. Autoimmune Pulmonary Alveolar Proteinosis. Am J Respir Crit Care Med 2022; 205:1016-1035. [PMID: 35227171 PMCID: PMC9851473 DOI: 10.1164/rccm.202112-2742so] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/24/2022] [Indexed: 01/23/2023] Open
Abstract
Autoimmune pulmonary alveolar proteinosis (PAP) is a rare disease characterized by myeloid cell dysfunction, abnormal pulmonary surfactant accumulation, and innate immune deficiency. It has a prevalence of 7-10 per million; occurs in individuals of all races, geographic regions, sex, and socioeconomic status; and accounts for 90% of all patients with PAP syndrome. The most common presentation is dyspnea of insidious onset with or without cough, production of scant white and frothy sputum, and diffuse radiographic infiltrates in a previously healthy adult, but it can also occur in children as young as 3 years. Digital clubbing, fever, and hemoptysis are not typical, and the latter two indicate that intercurrent infection may be present. Low prevalence and nonspecific clinical, radiological, and laboratory findings commonly lead to misdiagnosis as pneumonia and substantially delay an accurate diagnosis. The clinical course, although variable, usually includes progressive hypoxemic respiratory insufficiency and, in some patients, secondary infections, pulmonary fibrosis, respiratory failure, and death. Two decades of research have raised autoimmune PAP from obscurity to a paradigm of molecular pathogenesis-based diagnostic and therapeutic development. Pathogenesis is driven by GM-CSF (granulocyte/macrophage colony-stimulating factor) autoantibodies, which are present at high concentrations in blood and tissues and form the basis of an accurate, commercially available diagnostic blood test with sensitivity and specificity of 100%. Although whole-lung lavage remains the first-line therapy, inhaled GM-CSF is a promising pharmacotherapeutic approach demonstrated in well-controlled trials to be safe, well tolerated, and efficacious. Research has established GM-CSF as a pulmonary regulatory molecule critical to surfactant homeostasis, alveolar stability, lung function, and host defense.
Collapse
Affiliation(s)
- Cormac McCarthy
- Department of Respiratory Medicine, St. Vincent’s University Hospital, Dublin, Ireland
- University College Dublin, Dublin, Ireland
| | - Brenna C. Carey
- Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio; and
- University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Bruce C. Trapnell
- Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio; and
- University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
20
|
Puel A, Bastard P, Bustamante J, Casanova JL. Human autoantibodies underlying infectious diseases. J Exp Med 2022; 219:e20211387. [PMID: 35319722 PMCID: PMC8952682 DOI: 10.1084/jem.20211387] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/14/2022] Open
Abstract
The vast interindividual clinical variability observed in any microbial infection-ranging from silent infection to lethal disease-is increasingly being explained by human genetic and immunological determinants. Autoantibodies neutralizing specific cytokines underlie the same infectious diseases as inborn errors of the corresponding cytokine or response pathway. Autoantibodies against type I IFNs underlie COVID-19 pneumonia and adverse reactions to the live attenuated yellow fever virus vaccine. Autoantibodies against type II IFN underlie severe disease caused by environmental or tuberculous mycobacteria, and other intra-macrophagic microbes. Autoantibodies against IL-17A/F and IL-6 are less common and underlie mucocutaneous candidiasis and staphylococcal diseases, respectively. Inborn errors of and autoantibodies against GM-CSF underlie pulmonary alveolar proteinosis; associated infections are less well characterized. In individual patients, autoantibodies against cytokines preexist infection with the pathogen concerned and underlie the infectious disease. Human antibody-driven autoimmunity can interfere with cytokines that are essential for protective immunity to specific infectious agents but that are otherwise redundant, thereby underlying specific infectious diseases.
Collapse
Affiliation(s)
- Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, Paris, France
| |
Collapse
|
21
|
Liu S, Cui X, Xia K, Wang D, Han J, Yao X, Liu X, Bian L, Zhang J, Li G. A Bibliometric Analysis of Pulmonary Alveolar Proteinosis From 2001 to 2021. Front Med (Lausanne) 2022; 9:846480. [PMID: 35391885 PMCID: PMC8980592 DOI: 10.3389/fmed.2022.846480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/05/2022] Open
Abstract
Background Pulmonary alveolar proteinosis (PAP) is a rare syndrome first described by Rosen et al. in 1958. Despite our considerably evolved understanding of PAP over the past decades, no bibliometric studies have been reported on this field. We aimed to analyze and visualize the research hotspots and current trends of the PAP research field using a bibliometric analysis to help understand the future development of basic and clinical research. Methods The literature regarding PAP was culled from the Web of Science Core Collection (WoSCC) database. Data were extracted from the relevant articles and visually analyzed using CiteSpace and VOSviewer software. Results Nine hundred and nine qualifying articles were included in the analysis. Publications regarding PAP increased over time. These articles mainly come from 407 institutions of 57 countries. The leading countries were the USA and Japan. University of Cincinnati (USA) and Niigata University (Japan) featured the highest number of publications among all institutions. Bruce C Trapnell exerts a significant publication impact and has made the most outstanding contributions in the field of PAP. American Journal of Physiology-Lung Cellular and Molecular Physiology was the journal with the most publications, and American Journal of Respiratory and Critical Care Medicine was the most commonly cited journal. All the top 5 co-cited journals belong to Q1. Keyword citation bursts revealed that inflammation, deficiency, tissue resident macrophage, classification, autoimmune pulmonary alveolar proteinosis, sarcoidosis, gm csf, high resolution ct, and fetal monocyte were the emerging research hotspots. Conclusion Research on PAP is prosperous. International cooperation is also expected to deepen and strengthen in the future. Our results indicated that the etiology and pathogenesis of PAP, current and emerging therapies, especially the novel pathogenesis-based options will remain research hotspots in the future.
Collapse
Affiliation(s)
- Shixu Liu
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiangning Cui
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kun Xia
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dandan Wang
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Han
- Affilated Hospital of Weifang Medical University, Weifang, China
| | - Xiaoyan Yao
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaohong Liu
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lingjie Bian
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinzhi Zhang
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangxi Li
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Guangxi Li
| |
Collapse
|
22
|
Shima K, Arumugam P, Sallese A, Horio Y, Ma Y, Trapnell C, Wessendarp M, Chalk C, McCarthy C, Carey BC, Trapnell BC, Suzuki T. A murine model of hereditary pulmonary alveolar proteinosis caused by homozygous Csf2ra gene disruption. Am J Physiol Lung Cell Mol Physiol 2022; 322:L438-L448. [PMID: 35043685 PMCID: PMC8917935 DOI: 10.1152/ajplung.00175.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hereditary pulmonary alveolar proteinosis (hPAP) is a rare disorder caused by recessive mutations in GM-CSF receptor subunit α/β genes (CSF2RA/CSF2RB, respectively) characterized by impaired GM-CSF-dependent surfactant clearance by alveolar macrophages (AMs) resulting in alveolar surfactant accumulation and hypoxemic respiratory failure. Because hPAP is caused by CSF2RA mutations in most patients, we created an animal model of hPAP caused by Csf2ra gene disruption (Csf2ra-/- mice) and evaluated the effects on AMs and lungs. Macrophages from Csf2ra-/- mice were unable to bind and clear GM-CSF, did not exhibit GM-CSF signaling, and had functional defects in phagocytosis, cholesterol clearance, and surfactant clearance. Csf2ra-/- mice developed a time-dependent, progressive lung disease similar to hPAP in children caused by CSF2RA mutations with respect to the clinical, physiological, histopathological, biochemical abnormalities, biomarkers of PAP lung disease, and clinical course. In contrast, Csf2ra+/- mice had functionally normal AMs and no lung disease. Pulmonary macrophage transplantation (PMT) without myeloablation resulted in long-term engraftment, restoration of GM-CSF responsiveness to AMs, and a safe and durable treatment effect that lasted for the duration of the experiment (6 mo). Results demonstrate that homozygous (but not heterozygous) Csf2ra gene ablation caused hPAP identical to hPAP in children with CSF2RA mutations, identified AMs as the cellular site of hPAP pathogenesis in Csf2ra-/- mice, and have implications for preclinical studies supporting the translation of PMT as therapy of hPAP in humans.
Collapse
Affiliation(s)
- Kenjiro Shima
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Paritha Arumugam
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Anthony Sallese
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Yuko Horio
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Yan Ma
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Cole Trapnell
- 2Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Matthew Wessendarp
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Claudia Chalk
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Cormac McCarthy
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio,4Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Brenna C. Carey
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Bruce C. Trapnell
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio,3Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio,4Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Takuji Suzuki
- 1Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio,5Department of Respirology, Chiba University, Chiba, Japan
| |
Collapse
|
23
|
Wessendarp M, Watanabe-Chailland M, Liu S, Stankiewicz T, Ma Y, Kasam RK, Shima K, Chalk C, Carey B, Rosendale LR, Dominique Filippi M, Arumugam P. Role of GM-CSF in regulating metabolism and mitochondrial functions critical to macrophage proliferation. Mitochondrion 2021; 62:85-101. [PMID: 34740864 DOI: 10.1016/j.mito.2021.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 10/14/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) exerts pleiotropic effects on macrophages and is required for self-renewal but the mechanisms responsible are unknown. Using mouse models with disrupted GM-CSF signaling, we show GM-CSF is critical for mitochondrial turnover, functions, and integrity. GM-CSF signaling is essential for fatty acid β-oxidation and markedly increased tricarboxylic acid cycle activity, oxidative phosphorylation, and ATP production. GM-CSF also regulated cytosolic pathways including glycolysis, pentose phosphate pathway, and amino acid synthesis. We conclude that GM-CSF regulates macrophages in part through a critical role in maintaining mitochondria, which are necessary for cellular metabolism as well as proliferation and self-renewal.
Collapse
Affiliation(s)
- Matthew Wessendarp
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | | | - Serena Liu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Yan Ma
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | | | - Kenjiro Shima
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | - Claudia Chalk
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | - Brenna Carey
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | | | | | - Paritha Arumugam
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA.
| |
Collapse
|
24
|
Lazarus HM, Ragsdale CE, Gale RP, Lyman GH. Sargramostim (rhu GM-CSF) as Cancer Therapy (Systematic Review) and An Immunomodulator. A Drug Before Its Time? Front Immunol 2021; 12:706186. [PMID: 34484202 PMCID: PMC8416151 DOI: 10.3389/fimmu.2021.706186] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/26/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Sargramostim [recombinant human granulocyte-macrophage colony-stimulating factor (rhu GM-CSF)] was approved by US FDA in 1991 to accelerate bone marrow recovery in diverse settings of bone marrow failure and is designated on the list of FDA Essential Medicines, Medical Countermeasures, and Critical Inputs. Other important biological activities including accelerating tissue repair and modulating host immunity to infection and cancer via the innate and adaptive immune systems are reported in pre-clinical models but incompletely studied in humans. OBJECTIVE Assess safety and efficacy of sargramostim in cancer and other diverse experimental and clinical settings. METHODS AND RESULTS We systematically reviewed PubMed, Cochrane and TRIP databases for clinical data on sargramostim in cancer. In a variety of settings, sargramostim after exposure to bone marrow-suppressing agents accelerated hematologic recovery resulting in fewer infections, less therapy-related toxicity and sometimes improved survival. As an immune modulator, sargramostim also enhanced anti-cancer responses in solid cancers when combined with conventional therapies, for example with immune checkpoint inhibitors and monoclonal antibodies. CONCLUSIONS Sargramostim accelerates hematologic recovery in diverse clinical settings and enhances anti-cancer responses with a favorable safety profile. Uses other than in hematologic recovery are less-well studied; more data are needed on immune-enhancing benefits. We envision significantly expanded use of sargramostim in varied immune settings. Sargramostim has the potential to reverse the immune suppression associated with sepsis, trauma, acute respiratory distress syndrome (ARDS) and COVID-19. Further, sargramostim therapy has been promising in the adjuvant setting with vaccines and for anti-microbial-resistant infections and treating autoimmune pulmonary alveolar proteinosis and gastrointestinal, peripheral arterial and neuro-inflammatory diseases. It also may be useful as an adjuvant in anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Hillard M. Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | | | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Gary H. Lyman
- Public Health Sciences and Clinical Research Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
25
|
Karolak JA, Gambin T, Szafranski P, Maywald RL, Popek E, Heaney JD, Stankiewicz P. Perturbation of semaphorin and VEGF signaling in ACDMPV lungs due to FOXF1 deficiency. Respir Res 2021; 22:212. [PMID: 34315444 PMCID: PMC8314029 DOI: 10.1186/s12931-021-01797-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/01/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a rare lethal congenital lung disorder in neonates characterized by severe progressive respiratory failure and refractory pulmonary hypertension, resulting from underdevelopment of the peripheral pulmonary tree. Causative heterozygous single nucleotide variants (SNVs) or copy-number variant (CNV) deletions involving FOXF1 or its distant lung-specific enhancer on chromosome 16q24.1 have been identified in 80-90% of ACDMPV patients. FOXF1 maps closely to and regulates the oppositely oriented FENDRR, with which it also shares regulatory elements. METHODS To better understand the transcriptional networks downstream of FOXF1 that are relevant for lung organogenesis, using RNA-seq, we have examined lung transcriptomes in 12 histopathologically verified ACDMPV patients with or without pathogenic variants in the FOXF1 locus and analyzed gene expression profile in FENDRR-depleted fetal lung fibroblasts, IMR-90. RESULTS RNA-seq analyses in ACDMPV neonates revealed changes in the expression of several genes, including semaphorins (SEMAs), neuropilin 1 (NRP1), and plexins (PLXNs), essential for both epithelial branching and vascular patterning. In addition, we have found deregulation of the vascular endothelial growth factor (VEGF) signaling that also controls pulmonary vasculogenesis and a lung-specific endothelial gene TMEM100 known to be essential in vascular morphogenesis. Interestingly, we have observed a substantial difference in gene expression profiles between the ACDMPV samples with different types of FOXF1 defect. Moreover, partial overlap between transcriptome profiles of ACDMPV lungs with FOXF1 SNVs and FENDRR-depleted IMR-90 cells suggests contribution of FENDRR to ACDMPV etiology. CONCLUSIONS Our transcriptomic data imply potential crosstalk between several lung developmental pathways, including interactions between FOXF1-SHH and SEMA-NRP or VEGF/VEGFR2 signaling, and provide further insight into complexity of lung organogenesis in humans.
Collapse
Affiliation(s)
- Justyna A Karolak
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-781, Poznań, Poland
| | - Tomasz Gambin
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Institute of Computer Science, Warsaw University of Technology, 00-665, Warsaw, Poland
| | - Przemyslaw Szafranski
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Rebecca L Maywald
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Edwina Popek
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Paweł Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
Alasiri AM, Alasbali RA, Alaqil MA, Alahmari AM, Alshamrani ND, Badri RN. Autoimmune pulmonary alveolar proteinosis successfully treated with lung lavage in an adolescent patient: a case report. J Med Case Rep 2021; 15:340. [PMID: 34238362 PMCID: PMC8268574 DOI: 10.1186/s13256-021-02906-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/13/2021] [Indexed: 12/23/2022] Open
Abstract
Background Pulmonary alveolar proteinosis is a rare interstitial lung disease characterized by accumulating surfactant materials in the alveoli. The autoimmune form is by far the most common in adults, while in the pediatric age group, the vast majority of cases are congenital. We report a case of an adolescent patient diagnosed with autoimmune pulmonary alveolar proteinosis, which is unusual in this age group. Case presentation A-15 year-old Saudi male presented to the emergency department with a history of shortness of breath and low oxygen saturation. High-resolution computed tomography of his chest showed a global crazy-paving pattern. Autoantibodies against granulocyte-macrophage colony-stimulating factor were detected in his serum. A diagnosis of the autoimmune form of pulmonary alveolar proteinosis was confirmed after excluding other possible causes. The patient improved after he underwent whole lung lavage under general anesthesia, and he was independent of oxygen therapy after 6 months of follow-up. Conclusion The autoimmune form of pulmonary alveolar proteinosis is rare in the pediatric age group and should be considered when no apparent cause of this disease was found. Whole lung lavage should be the first treatment modality offered in this setting with close follow-up and monitoring.
Collapse
Affiliation(s)
| | | | - Meaad Ali Alaqil
- Department of internal medicine, Aseer Central Hospital, Abha, Saudi Arabia
| | | | | | - Rabab Nasir Badri
- Department of histopathology, Aseer Central Hospital, Abha, Saudi Arabia
| |
Collapse
|
27
|
Wu XK, Lin Q. Pulmonary alveolar proteinosis complicated with nocardiosis: A case report and review of the literature. World J Clin Cases 2021; 9:2874-2883. [PMID: 33969072 PMCID: PMC8058687 DOI: 10.12998/wjcc.v9.i12.2874] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/26/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pulmonary alveolar proteinosis (PAP) is a pulmonary syndrome wherein large volumes of phospholipid and protein-rich surfactants accumulate within the alveoli. PAP forms include primary (auto-immune PAP), secondary, and congenital. Nocardiosis is a form of suppurative disease induced upon infection with bacteria of the Nocardia genus. Clinically, cases of PAP complicated with Nocardia infections are rare, regardless of form. Unfortunately, as such, they are easily overlooked or misdiagnosed. We describe, here, the case of a patient suffering from simultaneous primary PAP and nocardiosis.
CASE SUMMARY A 45-year-old Chinese man, without history of relevant disease, was admitted to our hospital on August 8, 2018 to address complaints of activity-related respiratory exertion and cough lasting over 6 mo. Lung computed tomography (CT) revealed diffuse bilateral lung infiltration with local consolidation in the middle right lung lobe. Subsequent transbronchial lung biopsy and CT-guided lung biopsy led to a diagnosis of primary PAP (granulocyte-macrophage colony-stimulating factor antibody-positive) complicated with nocardiosis (periodic acid-Schiff-positive). After a 6 mo course of anti-infective treatment (sul-famethoxazole), the lesion was completely absorbed, such that only fibrous foci remained, and the patient exhibited significant symptom improvement. Follow-up also showed improvement in pulmonary function and the CT imaging findings of PAP. No whole-lung lavage has been conducted to date. This case highlights that active anti-nocardia treatment may effectively improve the symptoms and alleviate PAP in patients with PAP and nocardia, possibly reducing the need for whole-lung lavage.
CONCLUSION When evaluating patients presenting with PAP and pulmonary infections, the potential for nocardiosis should be considered.
Collapse
Affiliation(s)
- Xiao-Kang Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang Province, China
| | - Quan Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang Province, China
| |
Collapse
|
28
|
Hetzel M, Ackermann M, Lachmann N. Beyond "Big Eaters": The Versatile Role of Alveolar Macrophages in Health and Disease. Int J Mol Sci 2021; 22:3308. [PMID: 33804918 PMCID: PMC8036607 DOI: 10.3390/ijms22073308] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023] Open
Abstract
Macrophages act as immune scavengers and are important cell types in the homeostasis of various tissues. Given the multiple roles of macrophages, these cells can also be found as tissue resident macrophages tightly integrated into a variety of tissues in which they fulfill crucial and organ-specific functions. The lung harbors at least two macrophage populations: interstitial and alveolar macrophages, which occupy different niches and functions. In this review, we provide the latest insights into the multiple roles of alveolar macrophages while unraveling the distinct factors which can influence the ontogeny and function of these cells. Furthermore, we will highlight pulmonary diseases, which are associated with dysfunctional macrophages, concentrating on congenital diseases as well as pulmonary infections and impairment of immunological pathways. Moreover, we will provide an overview about different treatment approaches targeting macrophage dysfunction. Improved knowledge of the role of macrophages in the onset of pulmonary diseases may provide the basis for new pharmacological and/or cell-based immunotherapies and will extend our understanding to other macrophage-related disorders.
Collapse
Affiliation(s)
- Miriam Hetzel
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.H.); (M.A.)
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Mania Ackermann
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.H.); (M.A.)
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Nico Lachmann
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
29
|
Genetic Testing for Neonatal Respiratory Disease. CHILDREN-BASEL 2021; 8:children8030216. [PMID: 33799761 PMCID: PMC8001923 DOI: 10.3390/children8030216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/27/2021] [Accepted: 03/08/2021] [Indexed: 11/16/2022]
Abstract
Genetic mechanisms are now recognized as rare causes of neonatal lung disease. Genes potentially responsible for neonatal lung disease include those encoding proteins important in surfactant function and metabolism, transcription factors important in lung development, proteins involved in ciliary assembly and function, and various other structural and immune regulation genes. The phenotypes of infants with genetic causes of neonatal lung disease may have some features that are difficult to distinguish clinically from more common, reversible causes of lung disease, and from each other. Multigene panels are now available that can allow for a specific diagnosis, providing important information for treatment and prognosis. This review discusses genes in which abnormalities are known to cause neonatal lung disease and their associated phenotypes, and advantages and limitations of genetic testing.
Collapse
|
30
|
Goussard P, Pohunek P, Eber E, Midulla F, Di Mattia G, Merven M, Janson JT. Pediatric bronchoscopy: recent advances and clinical challenges. Expert Rev Respir Med 2021; 15:453-475. [PMID: 33512252 DOI: 10.1080/17476348.2021.1882854] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: During the last 40 years equipment has been improved with smaller instruments and sufficient size working channels. This has ensured that bronchoscopy offers therapeutic and interventional options.Areas covered: We provide a review of recent advances and clinical challenges in pediatric bronchoscopy. This includes single-use bronchoscopes, endobronchial ultrasound, and cryoprobe. Bronchoscopy in persistent preschool wheezing and asthma is included. The indications for interventional bronchoscopy have amplified and included balloon dilatation, endoscopic intubation, the use of airway stents, whole lung lavage, closing of fistulas and air leak, as well as an update on removal of foreign bodies. Others include the use of laser and microdebrider in airway surgery. Experience with bronchoscope during the COVID-19 pandemic has been included in this review. PubMed was searched for articles on pediatric bronchoscopy, including rigid bronchoscopy as well as interventional bronchoscopy with a focus on reviewing literature in the past 5 years.Expert opinion: As the proficiency of pediatric interventional pulmonologists continues to grow more interventions are being performed. There is a scarcity of published evidence in this field. Courses for pediatric interventional bronchoscopy need to be developed. The COVID-19 experience resulted in safer bronchoscopy practice for all involved.
Collapse
Affiliation(s)
- P Goussard
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - P Pohunek
- Division of Pediatric Respiratory Diseases, Pediatric Department, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - E Eber
- Department of Paediatrics and Adolescent Medicine, Head, Division of Paediatric Pulmonology and Allergology, Medical University of Graz, Graz, Austria
| | - F Midulla
- Department of Maternal Infantile and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - G Di Mattia
- Department of Maternal Infantile and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - M Merven
- Department Otorhinolaryngology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - J T Janson
- Department of Surgical Sciences, Division of Cardio-Thoracic Surgery, Stellenbosch University, and Tygerberg Hospital, Tygerberg, South Africa
| |
Collapse
|
31
|
|
32
|
Hadchouel A, Drummond D, Abou Taam R, Lebourgeois M, Delacourt C, de Blic J. Alveolar proteinosis of genetic origins. Eur Respir Rev 2020; 29:29/158/190187. [PMID: 33115790 DOI: 10.1183/16000617.0187-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/21/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is a rare form of chronic interstitial lung disease, characterised by the intra-alveolar accumulation of lipoproteinaceous material. Numerous conditions can lead to its development. Whereas the autoimmune type is the main cause in adults, genetic defects account for a large part of cases in infants and children. Even if associated extra-respiratory signs may guide the clinician during diagnostic work-up, next-generation sequencing panels represent an efficient diagnostic tool. Exome sequencing also allowed the discovery of new variants and genes involved in PAP. The aim of this article is to summarise our current knowledge of genetic causes of PAP.
Collapse
Affiliation(s)
- Alice Hadchouel
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France .,INSERM U1151, Institut Necker Enfants Malades, Paris, France.,Université de Paris, Faculté de Médecine, Paris, France
| | - David Drummond
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France
| | - Rola Abou Taam
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France
| | - Muriel Lebourgeois
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France
| | - Christophe Delacourt
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,INSERM U1151, Institut Necker Enfants Malades, Paris, France.,Université de Paris, Faculté de Médecine, Paris, France
| | - Jacques de Blic
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France
| |
Collapse
|
33
|
Mata-Suarez SM, Castro-Lalín A, Mc Loughlin S, De Domini J, Bianco JC. Whole-Lung Lavage-a Narrative Review of Anesthetic Management. J Cardiothorac Vasc Anesth 2020; 36:587-593. [PMID: 33386193 DOI: 10.1053/j.jvca.2020.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 01/15/2023]
Abstract
Pulmonary alveolar proteinosis is a rare disease characterized by progressive accumulation of lipoprotein material in the alveoli as a result of a dysfunction in surfactant clearance. The whole-lung lavage procedure is considered the current standard of care and consists of the sequential lavage of both lungs for mechanical removal of residual material in the alveoli. However, a lack of standardization has resulted in different procedural techniques among institutions. Even though whole-lung lavage is considered to be a safe procedure, unforeseen complications might occur, and proper knowledge of physiologic implications may allow clinicians to establish the appropriate therapy. This review provides an insight into the underlying physiology of the disease, the technical details of the procedure from an anesthesiologist's perspective, and discussion of potential intraoperative complications.
Collapse
Affiliation(s)
- Santiago M Mata-Suarez
- Department of Anesthesiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina.
| | - Agustina Castro-Lalín
- Department of Anesthesiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Santiago Mc Loughlin
- Department of Anesthesiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Juan De Domini
- Department of Anesthesiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Juan C Bianco
- Department of Anesthesiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
34
|
Abstract
Pulmonary alveolar proteinosis (PAP) is a respiratory pathology characterized by the accumulation and increase of surfactant-derived material in the lungs. In clinical practice, PAP may present as the primary form, which includes autoimmune and hereditary PAP, or as the secondary form. Diffuse alveolar radiopacities on chest x-ray and the crazy-paving pattern on high-resolution computed tomography are important, although not specific findings for PAP. Bronchoalveolar lavage biopsy is a diagnostic method, and whole-lung lavage remains the criterion standard for the treatment of PAP. Evidence is required regarding treatment with exogenous anti-granulocyte/macrophage colony-stimulating factor.Here, we present a 13-year-old male patient with hereditary PAP and a 15-year-old female patient with autoimmune PAP who presented with complaints of easy fatigability and weakness to emphasize the importance of keeping in mind PAP as a differential diagnosis in patients with respiratory failure findings.
Collapse
|
35
|
Abstract
Pulmonary alveolar proteinosis (PAP) is a rare respiratory syndrome characterised by the accumulation of surfactant lipoproteins within the alveoli. According to various pathogenetic mechanisms and aetiologies, PAP is classified as primary, secondary or congenital. Primary PAP is led by a granulocyte–macrophage colony-stimulating factor (GM-CSF) signalling disruption; the autoimmune form is driven by the presence of anti GM-CSF autoantibodies and represents 90% of all the PAP cases; and the hereditary form is the result of mutations in genes encoding GM-CSF receptor. Secondary PAP is associated with various diseases causing a reduction in function and/or number of alveolar macrophages. Congenital PAP emerges as a consequence of corrupted surfactant production, due to mutations in surfactant proteins or lipid transporter, or mutations affecting lung development. The clinical manifestations are various, ranging from insidious onset to acute or progressive respiratory failure, including premature death within the first days of life in neonates with congenital surfactant production disorders. The diagnostic workup includes clinical and radiological assessment (respiratory function test, high-resolution chest computed tomography), laboratory tests (anti-GM-CSF autoantibodies dosage, GM-CSF serum level and GM-CSF signalling test), and genetic tests. Whole-lung lavage is the current gold standard of care of PAP; however, the therapeutic approach depends on the pathogenic form and disease severity, including GM-CSF augmentation strategies in autoimmune PAP and other promising new treatments. A concise educational review of pulmonary alveolar proteinosis (PAP), a rare respiratory syndrome with various and heterogeneous aetiologies, caused by the impairment of pulmonary surfactant clearance or by abnormal surfactant productionhttps://bit.ly/3aFpQm9
Collapse
Affiliation(s)
| | - Ilaria Campo
- Pneumology Unit, IRCCS Policlinico San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
36
|
Bonaventura A, Vecchié A, Wang TS, Lee E, Cremer PC, Carey B, Rajendram P, Hudock KM, Korbee L, Van Tassell BW, Dagna L, Abbate A. Targeting GM-CSF in COVID-19 Pneumonia: Rationale and Strategies. Front Immunol 2020; 11:1625. [PMID: 32719685 PMCID: PMC7348297 DOI: 10.3389/fimmu.2020.01625] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/17/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a clinical syndrome ranging from mild symptoms to severe pneumonia that often leads to respiratory failure, need for mechanical ventilation, and death. Most of the lung damage is driven by a surge in inflammatory cytokines [interleukin-6, interferon-γ, and granulocyte-monocyte stimulating factor (GM-CSF)]. Blunting this hyperinflammation with immunomodulation may lead to clinical improvement. GM-CSF is produced by many cells, including macrophages and T-cells. GM-CSF-derived signals are involved in differentiation of macrophages, including alveolar macrophages (AMs). In animal models of respiratory infections, the intranasal administration of GM-CSF increased the proliferation of AMs and improved outcomes. Increased levels of GM-CSF have been recently described in patients with COVID-19 compared to healthy controls. While GM-CSF might be beneficial in some circumstances as an appropriate response, in this case the inflammatory response is maladaptive by virtue of being later and disproportionate. The inhibition of GM-CSF signaling may be beneficial in improving the hyperinflammation-related lung damage in the most severe cases of COVID-19. This blockade can be achieved through antagonism of the GM-CSF receptor or the direct binding of circulating GM-CSF. Initial findings from patients with COVID-19 treated with a single intravenous dose of mavrilimumab, a monoclonal antibody binding GM-CSF receptor α, showed oxygenation improvement and shorter hospitalization. Prospective, randomized, placebo-controlled trials are ongoing. Anti-GM-CSF monoclonal antibodies, TJ003234 and gimsilumab, will be tested in clinical trials in patients with COVID-19, while lenzilumab received FDA approval for compassionate use. These trials will help inform whether blunting the inflammatory signaling provided by the GM-CSF axis in COVID-19 is beneficial.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/therapeutic use
- Betacoronavirus/immunology
- COVID-19
- Coronavirus Infections/drug therapy
- Coronavirus Infections/immunology
- Coronavirus Infections/pathology
- Disease Models, Animal
- Drug Delivery Systems
- Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Humans
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/pathology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/pathology
- Pandemics
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/immunology
- Pneumonia, Viral/pathology
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- SARS-CoV-2
- Signal Transduction/drug effects
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Aldo Bonaventura
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, United States
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alessandra Vecchié
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, United States
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Tisha S. Wang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, United States
| | - Elinor Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, United States
| | - Paul C. Cremer
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brenna Carey
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | | | - Kristin M. Hudock
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, OH, United States
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Leslie Korbee
- Academic Regulatory & Monitoring Services, LLC, Cincinnati, OH, United States
| | - Benjamin W. Van Tassell
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, United States
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Abbate
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, United States
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
37
|
Gros M, Aissat A, Perez‐Martin S, Abou Taam R, Funalot B, Fanen P, Epaud R, Becdelievre A. Interstitial lung disease reveals 48,XXYY syndrome in a child. Acta Paediatr 2020; 109:1060-1061. [PMID: 31721284 DOI: 10.1111/apa.15090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Marion Gros
- Service de Pédiatrie Centre Intercommunal de Créteil Créteil France
| | - Abdel Aissat
- Laboratoire de Génétique Hôpital Henri Mondor AP‐HP Créteil France
- Inserm U955 Equipe 5 Créteil France
- DHU Ageing Thorax Vessel Blood Créteil France
- Faculté de Médecine Université Paris‐Est Créteil Créteil France
| | | | - Rola Abou Taam
- Service de pneumologie pédiatrique AP‐HP Hôpital Necker Enfants‐Malades Paris France
| | - Benoit Funalot
- Laboratoire de Génétique Hôpital Henri Mondor AP‐HP Créteil France
- Faculté de Médecine Université Paris‐Est Créteil Créteil France
| | - Pascale Fanen
- Laboratoire de Génétique Hôpital Henri Mondor AP‐HP Créteil France
- Inserm U955 Equipe 5 Créteil France
- DHU Ageing Thorax Vessel Blood Créteil France
- Faculté de Médecine Université Paris‐Est Créteil Créteil France
- Centre des Maladies Respiratoires Rare Respirare® Créteil France
| | - Ralph Epaud
- Service de Pédiatrie Centre Intercommunal de Créteil Créteil France
- Inserm U955 Equipe 5 Créteil France
- DHU Ageing Thorax Vessel Blood Créteil France
- Faculté de Médecine Université Paris‐Est Créteil Créteil France
- Centre des Maladies Respiratoires Rare Respirare® Créteil France
| | - Alix Becdelievre
- Laboratoire de Génétique Hôpital Henri Mondor AP‐HP Créteil France
- Inserm U955 Equipe 5 Créteil France
- DHU Ageing Thorax Vessel Blood Créteil France
- Centre des Maladies Respiratoires Rare Respirare® Créteil France
| |
Collapse
|
38
|
Suzuki T, McCarthy C, Carey BC, Borchers M, Beck D, Wikenheiser-Brokamp KA, Black D, Chalk C, Trapnell BC. Increased Pulmonary GM-CSF Causes Alveolar Macrophage Accumulation. Mechanistic Implications for Desquamative Interstitial Pneumonitis. Am J Respir Cell Mol Biol 2020; 62:87-94. [PMID: 31310562 PMCID: PMC6938130 DOI: 10.1165/rcmb.2018-0294oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 05/14/2019] [Indexed: 12/11/2022] Open
Abstract
Desquamative interstitial pneumonia (DIP) is a rare, smoking-related, diffuse parenchymal lung disease characterized by marked accumulation of alveolar macrophages (AMs) and emphysema, without extensive fibrosis or neutrophilic inflammation. Because smoking increases expression of pulmonary GM-CSF (granulocyte/macrophage-colony stimulating factor) and GM-CSF stimulates proliferation and activation of AMs, we hypothesized that chronic exposure of mice to increased pulmonary GM-CSF may recapitulate DIP. Wild-type (WT) mice were subjected to inhaled cigarette smoke exposure for 16 months, and AM numbers and pulmonary GM-CSF mRNA levels were measured. After demonstrating that smoke inhalation increased pulmonary GM-CSF in WT mice, transgenic mice overexpressing pulmonary GM-CSF (SPC-GM-CSF+/+) were used to determine the effects of chronic exposure to increased pulmonary GM-CSF (without smoke inhalation) on accumulation and activation of AMs, pulmonary matrix metalloproteinase (MMP) expression and activity, lung histopathology, development of polycythemia, and survival. In WT mice, smoke exposure markedly increased pulmonary GM-CSF and AM accumulation. In unexposed SPC-GM-CSF+/+ mice, AMs were spontaneously activated as shown by phosphorylation of STAT5 (signal inducer and activator of transcription 5) and accumulated progressively with involvement of 84% (interquartile range, 55-90%) of the lung parenchyma by 10 months of age. Histopathologic features also included scattered multinucleated giant cells, alveolar epithelial cell hyperplasia, and mild alveolar wall thickening. SPC-GM-CSF+/+ mice had increased pulmonary MMP-9 and MMP-12 levels, spontaneously developed emphysema and secondary polycythemia, and had increased mortality compared with WT mice. Results show cigarette smoke increased pulmonary GM-CSF and AM proliferation, and chronically increased pulmonary GM-CSF recapitulated the cardinal features of DIP, including AM accumulation, emphysema, secondary polycythemia, and increased mortality in mice. These observations suggest pulmonary GM-CSF may be involved in the pathogenesis of DIP.
Collapse
Affiliation(s)
- Takuji Suzuki
- Translational Pulmonary Science Center
- Division of Pulmonary Biology
| | - Cormac McCarthy
- Translational Pulmonary Science Center
- Division of Pulmonary Biology
- Division of Pulmonary Medicine, and
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Brenna C. Carey
- Translational Pulmonary Science Center
- Division of Pulmonary Biology
| | - Michael Borchers
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David Beck
- Translational Pulmonary Science Center
- Division of Pulmonary Biology
| | - Kathryn A. Wikenheiser-Brokamp
- Division of Pulmonary Biology
- Division of Pathology and Laboratory Medicine, Children’s Hospital Medical Center, Cincinnati, Ohio; and
| | - Dianna Black
- Translational Pulmonary Science Center
- Division of Pulmonary Biology
| | - Claudia Chalk
- Translational Pulmonary Science Center
- Division of Pulmonary Biology
| | - Bruce C. Trapnell
- Translational Pulmonary Science Center
- Division of Pulmonary Biology
- Division of Pulmonary Medicine, and
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
39
|
Happle C, Lachmann N, Ackermann M, Mirenska A, Göhring G, Thomay K, Mucci A, Hetzel M, Glomb T, Suzuki T, Chalk C, Glage S, Dittrich-Breiholz O, Trapnell B, Moritz T, Hansen G. Pulmonary Transplantation of Human Induced Pluripotent Stem Cell-derived Macrophages Ameliorates Pulmonary Alveolar Proteinosis. Am J Respir Crit Care Med 2019; 198:350-360. [PMID: 29652170 DOI: 10.1164/rccm.201708-1562oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Although the transplantation of induced pluripotent stem cell (iPSC)-derived cells harbors enormous potential for the treatment of pulmonary diseases, in vivo data demonstrating clear therapeutic benefits of human iPSC-derived cells in lung disease models are missing. OBJECTIVES We have tested the therapeutic potential of iPSC-derived macrophages in a humanized disease model of hereditary pulmonary alveolar proteinosis (PAP). Hereditary PAP is caused by a genetic defect of the GM-CSF (granulocyte-macrophage colony-stimulating factor) receptor, which leads to disturbed macrophage differentiation and protein/surfactant degradation in the lungs, subsequently resulting in severe respiratory insufficiency. METHODS Macrophages derived from human iPSCs underwent intrapulmonary transplantation into humanized PAP mice, and engraftment, in vivo differentiation, and therapeutic efficacy of the transplanted cells were analyzed. MEASUREMENTS AND MAIN RESULTS On intratracheal application, iPSC-derived macrophages engrafted in the lungs of humanized PAP mice. After 2 months, transplanted cells displayed the typical morphology, surface markers, functionality, and transcription profile of primary human alveolar macrophages. Alveolar proteinosis was significantly reduced as demonstrated by diminished protein content and surfactant protein D levels, decreased turbidity of the BAL fluid, and reduced surfactant deposition in the lungs of transplanted mice. CONCLUSIONS We here demonstrate for the first time that pulmonary transplantation of human iPSC-derived macrophages leads to pulmonary engraftment, their in situ differentiation to an alveolar macrophage phenotype, and a reduction of alveolar proteinosis in a humanized PAP model. To our knowledge, this finding presents the first proof-of-concept for the therapeutic potential of human iPSC-derived cells in a pulmonary disease and may have profound implications beyond the rare disease of PAP.
Collapse
Affiliation(s)
- Christine Happle
- 1 Department of Pediatric Pneumology, Allergology and Neonatology.,2 Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL)
| | - Nico Lachmann
- 3 Junior Research Group (JRG) Translational Hematology of Congenital Diseases, Regenerative Biology and Reconstructive Therapies (REBIRTH) Cluster of Excellence.,4 Institute of Experimental Hematology
| | - Mania Ackermann
- 3 Junior Research Group (JRG) Translational Hematology of Congenital Diseases, Regenerative Biology and Reconstructive Therapies (REBIRTH) Cluster of Excellence.,4 Institute of Experimental Hematology
| | - Anja Mirenska
- 1 Department of Pediatric Pneumology, Allergology and Neonatology
| | | | | | - Adele Mucci
- 4 Institute of Experimental Hematology.,6 Research Group-Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence
| | - Miriam Hetzel
- 4 Institute of Experimental Hematology.,6 Research Group-Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence
| | - Torsten Glomb
- 7 Core Unit Transcriptomics, Institute for Physiological Chemistry, and
| | - Takuji Suzuki
- 8 Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Claudia Chalk
- 8 Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Silke Glage
- 9 Central Animal Facility, Hannover Medical School, Hannover, Germany; and
| | | | - Bruce Trapnell
- 8 Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Thomas Moritz
- 4 Institute of Experimental Hematology.,6 Research Group-Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence
| | - Gesine Hansen
- 1 Department of Pediatric Pneumology, Allergology and Neonatology.,2 Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL)
| |
Collapse
|
40
|
McCarthy C, Kokosi M, Bonella F. Shaping the future of an ultra-rare disease: unmet needs in the diagnosis and treatment of pulmonary alveolar proteinosis. Curr Opin Pulm Med 2019; 25:450-458. [PMID: 31365379 DOI: 10.1097/mcp.0000000000000601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Pulmonary alveolar proteinosis (PAP) can be considered the archetype of ultra-rare diseases with a prevalence of under 10 cases per million. We discuss the classification of PAP, the current diagnostic practice and the supplementary role of genetic testing and granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling in the diagnosis of congenital and hereditary PAP. We report on novel therapeutic approaches such as GM-CSF substitution, stem cell transplantation, pioglitazone, statins and immunomodulation. RECENT FINDINGS The discovery of new genetic mutations underlying this syndrome raises the question whether the classification should be radically revised in the future. Serum GM-CSF autoantibody is the best diagnostic marker for autoimmune PAP, the most common form, but does not correlate with disease severity. Several circulating biomarkers have been investigated to assess disease activity and predict outcome. Imaging techniques have also enormously evolved and offer new tools to quantify disease burden and possibly drive therapeutic decisions. Promising clinical trials are ongoing and will generate new treatment strategies besides or in addition to whole lung lavage in the next future. SUMMARY Despite impressive advances in understanding pathogenesis, PAP remains a rare syndrome with several unanswered questions impacting diagnosis, management and treatment, and, as a result, patients' quality of life.
Collapse
Affiliation(s)
- Cormac McCarthy
- Department of Respiratory Medicine, Rare Lung Disease Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Maria Kokosi
- Interstitial Lung Disease Unit, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| | - Francesco Bonella
- Department of Pneumology, Centre for Interstitial and Rare Lung Disease, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| |
Collapse
|
41
|
Smith BB, Torres NE, Hyder JA, Barbara DW, Gillespie SM, Wylam ME, Smith MM. Whole-lung Lavage and Pulmonary Alveolar Proteinosis: Review of Clinical and Patient-centered Outcomes. J Cardiothorac Vasc Anesth 2019; 33:2453-2461. [DOI: 10.1053/j.jvca.2019.03.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/19/2019] [Accepted: 03/23/2019] [Indexed: 11/11/2022]
|
42
|
Racanelli AC, Ding BS. Manmade Macrophage Offers a New Therapy for Pulmonary Alveolar Proteinosis. Am J Respir Crit Care Med 2019; 198:297-298. [PMID: 29669215 DOI: 10.1164/rccm.201803-0478ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Alexandra C Racanelli
- 1 Division of Pulmonary and Critical Care Medicine Weill Cornell Medical College New York, New York.,2 New York-Presbyterian Hospital New York, New York and
| | - Bi-Sen Ding
- 3 Ansary Stem Cell Institute Weill Cornell Medicine New York, New York
| |
Collapse
|
43
|
Misra S, Das PK, Bal SK, Elayat A, Sahoo S, Dahl AB, Kurian D, Raphael IJ, Youness HA. Therapeutic Whole Lung Lavage for Alveolar Proteinosis. J Cardiothorac Vasc Anesth 2019; 34:250-257. [PMID: 31399308 DOI: 10.1053/j.jvca.2019.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Satyajeet Misra
- Department of Anesthesiology, All India Institute of Medical Sciences, Bhubaneswar, India.
| | - Prasanta Kumar Das
- Department of Anesthesiology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Shakti Kumar Bal
- Pulmonary Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Anirudh Elayat
- Department of Anesthesiology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Satyajeet Sahoo
- Pulmonary Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Aaron B Dahl
- Department of Anesthesia and Critical Care, University of Chicago Medical Center, Chicago, IL
| | - Dinesh Kurian
- Department of Anesthesia and Critical Care, University of Chicago Medical Center, Chicago, IL
| | - Ibrahim J Raphael
- Interventional Pulmonary Program, Section of Pulmonary, Critical Care, and Sleep, Oklahoma City VA Health Care System, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Houssein A Youness
- Interventional Pulmonary Program, Section of Pulmonary, Critical Care, and Sleep, Oklahoma City VA Health Care System, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
44
|
Hetzel M, Lopez-Rodriguez E, Mucci A, Nguyen AHH, Suzuki T, Shima K, Buchegger T, Dettmer S, Rodt T, Bankstahl JP, Malik P, Knudsen L, Schambach A, Hansen G, Trapnell BC, Lachmann N, Moritz T. Effective hematopoietic stem cell-based gene therapy in a murine model of hereditary pulmonary alveolar proteinosis. Haematologica 2019; 105:1147-1157. [PMID: 31289207 PMCID: PMC7109724 DOI: 10.3324/haematol.2018.214866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 07/05/2019] [Indexed: 12/29/2022] Open
Abstract
Hereditary pulmonary alveolar proteinosis due to GM-CSF receptor deficiency (herPAP) constitutes a life-threatening lung disease characterized by alveolar deposition of surfactant protein secondary to defective alveolar macrophage function. As current therapeutic options are primarily symptomatic, we have explored the potential of hematopoietic stem cell-based gene therapy. Using Csf2rb-/- mice, a model closely reflecting the human herPAP disease phenotype, we here demonstrate robust pulmonary engraftment of an alveolar macrophage population following intravenous transplantation of lentivirally corrected hematopoietic stem and progenitor cells. Engraftment was associated with marked improvement of critical herPAP disease parameters, including bronchoalveolar fluid protein, cholesterol and cytokine levels, pulmonary density on computed tomography scans, pulmonary deposition of Periodic Acid-Schiff+ material as well as respiratory mechanics. These effects were stable for at least nine months. With respect to engraftment and alveolar macrophage differentiation kinetics, we demonstrate the rapid development of CD11c+/SiglecF+ cells in the lungs from a CD11c-/SiglecF+ progenitor population within four weeks after transplantation. Based on these data, we suggest hematopoietic stem cell-based gene therapy as an effective and cause-directed treatment approach for herPAP.
Collapse
Affiliation(s)
- Miriam Hetzel
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Elena Lopez-Rodriguez
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Adele Mucci
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Ariane Hai Ha Nguyen
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Takuji Suzuki
- Translational Pulmonary Science Center, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Pulmonary Medicine, Jichi Medical University, Shimotsukeshi, Tochigi, Japan
| | - Kenjiro Shima
- Translational Pulmonary Science Center, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Theresa Buchegger
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Sabine Dettmer
- Department of Radiology, Hannover Medical School, Hannover, Germany
| | - Thomas Rodt
- Department of Radiology, Hannover Medical School, Hannover, Germany
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Punam Malik
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Disease Institute (CBDI), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gesine Hansen
- Department of Pediatrics, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Bruce C Trapnell
- Translational Pulmonary Science Center, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Pulmonary Medicine, Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nico Lachmann
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Thomas Moritz
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
45
|
Long-Term Safety and Efficacy of Gene-Pulmonary Macrophage Transplantation Therapy of PAP in Csf2ra -/- Mice. Mol Ther 2019; 27:1597-1611. [PMID: 31326401 DOI: 10.1016/j.ymthe.2019.06.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 01/08/2023] Open
Abstract
Hereditary pulmonary alveolar proteinosis (PAP) is a genetic lung disease characterized by surfactant accumulation and respiratory failure arising from disruption of GM-CSF signaling. While mutations in either CSF2RA or CSF2RB (encoding GM-CSF receptor α or β chains, respectively) can cause PAP, α chain mutations are responsible in most patients. Pulmonary macrophage transplantation (PMT) is a promising new cell therapy in development; however, no studies have evaluated this approach for hereditary PAP (hPAP) caused by Csf2ra mutations. Here, we report on the preclinical safety, tolerability, and efficacy of lentiviral-vector (LV)-mediated Csf2ra expression in macrophages and PMT of gene-corrected macrophages (gene-PMT therapy) in Csf2ra gene-ablated (Csf2ra-/-) mice. Gene-PMT therapy resulted in a stable transgene integration and correction of GM-CSF signaling and functions in Csf2ra-/- macrophages in vitro and in vivo and resulted in engraftment and long-term persistence of gene-corrected macrophages in alveoli; restoration of pulmonary surfactant homeostasis; correction of PAP-specific cytologic, histologic, and biomarker abnormalities; and reduced inflammation associated with disease progression in untreated mice. No adverse consequences of gene-PMT therapy in Csf2ra-/- mice were observed. Results demonstrate that gene-PMT therapy of hPAP in Csf2ra-/- mice was highly efficacious, durable, safe, and well tolerated.
Collapse
|
46
|
Vuga LJ, Aggarwal NR, Reineck LA, Kalantari R, Banerjee K, Kiley J. Rare Lung Disease Research: National Heart, Lung, and Blood Institute's Commitment to Partnership and Progress. Chest 2019; 156:438-444. [PMID: 31121150 DOI: 10.1016/j.chest.2019.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 10/26/2022] Open
Affiliation(s)
- Louis J Vuga
- National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, MD
| | - Neil R Aggarwal
- National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, MD
| | - Lora A Reineck
- National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, MD
| | - Roya Kalantari
- National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, MD
| | - Koyeli Banerjee
- National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, MD
| | - James Kiley
- National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, MD.
| |
Collapse
|
47
|
De Alessandris S, Ferguson GJ, Dodd AJ, Juss JK, Devaprasad A, Piper S, Wyatt O, Killick H, Corkill DJ, Cohen ES, Pandit A, Radstake TRDJ, Simmonds R, Condliffe AM, Sleeman MA, Cowburn AS, Finch DK, Chilvers ER. Neutrophil GM-CSF receptor dynamics in acute lung injury. J Leukoc Biol 2019; 105:1183-1194. [PMID: 30942918 PMCID: PMC6850700 DOI: 10.1002/jlb.3ma0918-347r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/18/2019] [Accepted: 03/12/2019] [Indexed: 11/13/2022] Open
Abstract
GM‐CSF is important in regulating acute, persistent neutrophilic inflammation in certain settings, including lung injury. Ligand binding induces rapid internalization of the GM‐CSF receptor (GM‐CSFRα) complex, a process essential for signaling. Whereas GM‐CSF controls many aspects of neutrophil biology, regulation of GM‐CSFRα expression is poorly understood, particularly the role of GM‐CSFRα in ligand clearance and whether signaling is sustained despite major down‐regulation of GM‐CSFRα surface expression. We established a quantitative assay of GM‐CSFRα surface expression and used this, together with selective anti‐GM‐CSFR antibodies, to define GM‐CSFRα kinetics in human neutrophils, and in murine blood and alveolar neutrophils in a lung injury model. Despite rapid sustained ligand‐induced GM‐CSFRα loss from the neutrophil surface, which persisted even following ligand removal, pro‐survival effects of GM‐CSF required ongoing ligand‐receptor interaction. Neutrophils recruited to the lungs following LPS challenge showed initially high mGM‐CSFRα expression, which along with mGM‐CSFRβ declined over 24 hr; this was associated with a transient increase in bronchoalveolar lavage fluid (BALF) mGM‐CSF concentration. Treating mice in an LPS challenge model with CAM‐3003, an anti‐mGM‐CSFRα mAb, inhibited inflammatory cell influx into the lung and maintained the level of BALF mGM‐CSF. Consistent with neutrophil consumption of GM‐CSF, human neutrophils depleted exogenous GM‐CSF, independent of protease activity. These data show that loss of membrane GM‐CSFRα following GM‐CSF exposure does not preclude sustained GM‐CSF/GM‐CSFRα signaling and that this receptor plays a key role in ligand clearance. Hence neutrophilic activation via GM‐CSFR may play an important role in neutrophilic lung inflammation even in the absence of high GM‐CSF levels or GM‐CSFRα expression.
Collapse
Affiliation(s)
| | - G John Ferguson
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Alison J Dodd
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Jatinder K Juss
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Abhinandan Devaprasad
- Department of Rheumatology and Clinical Immunology and Laboratory of Translational Immunology, University Medical Centre, Utrecht, Netherlands
| | - Siân Piper
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Owen Wyatt
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Helen Killick
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Dominic J Corkill
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - E Suzanne Cohen
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Aridaman Pandit
- Department of Rheumatology and Clinical Immunology and Laboratory of Translational Immunology, University Medical Centre, Utrecht, Netherlands
| | - Timothy R D J Radstake
- Department of Rheumatology and Clinical Immunology and Laboratory of Translational Immunology, University Medical Centre, Utrecht, Netherlands
| | - Rosalind Simmonds
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Alison M Condliffe
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Matthew A Sleeman
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Andrew S Cowburn
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Donna K Finch
- Respiratory, Inflammation and Autoimmunity, MedImmune Ltd., Cambridge, United Kingdom
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
48
|
The influence of genetics on therapeutic developments in pulmonary alveolar proteinosis. Curr Opin Pulm Med 2019; 25:294-299. [PMID: 30865035 DOI: 10.1097/mcp.0000000000000576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Pulmonary alveolar proteinosis (PAP) is characterized by the massive accumulation of lipoproteinaceous material within alveoli, which results in progressive respiratory failure. The abnormalities in surfactant clearance are caused by defective pulmonary macrophages, whose terminal differentiation is GM-CSF-dependent. In hereditary PAP, the rupture of GM-CSF signaling is because of mutations in the GM-CSF receptor genes. This review focus on the innovative technologies of gene-correction proposed for the development of new therapeutic strategies, for hereditary PAP patients. RECENT FINDINGS Hematopoietic stem cell gene therapy has been successfully experimented in murine models to restore the expression of the GM-CSF receptor, however, a therapeutic approach based on bone marrow transplantation requires a preconditioning, which could be hazardous in PAP patients, who are highly susceptible to pulmonary infections. Gene-corrected pulmonary macrophages, administered directly to the lung, could represent an improved approach. Finally, patient-derived induced pluripotent stem cells seem to be promising to overcome the limited availability of primary patient cells and to generate gene-corrected macrophages, able to recover pulmonary surfactant clearance. SUMMARY WLL is the gold standard therapy for PAP. However, its use in hereditary PAP is limited by the difficulty of performing this technique in paediatric patients and by its purely symptomatic efficacy. The recent advances in genome engineering could provide efficacious strategies for clinical application.
Collapse
|
49
|
Anderson K, Carey B, Martin A, Roark C, Chalk C, Nowell-Bostic M, Freed B, Aubrey M, Trapnell B, Fontenot A. Pulmonary alveolar proteinosis: An autoimmune disease lacking an HLA association. PLoS One 2019; 14:e0213179. [PMID: 30845238 PMCID: PMC6405167 DOI: 10.1371/journal.pone.0213179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 02/15/2019] [Indexed: 11/29/2022] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is a rare lung disease characterized by the accumulation of pulmonary surfactant in alveolar macrophages and alveoli, resulting in respiratory impairment and an increased risk of opportunistic infections. Autoimmune PAP is an autoimmune lung disease that is caused by autoantibodies directed against granulocyte-macrophage colony-stimulating factor (GM-CSF). A shared feature among many autoimmune diseases is a distinct genetic association to HLA alleles. In the present study, we HLA-typed patients with autoimmune PAP to determine if this disease had any HLA association. We analyzed amino acid and allele associations for HLA-A, B, C, DRB1, DQB1, DPB1, DRB3, DRB4 and DRB5 in 41 autoimmune PAP patients compared to 1000 ethnic-matched controls and did not find any HLA association with autoimmune PAP. Collectively, these data may suggest the absence of a genetic association to the HLA in the development of autoimmune PAP.
Collapse
Affiliation(s)
- Kirsten Anderson
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, United States of America
- ClinImmune Labs Aurora, CO, United States of America
- * E-mail: (KA); (AF)
| | - Brenna Carey
- Cincinatti Children’s Hospital Medical Center (CCHMC) Cincinnati, OH, United States of America
| | - Allison Martin
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, United States of America
| | | | - Claudia Chalk
- Cincinatti Children’s Hospital Medical Center (CCHMC) Cincinnati, OH, United States of America
| | - Marchele Nowell-Bostic
- Cincinatti Children’s Hospital Medical Center (CCHMC) Cincinnati, OH, United States of America
| | - Brian Freed
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, United States of America
- ClinImmune Labs Aurora, CO, United States of America
| | - Michael Aubrey
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, United States of America
- ClinImmune Labs Aurora, CO, United States of America
| | - Bruce Trapnell
- Cincinatti Children’s Hospital Medical Center (CCHMC) Cincinnati, OH, United States of America
| | - Andrew Fontenot
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, United States of America
- * E-mail: (KA); (AF)
| |
Collapse
|
50
|
Trapnell BC, Nakata K, Bonella F, Campo I, Griese M, Hamilton J, Wang T, Morgan C, Cottin V, McCarthy C. Pulmonary alveolar proteinosis. Nat Rev Dis Primers 2019; 5:16. [PMID: 30846703 DOI: 10.1038/s41572-019-0066-3] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pulmonary alveolar proteinosis (PAP) is a syndrome characterized by the accumulation of alveolar surfactant and dysfunction of alveolar macrophages. PAP results in progressive dyspnoea of insidious onset, hypoxaemic respiratory failure, secondary infections and pulmonary fibrosis. PAP can be classified into different types on the basis of the pathogenetic mechanism: primary PAP is characterized by the disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling and can be autoimmune (caused by elevated levels of GM-CSF autoantibodies) or hereditary (due to mutations in CSF2RA or CSF2RB, encoding GM-CSF receptor subunits); secondary PAP results from various underlying conditions; and congenital PAP is caused by mutations in genes involved in surfactant production. In most patients, pathogenesis is driven by reduced GM-CSF-dependent cholesterol clearance in alveolar macrophages, which impairs alveolar surfactant clearance. PAP has a prevalence of at least 7 cases per million individuals in large population studies and affects men, women and children of all ages, ethnicities and geographical locations irrespective of socioeconomic status, although it is more-prevalent in smokers. Autoimmune PAP accounts for >90% of all cases. Management aims at improving symptoms and quality of life; whole-lung lavage effectively removes excessive surfactant. Novel pathogenesis-based therapies are in development, targeting GM-CSF signalling, immune modulation and cholesterol homeostasis.
Collapse
Affiliation(s)
- Bruce C Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Koh Nakata
- Bioscience Medical Research Center, Niigata University, Niigata, Japan
| | - Francesco Bonella
- Interstitial and Rare Lung Disease Unit, Pneumology Department, Ruhrlandklinik University Hospital, University of Essen, Essen, Germany
| | - Ilaria Campo
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Matthias Griese
- Pediatric Pneumology, University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - John Hamilton
- University of Melbourne, Parkville, Victoria, Australia
| | - Tisha Wang
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Cliff Morgan
- Department of Critical Care and Anaesthesia, Royal Brompton Hospital, London, UK
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases, University of Lyon, Lyon, France
| | - Cormac McCarthy
- Department of Medicine, St. Vincent's University Hospital and University College Dublin, Dublin, Ireland
| |
Collapse
|