1
|
Darie AM, Stolz D. Is There a Role for Bronchoscopy in Aspiration Pneumonia? Semin Respir Crit Care Med 2024. [PMID: 39447600 DOI: 10.1055/s-0044-1791739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Aspiration represents the passage of oropharyngeal content to the lower respiratory tract. The interplay between the host and the aspirate proprieties determines the subsequent aspiration syndrome. A low pH, typical of gastric aspirate, favors chemical pneumonitis, whereas an increased bacterial inoculum causes aspiration pneumonia. About a quarter of patients with aspiration pneumonitis will develop a bacterial superinfection during the course of recovery. While antibiotic therapy is indicated for aspiration pneumonia, supportive care remains the cornerstone of treatment in aspiration pneumonitis. However, the overlapping clinical features of these syndromes lead to initiation of antimicrobial therapy in most cases of aspiration. Bronchoscopy can aid in clinical decision-making by direct airway visualization and also by providing access to a series of emerging biomarkers. Invasive microbiological studies increase diagnostic yield and enable a tailored antibiotic treatment. In conjunction with stewardship programs, invasive sampling and novel molecular diagnostics can decrease the amount of inappropriate antibiotic therapy. In the context of foreign body aspiration, bronchoscopy represents both diagnostic and treatment gold standard.
Collapse
Affiliation(s)
- Andrei M Darie
- Clinic of Respiratory Medicine, University Hospital Basel, Switzerland
| | - Daiana Stolz
- Clinic of Respiratory Medicine, University of Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
2
|
Guyard A, Cazes A. [Exogenous lipid pneumonia]. Ann Pathol 2024:S0242-6498(24)00189-5. [PMID: 39322540 DOI: 10.1016/j.annpat.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/27/2024]
Abstract
Exogenous lipid pneumonia is an underdiagnosed pathology due to acute or chronic inhalation of lipid substances (paraffin oil, nasal drops, etc.). The clinical and CT appearance is polymorphic, ranging from fibrosing interstitial lung disease to a pseudo-tumoral nodule. The key to CT diagnosis is fatty hypodensity within condensations. The key to histopathological diagnosis is the presence of macrovacuoles in the macrophages, Oil-red-O+(on fresh/frozen material). The differential diagnosis is represented by endogenous lipid pneumonia, common and associated with bronchial obstruction, with necrotic changes, and specific entities such as alveolar proteinosis, hoarding (Niemann-Pick disease) or cordarone treatment.
Collapse
Affiliation(s)
- Alice Guyard
- Département de pathologie, Hôpital Bichat, AP-HP, rue Henri Huchard, 75018 Paris, France
| | - Aurélie Cazes
- Département de pathologie, Hôpital Bichat, AP-HP, rue Henri Huchard, 75018 Paris, France.
| |
Collapse
|
3
|
Ackermann M, Werlein C, Plucinski E, Leypold S, Kühnel MP, Verleden SE, Khalil HA, Länger F, Welte T, Mentzer SJ, Jonigk DD. The role of vasculature and angiogenesis in respiratory diseases. Angiogenesis 2024; 27:293-310. [PMID: 38580869 PMCID: PMC11303512 DOI: 10.1007/s10456-024-09910-2] [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: 12/20/2023] [Accepted: 02/11/2024] [Indexed: 04/07/2024]
Abstract
In European countries, nearly 10% of all hospital admissions are related to respiratory diseases, mainly chronic life-threatening diseases such as COPD, pulmonary hypertension, IPF or lung cancer. The contribution of blood vessels and angiogenesis to lung regeneration, remodeling and disease progression has been increasingly appreciated. The vascular supply of the lung shows the peculiarity of dual perfusion of the pulmonary circulation (vasa publica), which maintains a functional blood-gas barrier, and the bronchial circulation (vasa privata), which reveals a profiled capacity for angiogenesis (namely intussusceptive and sprouting angiogenesis) and alveolar-vascular remodeling by the recruitment of endothelial precursor cells. The aim of this review is to outline the importance of vascular remodeling and angiogenesis in a variety of non-neoplastic and neoplastic acute and chronic respiratory diseases such as lung infection, COPD, lung fibrosis, pulmonary hypertension and lung cancer.
Collapse
Affiliation(s)
- Maximilian Ackermann
- Institute of Pathology, University Clinics of RWTH University, Aachen, Germany.
- Institute of Pathology and Molecular Pathology, Helios University Clinic Wuppertal, University of Witten/Herdecke, Witten, Germany.
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.
| | | | - Edith Plucinski
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Sophie Leypold
- Institute of Pathology, University Clinics of RWTH University, Aachen, Germany
| | - Mark P Kühnel
- Institute of Pathology, University Clinics of RWTH University, Aachen, Germany
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Stijn E Verleden
- Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Antwerp, Belgium
| | - Hassan A Khalil
- Division of Thoracic and Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, USA
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Florian Länger
- Institute of Pathology, University Clinics of RWTH University, Aachen, Germany
| | - Tobias Welte
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Steven J Mentzer
- Division of Thoracic and Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, USA
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Danny D Jonigk
- Institute of Pathology, University Clinics of RWTH University, Aachen, Germany
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| |
Collapse
|
4
|
Zhu Y, Choi D, Somanath PR, Zhang D. Lipid-Laden Macrophages in Pulmonary Diseases. Cells 2024; 13:889. [PMID: 38891022 PMCID: PMC11171561 DOI: 10.3390/cells13110889] [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: 04/21/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Pulmonary surfactants play a crucial role in managing lung lipid metabolism, and dysregulation of this process is evident in various lung diseases. Alternations in lipid metabolism lead to pulmonary surfactant damage, resulting in hyperlipidemia in response to lung injury. Lung macrophages are responsible for recycling damaged lipid droplets to maintain lipid homeostasis. The inflammatory response triggered by external stimuli such as cigarette smoke, bleomycin, and bacteria can interfere with this process, resulting in the formation of lipid-laden macrophages (LLMs), also known as foamy macrophages. Recent studies have highlighted the potential significance of LLM formation in a range of pulmonary diseases. Furthermore, growing evidence suggests that LLMs are present in patients suffering from various pulmonary conditions. In this review, we summarize the essential metabolic and signaling pathways driving the LLM formation in chronic obstructive pulmonary disease, pulmonary fibrosis, tuberculosis, and acute lung injury.
Collapse
Affiliation(s)
- Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Dooyoung Choi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA (D.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
5
|
Vasse GF, Melgert BN. Microplastic and plastic pollution: impact on respiratory disease and health. Eur Respir Rev 2024; 33:230226. [PMID: 39009408 PMCID: PMC11262622 DOI: 10.1183/16000617.0226-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/28/2024] [Indexed: 07/17/2024] Open
Abstract
Throughout their lifecycle, from production to use and upon disposal, plastics release chemicals and particles known as micro- and nanoplastics (MNPs) that can accumulate in the environment. MNPs have been detected in different locations of the human body, including in our lungs. This is likely a consequence of MNP exposure through the air we breathe. Yet, we still lack a comprehensive understanding of the impact that MNP exposure may have on respiratory disease and health. In this review, we have collated the current body of evidence on the implications of MNP inhalation on human lung health from in vitro, in vivo and occupational exposure studies. We focused on interactions between MNP pollution and different specific lung-resident cells and respiratory diseases. We conclude that it is evident that MNPs possess the capacity to affect lung tissue in disease and health. Yet, it remains unclear to which extent this occurs upon exposure to ambient levels of MNPs, emphasising the need for a more comprehensive evaluation of environmental MNP exposure levels in everyday life.
Collapse
Affiliation(s)
- Gwenda F Vasse
- Groningen Research Institute for Pharmacy, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Barbro N Melgert
- Groningen Research Institute for Pharmacy, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Montero-Vega MT, Matilla J, Bazán E, Reimers D, De Andrés-Martín A, Gonzalo-Gobernado R, Correa C, Urbano F, Gómez-Coronado D. Fluvastatin Converts Human Macrophages into Foam Cells with Increased Inflammatory Response to Inactivated Mycobacterium tuberculosis H37Ra. Cells 2024; 13:536. [PMID: 38534380 DOI: 10.3390/cells13060536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Cholesterol biosynthesis inhibitors (statins) protect hypercholesterolemic patients against developing active tuberculosis, suggesting that these drugs could help the host to control the pathogen at the initial stages of the disease. This work studies the effect of fluvastatin on the early response of healthy peripheral blood mononuclear cells (PBMCs) to inactivated Mycobacterium tuberculosis (Mtb) H37Ra. We found that in fluvastatin-treated PBMCs, most monocytes/macrophages became foamy cells that overproduced NLRP3 inflammasome components in the absence of immune stimulation, evidencing important cholesterol metabolism/immunity connections. When both fluvastatin-treated and untreated PBMCs were exposed to Mtb H37Ra, a small subset of macrophages captured large amounts of bacilli and died, concentrating the bacteria in necrotic areas. In fluvastatin-untreated cultures, most of the remaining macrophages became epithelioid cells that isolated these areas of cell death in granulomatous structures that barely produced IFNγ. By contrast, in fluvastatin-treated cultures, foamy macrophages surrounded the accumulated bacteria, degraded them, markedly activated caspase-1 and elicited a potent IFNγ/cytotoxic response. In rabbits immunized with the same bacteria, fluvastatin increased the tuberculin test response. We conclude that statins may enhance macrophage efficacy to control Mtb, with the help of adaptive immunity, offering a promising tool in the design of alternative therapies to fight tuberculosis.
Collapse
Affiliation(s)
- María Teresa Montero-Vega
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Joaquín Matilla
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Eulalia Bazán
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Diana Reimers
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Ana De Andrés-Martín
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Rafael Gonzalo-Gobernado
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain
| | - Carlos Correa
- Unidad de Cirugía Experimental y Animalario, Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Francisco Urbano
- Servicio Interdepartamental de Investigación (SIdI), Facultad de Medicina, Universidad Autónoma, 28029 Madrid, Spain
| | - Diego Gómez-Coronado
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| |
Collapse
|
7
|
Tirelli C, Rondinone O, Italia M, Mira S, Belmonte LA, De Grassi M, Guido G, Maggioni S, Mondoni M, Miozzo MR, Centanni S. The Genetic Basis, Lung Involvement, and Therapeutic Options in Niemann-Pick Disease: A Comprehensive Review. Biomolecules 2024; 14:211. [PMID: 38397448 PMCID: PMC10886890 DOI: 10.3390/biom14020211] [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: 01/06/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Niemann-Pick Disease (NPD) is a rare autosomal recessive disease belonging to lysosomal storage disorders. Three types of NPD have been described: NPD type A, B, and C. NPD type A and B are caused by mutations in the gene SMPD1 coding for sphingomyelin phosphodiesterase 1, with a consequent lack of acid sphingomyelinase activity. These diseases have been thus classified as acid sphingomyelinase deficiencies (ASMDs). NPD type C is a neurologic disorder due to mutations in the genes NPC1 or NPC2, causing a defect of cholesterol trafficking and esterification. Although all three types of NPD can manifest with pulmonary involvement, lung disease occurs more frequently in NPD type B, typically with interstitial lung disease, recurrent pulmonary infections, and respiratory failure. In this sense, bronchoscopy with broncho-alveolar lavage or biopsy together with high-resolution computed tomography are fundamental diagnostic tools. Although several efforts have been made to find an effective therapy for NPD, to date, only limited therapeutic options are available. Enzyme replacement therapy with Olipudase α is the first and only approved disease-modifying therapy for patients with ASMD. A lung transplant and hematopoietic stem cell transplantation are also described for ASMD in the literature. The only approved disease-modifying therapy in NPD type C is miglustat, a substrate-reduction treatment. The aim of this review was to delineate a state of the art on the genetic basis and lung involvement in NPD, focusing on clinical manifestations, radiologic and histopathologic characteristics of the disease, and available therapeutic options, with a gaze on future therapeutic strategies.
Collapse
Affiliation(s)
- Claudio Tirelli
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Ornella Rondinone
- Medical Genetics Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (O.R.); (M.R.M.)
| | - Marta Italia
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Sabrina Mira
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Luca Alessandro Belmonte
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Mauro De Grassi
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Gabriele Guido
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Sara Maggioni
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Michele Mondoni
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| | - Monica Rosa Miozzo
- Medical Genetics Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (O.R.); (M.R.M.)
| | - Stefano Centanni
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.I.); (S.M.); (L.A.B.); (M.D.G.); (G.G.); (S.M.); (M.M.); (S.C.)
| |
Collapse
|
8
|
Sun Y, Saito K, Ushiki A, Abe M, Saito Y, Kashiwada T, Horimasu Y, Gemma A, Tatsumi K, Hattori N, Tsushima K, Takemoto K, Ishikawa R, Momiyama T, Matsuyama SI, Arakawa N, Akane H, Toyoda T, Ogawa K, Sato M, Takamatsu K, Mori K, Nishiya T, Izumi T, Ohno Y, Saito Y, Hanaoka M. Identification of kynurenine and quinolinic acid as promising serum biomarkers for drug-induced interstitial lung diseases. Respir Res 2024; 25:31. [PMID: 38221627 PMCID: PMC10788992 DOI: 10.1186/s12931-023-02653-6] [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: 08/10/2023] [Accepted: 12/24/2023] [Indexed: 01/16/2024] Open
Abstract
BACKGROUND Drug-induced interstitial lung disease (DILD) is a lung injury caused by various types of drugs and is a serious problem in both clinical practice and drug development. Clinical management of the condition would be improved if there were DILD-specific biomarkers available; this study aimed to meet that need. METHODS Biomarker candidates were identified by non-targeted metabolomics focusing on hydrophilic molecules, and further validated by targeted approaches using the serum of acute DILD patients, DILD recovery patients, DILD-tolerant patients, patients with other related lung diseases, and healthy controls. RESULTS Serum levels of kynurenine and quinolinic acid (and kynurenine/tryptophan ratio) were elevated significantly and specifically in acute DILD patients. The diagnostic potentials of these biomarkers were superior to those of conventional lung injury biomarkers, Krebs von den Lungen-6 and surfactant protein-D, in discriminating between acute DILD patients and patients with other lung diseases, including idiopathic interstitial pneumonia and lung diseases associated with connective tissue diseases. In addition to identifying and evaluating the biomarkers, our data showed that kynurenine/tryptophan ratios (an indicator of kynurenine pathway activation) were positively correlated with serum C-reactive protein concentrations in patients with DILD, suggesting the potential association between the generation of these biomarkers and inflammation. Our in vitro experiments demonstrated that macrophage differentiation and inflammatory stimulations typified by interferon gamma could activate the kynurenine pathway, resulting in enhanced kynurenine levels in the extracellular space in macrophage-like cell lines or lung endothelial cells. Extracellular quinolinic acid levels were elevated only in macrophage-like cells but not endothelial cells owing to the lower expression levels of metabolic enzymes converting kynurenine to quinolinic acid. These findings provide clues about the molecular mechanisms behind their specific elevation in the serum of acute DILD patients. CONCLUSIONS The serum concentrations of kynurenine and quinolinic acid as well as kynurenine/tryptophan ratios are promising and specific biomarkers for detecting and monitoring DILD and its recovery, which could facilitate accurate decisions for appropriate clinical management of patients with DILD.
Collapse
Affiliation(s)
- Yuchen Sun
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Kosuke Saito
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Atsuhito Ushiki
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Mitsuhiro Abe
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8677, Japan
| | - Yoshinobu Saito
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Takeru Kashiwada
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Yasushi Horimasu
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Koichiro Tatsumi
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8677, Japan
| | - Noboru Hattori
- Department of Respiratory Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Kenji Tsushima
- Division of General Internal Medicine, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Kazuhisa Takemoto
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Rika Ishikawa
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Toshiko Momiyama
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Shin-Ichiro Matsuyama
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Noriaki Arakawa
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Hirotoshi Akane
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Takeshi Toyoda
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Motonobu Sato
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Kazuhiko Takamatsu
- Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Kazuhiko Mori
- Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo, 134-8630, Japan
| | - Takayoshi Nishiya
- Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo, 134-8630, Japan
| | - Takashi Izumi
- Kihara Memorial Yokohama Foundation, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Yasuo Ohno
- Kihara Memorial Yokohama Foundation, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Yoshiro Saito
- Division of Medicinal Safety Science, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan.
| | - Masayuki Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| |
Collapse
|
9
|
Antonini JM, Kodali V, Meighan TG, McKinney W, Cumpston JL, Leonard HD, Cumpston JB, Friend S, Leonard SS, Andrews R, Zeidler-Erdely PC, Erdely A, Lee EG, Afshari AA. Lung toxicity, deposition, and clearance of thermal spray coating particles with different metal profiles after inhalation in rats. Nanotoxicology 2023; 17:669-686. [PMID: 38116948 PMCID: PMC10872229 DOI: 10.1080/17435390.2023.2297048] [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: 09/28/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023]
Abstract
Thermal spray coating is a process in which molten metal is sprayed onto a surface. Little is known about the health effects associated with these aerosols. Sprague-Dawley rats were exposed to aerosols (25 mg/m3 × 4 hr/d × 4 d) generated during thermal spray coating using different consumables [i.e. stainless-steel wire (PMET731), Ni-based wire (PMET885), Zn-based wire (PMET540)]. Control animals received air. Bronchoalveolar lavage was performed at 4 and 30 d post-exposure to assess lung toxicity. The particles were chain-like agglomerates and similar in size (310-378 nm). Inhalation of PMET885 aerosol caused a significant increase in lung injury and inflammation at both time points. Inhalation of PMET540 aerosol caused a slight but significant increase in lung toxicity at 4 but not 30 d. Exposure to PMET731 aerosol had no effect on lung toxicity. Overall, the lung responses were in the order: PMET885≫PMET540 >PMT731. Following a shorter exposure (25 mg/m3 × 4 h/d × 1d), lung burdens of metals from the different aerosols were determined by ICP-AES at 0, 1, 4 and 30 d post-exposure. Zn was cleared from the lungs at the fastest rate with complete clearance by 4 d post-exposure. Ni, Cr, and Mn had similar rates of clearance as nearly half of the deposited metal was cleared by 4 d. A small but significant percentage of each of these metals persisted in the lungs at 30 d. The pulmonary clearance of Fe was difficult to assess because of inherently high levels of Fe in control lungs.
Collapse
Affiliation(s)
- James M Antonini
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Vamsi Kodali
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Terence G Meighan
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Walter McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Jared L Cumpston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Howard D Leonard
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - James B Cumpston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Sherri Friend
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Stephen S Leonard
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Ronnee Andrews
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Patti C Zeidler-Erdely
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Aaron Erdely
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Eun Gyung Lee
- Respiratory Health Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Aliakbar A Afshari
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| |
Collapse
|
10
|
Janysek D, Joginpalli S, Thanawala M, Dutta A, Silva-Carmona M, Pereira M. 13-Year-Old Female With New Onset Multifocal Pulmonary Ground-Glass Opacities. Pediatrics 2023; 152:e2023061486. [PMID: 37850254 DOI: 10.1542/peds.2023-061486] [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] [Accepted: 07/17/2023] [Indexed: 10/19/2023] Open
Abstract
A 13-year-old female who recently emigrated from Honduras presented to an emergency department in Texas with a 2-month history of weight loss, fatigue, cough, and progressive shortness of breath. Her symptoms started with a nonproductive cough, and she later developed dyspnea on exertion and orthopnea. On physical examination, she was tachycardic and tachypneic. She had a thin, emaciated body habitus. She was visibly in respiratory distress with nasal flaring, tracheal tugging, and intercostal and subcostal retractions. She had diminished breath sounds at the bases and bibasilar crackles. A computed tomography scan of the chest revealed multifocal ground-glass opacities throughout all lobes of both lungs with small bilateral pleural effusions and prominent bilateral hilar lymph nodes. We will discuss the approach to the initial evaluation and subsequent diagnosis.
Collapse
Affiliation(s)
- Dawn Janysek
- Baylor College of Medicine, Houston, Texas; and Texas Children's Hospital, Houston, Texas
| | - Sharanya Joginpalli
- Baylor College of Medicine, Houston, Texas; and Texas Children's Hospital, Houston, Texas
| | - Mitali Thanawala
- Baylor College of Medicine, Houston, Texas; and Texas Children's Hospital, Houston, Texas
| | - Ankhi Dutta
- Baylor College of Medicine, Houston, Texas; and Texas Children's Hospital, Houston, Texas
| | - Manuel Silva-Carmona
- Baylor College of Medicine, Houston, Texas; and Texas Children's Hospital, Houston, Texas
| | - Maria Pereira
- Baylor College of Medicine, Houston, Texas; and Texas Children's Hospital, Houston, Texas
| |
Collapse
|
11
|
Zekre F, Duncan A, Laurent A, Tusseau M, Pescarmona R, Collardeau-Frachon S, Ohlmann C, Viel S, Reix P, Benezech S, Belot A. Rescue of Pap-Mas in Systemic JIA Using Janus Kinase Inhibitors, Case Report and Systematic Review. J Clin Med 2023; 12:jcm12072702. [PMID: 37048785 PMCID: PMC10095381 DOI: 10.3390/jcm12072702] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
INTRODUCTION Biological disease-modifying anti-rheumatic drugs (bDMARDs) targeting interleukin (IL)-6 and IL-1β represent a steroid-sparing first-line therapy used in systemic-onset juvenile idiopathic arthritis (sJIA). Recently, the occurrence of pulmonary alveolar proteinosis (PAP) in sJIA patients was reported with early-onset and exposure to bDMARDs as potential risk factors. We report on a new case with longitudinal immunomonitoring successfully treated by Janus Kinase inhibitors (JAKi) and review past clinical descriptions of this new entity. METHODS We report one case of pulmonary alveolar proteinosis and macrophage activation syndrome (PAP-MAS) with longitudinal immunomonitoring. We then conducted a review of the literature of seven publications reporting 107 cases of PAP-MAS sJIA, and included the main characteristics and evolution under treatment. RESULTS Of the seven articles analyzed, the incidence of PAP-MAS among sJIA patients varied from 1.28% to 12.9%. We report here a single case among a cohort of 537 sJIA patients followed in the pediatric department of the Hospices Civils de Lyon over the last 15 years. This child presented with all clinical and immunological characteristics of PAP-MAS. After several lines of treatment, he benefited from JAKi and improved with respect to both systemic symptoms and lung disease. In the literature, strategies with monoclonal antibodies targeting either INF-γ or IL-1β/IL-18 have been tested with variable results. Orally taken JAKi presents the advantage of targeting multiple cytokines and avoiding parenteral injections of monoclonal antibodies that may contribute to the pathogenesis. CONCLUSIONS JAKi represent a promising option in the treatment of lung disease associated with sJIA.
Collapse
Affiliation(s)
- Franck Zekre
- Nephrology, Rheumatology and Dermatology Unit, National Reference Centre for Rare Rheumatic Autoimmune and Systemic Diseases in Children (RAISE), Mère Enfant Hospital, Hospices Civils de Lyon, 69500 Bron, France
- International Center of Infectiology Research (CIRI), INSERM U1111, CNRS, UMR5308, ENS of Lyon, Claude Bernard University Lyon 1, 69007 Lyon, France
| | - Anita Duncan
- Nephrology, Rheumatology and Dermatology Unit, National Reference Centre for Rare Rheumatic Autoimmune and Systemic Diseases in Children (RAISE), Mère Enfant Hospital, Hospices Civils de Lyon, 69500 Bron, France
| | - Audrey Laurent
- Nephrology, Rheumatology and Dermatology Unit, National Reference Centre for Rare Rheumatic Autoimmune and Systemic Diseases in Children (RAISE), Mère Enfant Hospital, Hospices Civils de Lyon, 69500 Bron, France
| | - Maud Tusseau
- International Center of Infectiology Research (CIRI), INSERM U1111, CNRS, UMR5308, ENS of Lyon, Claude Bernard University Lyon 1, 69007 Lyon, France
- Department of Clinical Genetics, Hospices Civils de Lyon, 69500 Lyon, France
| | - Rémi Pescarmona
- Immunology Unit, Hôpital Edouard Herriot, 69003 Lyon, France
| | | | - Camille Ohlmann
- Service de Pneumologie Pédiatrique, Allergologie et Mucoviscidose, Mère-Enfant Hospital, Hospices Civils de Lyon, 69500 Bron, France
| | - Sébastien Viel
- International Center of Infectiology Research (CIRI), INSERM U1111, CNRS, UMR5308, ENS of Lyon, Claude Bernard University Lyon 1, 69007 Lyon, France
- Immunology, Allergy and Immunomonitoring Unit, Groupement Hôpitaux du Sud, 69310 Lyon, France
| | - Philippe Reix
- Service de Pneumologie Pédiatrique, Allergologie et Mucoviscidose, Mère-Enfant Hospital, Hospices Civils de Lyon, 69500 Bron, France
- UMR CNRS 5558 (équipe EMET), Laboratoire de Biométrie et Biologie Evolutive (LBBE), Université Claude Bernard Lyon 1, 69622 Villeurbanne, France
| | - Sarah Benezech
- International Center of Infectiology Research (CIRI), INSERM U1111, CNRS, UMR5308, ENS of Lyon, Claude Bernard University Lyon 1, 69007 Lyon, France
- Institute of Hematology and Pediatric Oncology, 69008 Lyon, France
| | - Alexandre Belot
- Nephrology, Rheumatology and Dermatology Unit, National Reference Centre for Rare Rheumatic Autoimmune and Systemic Diseases in Children (RAISE), Mère Enfant Hospital, Hospices Civils de Lyon, 69500 Bron, France
- International Center of Infectiology Research (CIRI), INSERM U1111, CNRS, UMR5308, ENS of Lyon, Claude Bernard University Lyon 1, 69007 Lyon, France
| |
Collapse
|
12
|
Agarwal M, Cummings K, Larsen B, Chopra M, Rodriguez-Pla A. Late Onset of Rivaroxaban-Associated Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis. J Investig Med High Impact Case Rep 2023; 11:23247096231207689. [PMID: 37902294 PMCID: PMC10627018 DOI: 10.1177/23247096231207689] [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: 03/08/2023] [Revised: 08/31/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Although anti-thyroid drugs (ATDs) are the most common cause of drug-associated anti-neutrophil cytoplasmic antibody (ANCA) vasculitis (AAV), many other classes of drugs can lead to drug-associated AAV. We present a unique case of rivaroxaban-associated AAV. A 76-year-old female with a past medical history of atrial fibrillation on rivaroxaban presented with fatigue, bilateral lower extremity purpura, and hemoptysis to an outside hospital. Investigations revealed a positive cytoplasmic-ANCA (c-ANCA) titer of 1:320 and a positive anti-myeloperoxidase (anti-MPO), and negative perinuclear-ANCA (p-ANCA) and anti-proteinase 3 (anti-PR3). In addition, chest imaging demonstrated bilateral ground-glass opacities which raised suspicion for diffuse alveolar hemorrhage (DAH). A lung biopsy revealed acute and ongoing DAH with focally active capillaritis and characteristic pathological findings, which strongly suggested that was likely secondary to rivaroxaban. Rivaroxaban was discontinued, and the patient received pulses of intravenous glucocorticosteroids and rituximab. Her symptoms improved. She continued immunosuppressive therapy with rituximab for 2 years. She presented to our hospital for a second opinion regarding the discontinuation of rituximab, and we decided to discontinue rituximab. After discontinuation, the patient remained stable after 1.5 years of follow-up and did not have any relapses. This is a unique case of rivaroxaban-associated AAV. Clinicians should consider drug-associated AAV in all patients who present with an atypical clinical presentation and/or pathological findings of AAV. Given the broad and rapidly increasing use of novel anticoagulants, it is important to raise awareness of this potential complication. Prompt discontinuation of the drug and initiation of immunosuppressant treatment in severe cases may be lifesaving.
Collapse
Affiliation(s)
- Muskan Agarwal
- Department of Internal Medicine, Mayo Clinic, Phoenix AZ, USA
| | | | - Brandon Larsen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, USA
| | - Madhav Chopra
- Division of Pulmonary, The University of Arizona, Tucson, AZ, USA
| | - Alicia Rodriguez-Pla
- Division of Rheumatology, University of California San Francisco in Fresno, Fresno, CA, USA
| |
Collapse
|
13
|
Soto B, Costanzo L, Puskoor A, Akkari N, Geraghty P. The implications of Vitamin E acetate in E-cigarette, or vaping, product use-associated lung injury. Ann Thorac Med 2023; 18:1-9. [PMID: 36968330 PMCID: PMC10034821 DOI: 10.4103/atm.atm_144_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 11/05/2022] [Indexed: 01/26/2023] Open
Abstract
In the summer of 2019, a cluster of cases were observed with users of battery-operated or superheating devices presenting with multiple symptoms, such as dyspnea, cough, fever, constitutional symptoms, gastrointestinal upset, and hemoptysis, that is now termed e-cigarette, or vaping, product use-associated lung injury (EVALI). The Centers for Disease Control and Prevention reported 2807 cases within the USA leading to at least 68 deaths as of February 18, 2020. The heterogeneous presentations of EVALI make diagnosis and treatment difficult; however, treatment focused on identifying and removal of the noxious substance and providing supportive care. Vitamin E acetate (VEA) is a likely cause of this lung injury, and others have reported other components to play a possible role, such as nicotine and vegetable glycerin/propylene glycol. EVALI is usually observed in adolescents, with a history of vaping product usage within 90 days typically containing tetrahydrocannabinol, and presenting on chest radiograph with pulmonary infiltrates or computed tomography scan with ground-glass opacities. Diagnosis requires a high degree of suspicion to diagnose and exclusion of other possible causes of lung disease. Here, we review the current literature to detail the major factors contributing to EVALI and primarily discuss the potential role of VEA in EVALI. We will also briefly discuss other constituents other than just VEA, as a small number of EVALI cases are reported without the detection of VEA, but with the same clinical diagnosis.
Collapse
Affiliation(s)
- Brian Soto
- Department of Medicine, State University of New York Downstate Health Sciences University, NY, USA
| | - Louis Costanzo
- Department of Medicine, State University of New York Downstate Health Sciences University, NY, USA
| | - Anoop Puskoor
- Department of Medicine, State University of New York Downstate Health Sciences University, NY, USA
| | - Nada Akkari
- Department of Medicine, State University of New York Downstate Health Sciences University, NY, USA
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, NY, USA
| |
Collapse
|
14
|
Calabrese F, Roden AC, Pavlisko E, Lunardi F, Neil D, Adam B, Hwang D, Goddard M, Berry GJ, Ivanovic M, Thüsen JVD, Gibault L, Lin CY, Wassilew K, Glass C, Westall G, Zeevi A, Levine DJ, Roux A. LUNG ALLOGRAFT STANDARDIZED HISTOLOGICAL ANALYSIS (LASHA) TEMPLATE: A RESEARCH CONSENSUS PROPOSAL. J Heart Lung Transplant 2022; 41:1487-1500. [DOI: 10.1016/j.healun.2022.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/10/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
|
15
|
Ramos-Martínez E, Falfán-Valencia R, Pérez-Rubio G, Mejía M, Mejía-Hurtado JG, Buendía-Roldán I, González-Pérez MI, Mateos-Toledo HN, Rojas-Serrano J. Interstitial lung disease progression in patients with anti-aminoacyl transfer-RNA-synthetase autoantibodies is characterized by higher levels of sCD163. Immunol Lett 2022; 248:56-61. [PMID: 35728747 DOI: 10.1016/j.imlet.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/22/2022] [Accepted: 06/17/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Patients with anti-tRNA autoantibodies are characterized by arthritis, mechanic´s hands, fever, Raynaud´s phenomenon, and interstitial lung disease (ILD), in at least two clinical scenarios: the antisynthetase syndrome (ASSD) and interstitial pneumonia with autoimmune features (IPAF). The anti-tRNA-ILD treatment is centered on the administration of corticosteroids and a wide variety of immunosuppressive drugs; however, the effectiveness of the treatment depends on factors not fully understood. This research work aimed to quantify the serum levels of two molecules related to pulmonary fibrosis and explore their relationship with the progression of ILD associated with ASSD METHODOLOGY: Serum levels of sCD163 and TGF-β1 from baseline and after six months of treatment of ILD patients' positives to anti-tRNA were included in the current study. At six months, patients were classified as with or without ILD progression RESULTS: Forty patients were included (anti-Jo1, anti-PL7, anti-PL12, and anti-Ej). Five patients (12.5%) had ILD progression and were characterized by higher levels of sCD163 at baseline. Baseline sCD163 serum levels showed good discriminatory capacity in patients with ILD progression. On the other hand, at follow-up, serum TGF-β1 levels significantly increased in both patients' groups, with and without progression CONCLUSION: Basal levels of sCD163 were higher in patients who later developed ILD progression and kinetics of both molecules suggests the participation of M2 macrophages in the development of ILD.
Collapse
Affiliation(s)
- Espiridión Ramos-Martínez
- Experimental Medicine Research Unit, Facultad de Medicina, Universidad Nacional Autónoma de México 06720 México City Mexico
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México City 14080, Mexico
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México City 14080, Mexico
| | - Mayra Mejía
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ismael Cosío Villegas, Calz. Tlalpan 4502, Sección XVI, Tlalpan, México City, Ciudad de México 14080, Mexico
| | - José Guillermo Mejía-Hurtado
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ismael Cosío Villegas, Calz. Tlalpan 4502, Sección XVI, Tlalpan, México City, Ciudad de México 14080, Mexico
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México City 14080, Mexico
| | - Montserrat I González-Pérez
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ismael Cosío Villegas, Calz. Tlalpan 4502, Sección XVI, Tlalpan, México City, Ciudad de México 14080, Mexico
| | - Heidegger N Mateos-Toledo
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ismael Cosío Villegas, Calz. Tlalpan 4502, Sección XVI, Tlalpan, México City, Ciudad de México 14080, Mexico
| | - Jorge Rojas-Serrano
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ismael Cosío Villegas, Calz. Tlalpan 4502, Sección XVI, Tlalpan, México City, Ciudad de México 14080, Mexico; Profesor, Programa de Maestría y Doctorado en Ciencias Médicas, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico.
| |
Collapse
|
16
|
Musiu C, Caligola S, Fiore A, Lamolinara A, Frusteri C, Del Pizzo FD, De Sanctis F, Canè S, Adamo A, Hofer F, Barouni RM, Grilli A, Zilio S, Serafini P, Tacconelli E, Donadello K, Gottin L, Polati E, Girelli D, Polidoro I, Iezzi PA, Angelucci D, Capece A, Chen Y, Shi ZL, Murray PJ, Chilosi M, Amit I, Bicciato S, Iezzi M, Bronte V, Ugel S. Fatal cytokine release syndrome by an aberrant FLIP/STAT3 axis. Cell Death Differ 2022; 29:420-438. [PMID: 34518653 PMCID: PMC8435761 DOI: 10.1038/s41418-021-00866-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammatory responses rapidly detect pathogen invasion and mount a regulated reaction. However, dysregulated anti-pathogen immune responses can provoke life-threatening inflammatory pathologies collectively known as cytokine release syndrome (CRS), exemplified by key clinical phenotypes unearthed during the SARS-CoV-2 pandemic. The underlying pathophysiology of CRS remains elusive. We found that FLIP, a protein that controls caspase-8 death pathways, was highly expressed in myeloid cells of COVID-19 lungs. FLIP controlled CRS by fueling a STAT3-dependent inflammatory program. Indeed, constitutive expression of a viral FLIP homolog in myeloid cells triggered a STAT3-linked, progressive, and fatal inflammatory syndrome in mice, characterized by elevated cytokine output, lymphopenia, lung injury, and multiple organ dysfunctions that mimicked human CRS. As STAT3-targeting approaches relieved inflammation, immune disorders, and organ failures in these mice, targeted intervention towards this pathway could suppress the lethal CRS inflammatory state.
Collapse
Affiliation(s)
- Chiara Musiu
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Simone Caligola
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Alessandra Fiore
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy ,grid.418615.f0000 0004 0491 845XMax Planck Institute of Biochemistry, Martinsried, Planegg, Germany
| | - Alessia Lamolinara
- grid.412451.70000 0001 2181 4941CAST - Center for Advanced Studies and Technology, Department of Neurosciences Imaging and Clinical Sciences, University of G. D’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Cristina Frusteri
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Francesco Domenico Del Pizzo
- grid.412451.70000 0001 2181 4941CAST - Center for Advanced Studies and Technology, Department of Neurosciences Imaging and Clinical Sciences, University of G. D’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Francesco De Sanctis
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Stefania Canè
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Annalisa Adamo
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Francesca Hofer
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Roza Maria Barouni
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Andrea Grilli
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Serena Zilio
- grid.26790.3a0000 0004 1936 8606Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Paolo Serafini
- grid.26790.3a0000 0004 1936 8606Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Evelina Tacconelli
- grid.411475.20000 0004 1756 948XDivision of Infectious Diseases, Department of Diagnostics and Public Health, University and Hospital Trust of Verona, Verona, Italy
| | - Katia Donadello
- grid.411475.20000 0004 1756 948XIntensive Care Unit, Department of Surgery, Dentistry, Maternity and Infant, University and Hospital Trust of Verona, Verona, Italy
| | - Leonardo Gottin
- grid.411475.20000 0004 1756 948XIntensive Care Unit, Department of Surgery, Dentistry, Maternity and Infant, University and Hospital Trust of Verona, Verona, Italy
| | - Enrico Polati
- grid.411475.20000 0004 1756 948XIntensive Care Unit, Department of Surgery, Dentistry, Maternity and Infant, University and Hospital Trust of Verona, Verona, Italy
| | - Domenico Girelli
- grid.411475.20000 0004 1756 948XDivision of Internal Medicine, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Ildo Polidoro
- Complex Operational Unit of Forensic Medicine, Local Health Authority of Pescara, Pescara, Italy
| | - Piera Amelia Iezzi
- Complex Operational Unit of Forensic Medicine, Local Health Authority of Pescara, Pescara, Italy
| | - Domenico Angelucci
- Pathological Anatomy Unit, Local Health Authority of Lanciano-Vasto-Chieti, Vasto, Italy
| | - Andrea Capece
- Pathological Anatomy Unit, Local Health Authority of Lanciano-Vasto-Chieti, Vasto, Italy
| | - Ying Chen
- grid.439104.b0000 0004 1798 1925CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei People’s Republic of China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Zheng-Li Shi
- grid.439104.b0000 0004 1798 1925CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei People’s Republic of China
| | - Peter J. Murray
- grid.418615.f0000 0004 0491 845XMax Planck Institute of Biochemistry, Martinsried, Planegg, Germany
| | - Marco Chilosi
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Ido Amit
- grid.13992.300000 0004 0604 7563Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Silvio Bicciato
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Iezzi
- grid.412451.70000 0001 2181 4941CAST - Center for Advanced Studies and Technology, Department of Neurosciences Imaging and Clinical Sciences, University of G. D’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Vincenzo Bronte
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| | - Stefano Ugel
- grid.411475.20000 0004 1756 948XImmunology Section, Department of Medicine, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|
17
|
Todd NW, Atamas SP, Hines SE, Luzina IG, Shah NG, Britt EJ, Ghio AJ, Galvin JR. Demystifying idiopathic interstitial pneumonia: time for more etiology-focused nomenclature in interstitial lung disease. Expert Rev Respir Med 2022; 16:235-245. [PMID: 35034567 PMCID: PMC8983480 DOI: 10.1080/17476348.2022.2030710] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION A major focus of interstitial lung disease (ILD) has centered on disorders termed idiopathic interstitial pneumonias (IIPs) which include, among others, idiopathic pulmonary fibrosis, idiopathic nonspecific interstitial pneumonia, cryptogenic organizing pneumonia, and respiratory bronchiolitis-interstitial lung disease. AREAS COVERED We review the radiologic and histologic patterns for the nine disorders classified by multidisciplinary approach as IIP, and describe the remarkable amount of published epidemiologic, translational, and molecular studies demonstrating their associations with numerous yet definitive environmental exposures, occupational exposures, pulmonary diseases, systemic diseases, medication toxicities, and genetic variants. EXPERT OPINION In the 21st century, these disorders termed IIPs are rarely idiopathic, but rather are well-described radiologic and histologic patterns of lung injury that are associated with a wide array of diverse etiologies. Accordingly, the idiopathic nomenclature is misleading and confusing, and may also promote a lack of inquisitiveness, suggesting the end rather than the beginning of a thorough diagnostic process to identify ILD etiology and initiate patient-centered management. A shift toward more etiology-focused nomenclature will be beneficial to all, including patients hoping for better life quality and disease outcome, general medicine and pulmonary physicians furthering their ILD knowledge, and expert ILD clinicians and researchers who are advancing the ILD field.
Collapse
Affiliation(s)
- Nevins W. Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | - Sergei P. Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | - Stella E. Hines
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Irina G. Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,,Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | - Nirav G. Shah
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Edward J. Britt
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrew J. Ghio
- Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Jeffrey R. Galvin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,,Department of Diagnostic Radiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Kim YC, Kim J, Kim S, Bae B, Kim RL, Jeong EM, Cho SH, Kang HR. Transglutaminase 2 mediates lung inflammation and remodeling by transforming growth factor beta 1 via alveolar macrophage modulation. Exp Lung Res 2021; 47:465-475. [PMID: 34818962 DOI: 10.1080/01902148.2021.1998733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Transforming growth factor beta 1 (TGF-β1) induces pulmonary fibrosis by enhancing epithelial apoptosis and affects the enzymatic activity of transglutaminase 2 (TG2). The aim of this study was to determine the role of TG2 in TGF-β1-induced lung remodeling and alveolar macrophage modulation. We characterized the in vivo effects of TGF-β1 and TG2 on lung inflammation, fibrosis, and macrophage activity using transgenic C57BL/6 mice with wild and null TG2 loci. The effect of TG2 inhibition on in vitro TGF-β1-stimulated alveolar macrophages was assessed through mRNA analysis. TG2 was remarkably upregulated in the lungs of TGF-β1 transgenic (TGF-β1 Tg) mice, especially in alveolar macrophages and epithelial cells. In the absence of TG2, TGF-β1-induced inflammation was suppressed, decreasing the number of macrophages in the bronchoalveolar lavage fluid. In addition, the alveolar destruction and peribronchial fibrosis induced by TGF-β1 overexpression were significantly reduced, which correlated with decreases in the expression of fibroblast growth factor and matrix metallopeptidase 12, respectively. However, TG2 deficiency did not compromise the phagocytic activity of alveolar macrophages in TGF-β1 Tg mice. At the same time, TG2 contributed to the regulation of TGF-β1-induced macrophage activation. Inhibition of TG2 did not affect the TGF-β1-induced expression of CD86, an M1 marker, in macrophages, but it did reverse the TGF-β1-induced expression of CD206. This result suggests that TG2 mediates TGF-β1-induced M2-like polarization but does not contribute to TGF-β1-induced M1 polarization. In conclusion, TG2 regulates macrophage modulation and plays an important role in TGF-β1-induced lung inflammation, destruction, and fibrosis.
Collapse
Affiliation(s)
- Young Chan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Jeonghyeon Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea.,Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Subin Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Boram Bae
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Ruth Lee Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea.,Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eui-Man Jeong
- Department of Pharmacy, Jeju National University College of Pharmacy, Jeju, Korea
| | - Sang-Heon Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Hye-Ryun Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
19
|
Abstract
Cell membrane fusion and multinucleation in macrophages are associated with physiologic homeostasis as well as disease. Osteoclasts are multinucleated macrophages that resorb bone through increased metabolic activity resulting from cell fusion. Fusion of macrophages also generates multinucleated giant cells (MGCs) in white adipose tissue (WAT) of obese individuals. For years, our knowledge of MGCs in WAT has been limited to their description as part of crown-like structures (CLS) surrounding damaged adipocytes. However, recent evidence indicates that these cells can phagocytose oversized lipid remnants, suggesting that, as in osteoclasts, cell fusion and multinucleation are required for specialized catabolic functions. We thus reason that WAT MGCs can be viewed as functionally analogous to osteoclasts and refer to them in this article as adipoclasts. We first review current knowledge on adipoclasts and their described functions. In view of recent advances in single cell genomics, we describe WAT macrophages from a ‘fusion perspective’ and speculate on the ontogeny of adipoclasts. Specifically, we highlight the role of CD9 and TREM2, two plasma membrane markers of lipid-associated macrophages in WAT, which have been previously described as regulators of fusion and multinucleation in osteoclasts and MGCs. Finally, we consider whether strategies aiming to target WAT macrophages can be more selectively directed against adipoclasts.
Collapse
|
20
|
Colbenson GA, Dempsey TM, Cecchini MJ, Aubry MC, Moua T, Ryu JH. A Bodybuilder with Dyspnea and Hypercalcemia. Am J Med 2021; 134:e363-e365. [PMID: 33316251 DOI: 10.1016/j.amjmed.2020.10.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 11/27/2022]
Affiliation(s)
| | | | - Matthew J Cecchini
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | | | - Teng Moua
- Division of Pulmonary and Critical Care Medicine
| | - Jay H Ryu
- Division of Pulmonary and Critical Care Medicine
| |
Collapse
|
21
|
Li Y, Jiang Y, Zhang Y, Li N, Yin Q, Liu L, Lv X, Liu Y, Li A, Fang B, Li J, Ye H, Yang G, Cui X, Liu Y, Qu Y, Li C, Li J, Li D, Gai Z, Wang S, Zhan F, Liang M. Abnormal upregulation of cardiovascular disease biomarker PLA2G7 induced by proinflammatory macrophages in COVID-19 patients. Sci Rep 2021; 11:6811. [PMID: 33762651 PMCID: PMC7990942 DOI: 10.1038/s41598-021-85848-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/08/2021] [Indexed: 01/07/2023] Open
Abstract
High rate of cardiovascular disease (CVD) has been reported among patients with coronavirus disease 2019 (COVID-19). Importantly, CVD, as one of the comorbidities, could also increase the risks of the severity of COVID-19. Here we identified phospholipase A2 group VII (PLA2G7), a well-studied CVD biomarker, as a hub gene in COVID-19 though an integrated hypothesis-free genomic analysis on nasal swabs (n = 486) from patients with COVID-19. PLA2G7 was further found to be predominantly expressed by proinflammatory macrophages in lungs emerging with progression of COVID-19. In the validation stage, RNA level of PLA2G7 was identified in nasal swabs from both COVID-19 and pneumonia patients, other than health individuals. The positive rate of PLA2G7 were correlated with not only viral loads but also severity of pneumonia in non-COVID-19 patients. Serum protein levels of PLA2G7 were found to be elevated and beyond the normal limit in COVID-19 patients, especially among those re-positive patients. We identified and validated PLA2G7, a biomarker for CVD, was abnormally enhanced in COVID-19 at both nucleotide and protein aspects. These findings provided indications into the prevalence of cardiovascular involvements seen in patients with COVID-19. PLA2G7 could be a potential prognostic and therapeutic target in COVID-19.
Collapse
Affiliation(s)
- Yang Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yongzhong Jiang
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430065, China
| | - Yi Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Naizhe Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Qiangling Yin
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Linlin Liu
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430065, China
| | - Xin Lv
- Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University and Jinan Children's Hospital, Jinan, 250022, China
| | - Yan Liu
- Department of Microbiology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, China
| | - Aqian Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Bin Fang
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430065, China
| | - Jiajia Li
- The Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hengping Ye
- Xiantao Center for Disease Control and Prevention, Xiantao, 433000, China
| | - Gang Yang
- Xiangyang Center for Disease Control and Prevention, Xiangyang, 441000, China
| | - Xiaoxian Cui
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai, 200336, China
| | - Yang Liu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yuanyuan Qu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Chuan Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Jiandong Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Dexin Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Zhongtao Gai
- Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University and Jinan Children's Hospital, Jinan, 250022, China
| | - Shiwen Wang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
- CDC-WIV Joint Research Center for Emerging Diseases and Biosafety, Wuhan, 430071, China.
| | - Faxian Zhan
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430065, China.
| | - Mifang Liang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
- CDC-WIV Joint Research Center for Emerging Diseases and Biosafety, Wuhan, 430071, China.
| |
Collapse
|
22
|
Meidaninikjeh S, Sabouni N, Marzouni HZ, Bengar S, Khalili A, Jafari R. Monocytes and macrophages in COVID-19: Friends and foes. Life Sci 2021; 269:119010. [PMID: 33454368 PMCID: PMC7834345 DOI: 10.1016/j.lfs.2020.119010] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/27/2020] [Accepted: 12/27/2020] [Indexed: 01/08/2023]
Abstract
The COVID-19 is a novel infectious disease caused by SARS-CoV-2 and is known as a pandemic emergency that has led to a high rate of mortality throughout the world. Evidence has indicated that hyperinflammatory responses triggered by SARS-CoV-2 are the main cause of pathogenicity in the severe cases of patients who have died during the current viral disease. Monocytes and macrophages as the most important cells of the innate arm of the immune system play a substantial part in the body's defense against viral infections. They mainly respond to the microbial antigens by producing inflammatory mediators to remove pathogens and repair tissue injury. Nevertheless, aberrant alterations in their function such as cytokine storm can be so harmful to the host in the acute respiratory distress syndrome cases caused by SARS-CoV-2. Moreover, inflammatory responses stimulated by SARS-CoV-2 have affected the other vital organs of the body including the heart. As cardiovascular complications in COVID-19 patients have been reported in several studies. During the infection, monocytes and macrophages may be involved in the hypersensitive and exacerbated reactions that contribute to the tissue damage, especially lung injury resulted in its dysfunction and respiratory disorder. In this review, we discuss both advantageous and disadvantageous about the pathological potential of monocytes and macrophages during the infection of SARS-CoV-2 to clarify their mutual effects on immune processing as a fist line defender in the current disease.
Collapse
Affiliation(s)
- Sepideh Meidaninikjeh
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Nasim Sabouni
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Zare Marzouni
- Qaen School of Nursing and Midwifery, Birjand University of Medical Science, Birjand, Iran
| | - Sajad Bengar
- Department of Microbiology, Faculty of Science, Shahre Ghods Branch, Islamic Azad University, Shahre Ghods, Tehran, Iran
| | - Ahmad Khalili
- Cancer Biomedical Center (CBC) Research Institute, Tehran, Iran.
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
23
|
Cecchini MJ, Mukhopadhyay S, Arrossi AV, Beasley MB, Butt YM, Jones KD, Pambuccian S, Mehrad M, Monaco SE, Saqi A, Smith ML, Tazelaar HD, Larsen BT. E-Cigarette or Vaping Product Use-Associated Lung Injury: A Review for Pathologists. Arch Pathol Lab Med 2021; 144:1490-1500. [PMID: 32401055 DOI: 10.5858/arpa.2020-0024-ra] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2010] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Vaping is the inhalation of heated aerosol from a small battery-powered device as a method to deliver nicotine or other substances. A recent outbreak of severe respiratory illness primarily in the United States has put a spotlight on vaping and its potential risks. OBJECTIVE.— To familiarize pathologists with vaping, the cytologic and histopathologic features of vaping-associated acute lung injury, and the role of pathology in this diagnosis. DATA SOURCES.— A targeted literature review was performed. CONCLUSIONS.— Most cases of vaping-associated lung injury have been linked to vaping products containing tetrahydrocannabinol or other cannabinoids. Lung biopsies show a spectrum of nonspecific acute lung injury patterns (organizing pneumonia, diffuse alveolar damage, acute fibrinous, and organizing pneumonia, or combinations of the above), accompanied by prominent, foamy macrophage accumulation. Injury is usually accentuated around small airways. Lipid-laden macrophages can be identified in bronchioloalveolar lavage fluid in most patients and these can be highlighted using lipid stains, such as oil red O, but the clinical utility of this finding remains unclear, as lipid-laden macrophages can be seen in a wide variety of processes and should not be relied upon to make the diagnosis. Classic histologic features of exogenous lipoid pneumonia have not been identified in tissue samples. Lightly pigmented macrophages, similar to those seen with traditional cigarette smoking, are present in some cases but are usually a minor feature. To date, no specific pathologic features for vaping-related injury have been identified, and it remains a diagnosis of exclusion that requires clinicopathologic correlation.
Collapse
Affiliation(s)
- Matthew J Cecchini
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Cecchini)
| | - Sanjay Mukhopadhyay
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio (Mukhopadhyay, Arrossi)
| | - Andrea V Arrossi
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio (Mukhopadhyay, Arrossi)
| | - Mary B Beasley
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York (Beasley)
| | - Yasmeen M Butt
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Butt, Smith, Tazelaar, Larsen)
| | - Kirk D Jones
- Department of Pathology, University of California San Francisco, San Francisco (Jones)
| | - Stefan Pambuccian
- Department of Pathology, Loyola University Medical Center and Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois (Pambuccian)
| | - Mitra Mehrad
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Mehrad)
| | - Sara E Monaco
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Monaco)
| | - Anjali Saqi
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York (Saqi)
| | - Maxwell L Smith
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Butt, Smith, Tazelaar, Larsen)
| | - Henry D Tazelaar
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Butt, Smith, Tazelaar, Larsen)
| | - Brandon T Larsen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Butt, Smith, Tazelaar, Larsen)
| |
Collapse
|
24
|
Reagan-Steiner S, Gary J, Matkovic E, Ritter JM, Shieh WJ, Martines RB, Werner AK, Lynfield R, Holzbauer S, Bullock H, Denison AM, Bhatnagar J, Bollweg BC, Patel M, Evans ME, King BA, Rose DA, Baldwin GT, Jones CM, Krishnasamy V, Briss PA, Weissman DN, Meaney-Delman D, Zaki SR, Jatlaoui T, Koumans E, Kiernan E, Petersen E, Karwowski MP, Valentin-Blasini L, Blount BC, Cummings KJ, Ghinai I, Feder K, Wells EV, Turabelidze G, Byers P, Tanz LJ, Navarette KA, Ramadugu K, Dewart C, Miller J, Squires K, Marsden L, Fields CA. Pathological findings in suspected cases of e-cigarette, or vaping, product use-associated lung injury (EVALI): a case series. THE LANCET RESPIRATORY MEDICINE 2020; 8:1219-1232. [DOI: 10.1016/s2213-2600(20)30321-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
|
25
|
McCarthy C, Keane MP, Fabre A. Lipid-Laden Macrophages Are Not Diagnostic of Pulmonary Alveolar Proteinosis Syndrome and Can Indicate Lung Injury. Am J Respir Crit Care Med 2020; 202:1197-1198. [PMID: 32673084 PMCID: PMC7560793 DOI: 10.1164/rccm.202005-1880le] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Cormac McCarthy
- St. Vincent's University Hospital Dublin, Ireland and.,University College Dublin Dublin, Ireland
| | - Michael P Keane
- St. Vincent's University Hospital Dublin, Ireland and.,University College Dublin Dublin, Ireland
| | - Aurelie Fabre
- St. Vincent's University Hospital Dublin, Ireland and.,University College Dublin Dublin, Ireland
| |
Collapse
|
26
|
Murakami Y, Sakamoto K, Okumura Y, Suzuki A, Mii S, Sato M, Yokoi T, Hashimoto N, Hasegawa Y. Acute Exacerbation of Pleuroparenchymal Fibroelastosis Secondary to Allogenic Hematopoietic Stem Cell Transplantation. Intern Med 2020; 59:2737-2743. [PMID: 32669500 PMCID: PMC7691016 DOI: 10.2169/internalmedicine.4995-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
In this article, we report a case with pleuroparenchymal fibroelastosis (PPFE) following hematopoietic stem cell transplantation (HSCT) that developed acute respiratory failure with new bilateral ground glass opacity, which could not be explained by either a pulmonary infection, drug toxicity or extraparenchymal causes. Although combination therapy with multiple immunosuppressants was transiently effective, the patient died from a recurrent exacerbation. Autopsied lungs demonstrated diffuse alveolar damage superimposed on PPFE. There was no evidence of any coexisting interstitial pneumonia with the usual interstitial pneumonia (UIP) pattern. Our case suggests that acute exacerbation can occur in patients with post-HSCT PPFE, even when a coexisting UIP pattern is absent.
Collapse
Affiliation(s)
- Yasushi Murakami
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Japan
| | - Koji Sakamoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Japan
| | - Yuki Okumura
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Japan
| | - Atsushi Suzuki
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Japan
| | - Mitsuo Sato
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Japan
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Japan
| | | | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Japan
| | - Yoshinori Hasegawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Japan
- Department of Respiratory Medicine, National Hospital Organization, Nagoya Medical Center, Japan
| |
Collapse
|
27
|
Armstrong DA, Lee MK, Hazlett HF, Dessaint JA, Mellinger DL, Aridgides DS, Hendricks GM, Abdalla MAK, Christensen BC, Ashare A. Extracellular Vesicles from Pseudomonas aeruginosa Suppress MHC-Related Molecules in Human Lung Macrophages. Immunohorizons 2020; 4:508-519. [PMID: 32819967 DOI: 10.4049/immunohorizons.2000026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/01/2020] [Indexed: 12/18/2022] Open
Abstract
Pseudomonas aeruginosa, a Gram-negative bacterium, is one of the most common pathogens colonizing the lungs of cystic fibrosis patients. P. aeruginosa secrete extracellular vesicles (EVs) that contain LPS and other virulence factors that modulate the host's innate immune response, leading to an increased local proinflammatory response and reduced pathogen clearance, resulting in chronic infection and ultimately poor patient outcomes. Lung macrophages are the first line of defense in the airway innate immune response to pathogens. Proper host response to bacterial infection requires communication between APC and T cells, ultimately leading to pathogen clearance. In this study, we investigate whether EVs secreted from P. aeruginosa alter MHC Ag expression in lung macrophages, thereby potentially contributing to decreased pathogen clearance. Primary lung macrophages from human subjects were collected via bronchoalveolar lavage and exposed to EVs isolated from P. aeruginosa in vitro. Gene expression was measured with the NanoString nCounter gene expression assay. DNA methylation was measured with the EPIC array platform to assess changes in methylation. P. aeruginosa EVs suppress the expression of 11 different MHC-associated molecules in lung macrophages. Additionally, we show reduced DNA methylation in a regulatory region of gene complement factor B (CFB) as the possible driving mechanism of widespread MHC gene suppression. Our results demonstrate MHC molecule downregulation by P. aeruginosa-derived EVs in lung macrophages, which is consistent with an immune evasion strategy employed by a prokaryote in a host-pathogen interaction, potentially leading to decreased pulmonary bacterial clearance.
Collapse
Affiliation(s)
- David A Armstrong
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756;
| | - Min Kyung Lee
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Haley F Hazlett
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - John A Dessaint
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Diane L Mellinger
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Daniel S Aridgides
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Gregory M Hendricks
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Moemen A K Abdalla
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21526, Egypt; and
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756.,Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Alix Ashare
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| |
Collapse
|
28
|
Fathima S, Zhang H. Histologic patterns of lung injury in patients using e-cigarettes. Proc (Bayl Univ Med Cent) 2020; 33:619-620. [PMID: 33100546 DOI: 10.1080/08998280.2020.1775052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
In recent years, e-cigarette use has become more popular. Until recently, it was considered safer than smoking. We report two cases of acute pulmonary illness associated with vaping, focusing on their histologic patterns.
Collapse
Affiliation(s)
- Samreen Fathima
- Department of Pathology, Baylor University Medical Center, Dallas, Texas
| | - Haiying Zhang
- Department of Pathology, Baylor University Medical Center, Dallas, Texas
| |
Collapse
|
29
|
Kulshrestha R, Dhanda H, Pandey A, Singh A, Kumar R. Immunopathogenesis and therapeutic potential of macrophage influx in diffuse parenchymal lung diseases. Expert Rev Respir Med 2020; 14:917-928. [PMID: 32600077 DOI: 10.1080/17476348.2020.1776117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION The diffuse parenchymal lung diseases (DPLD)/interstitial lung diseases (ILD) are progressive lung disorders with usually unclear etiology, poor long-term survival and no effective treatment. Their pathogenesis is characterized by alveolar epithelial cell injury, inflammation, epithelial-mesenchymal transition, and parenchymal fibrosis. Macrophages play diverse roles in their development, both in the acute phase and in tissue repair. AREAS COVERED In this review, we summarize the current state of knowledge regarding the role of macrophages and their phenotypes in the immunopathogenesis of DPLDs; CVD-ILD, UIP, NSIP, DIP, RB-ILD, AIP, HP, Sarcoidosis, etc. Our goal is to update the understanding of the immune mechanisms underlying the initiation and progression of fibrosis in DPLDs. This will help in identification of biomarkers and in developing novel therapeutic strategies for DPLDs. A thorough literature search of the published studies in PubMed (from 1975 to 2020) was done. EXPERT OPINION The macrophage associated inflammatory markers needs to be explored for their potential as biomarkers of disease activity and progression. Pharmacological targeting of macrophage activation may reduce the risk of macrophage activation syndrome (MAS) and help improving the survival and prognosis of these patients.
Collapse
Affiliation(s)
| | - Himanshu Dhanda
- Department of Pathology, V.P.Chest Institute , New Delhi, India
| | - Apoorva Pandey
- Department of Pathology, V.P.Chest Institute , New Delhi, India
| | - Amit Singh
- Department of Pathology, V.P.Chest Institute , New Delhi, India
| | - Raj Kumar
- Department of Pulmonary Medicine, V.P.Chest Institute , New Delhi, India
| |
Collapse
|
30
|
Eskes ECB, Sjouke B, Vaz FM, Goorden SMI, van Kuilenburg ABP, Aerts JMFG, Hollak CEM. Biochemical and imaging parameters in acid sphingomyelinase deficiency: Potential utility as biomarkers. Mol Genet Metab 2020; 130:16-26. [PMID: 32088119 DOI: 10.1016/j.ymgme.2020.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/19/2022]
Abstract
Acid Sphingomyelinase Deficiency (ASMD), or Niemann-Pick type A/B disease, is a rare lipid storage disorder leading to accumulation of sphingomyelin and its precursors primarily in macrophages. The disease has a broad phenotypic spectrum ranging from a fatal infantile form with severe neurological involvement (the infantile neurovisceral type) to a primarily visceral form with different degrees of pulmonary, liver, spleen and skeletal involvement (the chronic visceral type). With the upcoming possibility of treatment with enzyme replacement therapy, the need for biomarkers that predict or reflect disease progression has increased. Biomarkers should be validated for their use as surrogate markers of clinically relevant endpoints. In this review, clinically important endpoints as well as biochemical and imaging markers of ASMD are discussed and potential new biomarkers are identified. We suggest as the most promising biomarkers that may function as surrogate endpoints in the future: diffusion capacity measured by spirometry, spleen volume, platelet count, low-density lipoprotein cholesterol, liver fibrosis measured with a fibroscan, lysosphingomyelin and walked distance in six minutes. Currently, no biomarkers have been validated. Several plasma markers of lipid-laden cells, fibrosis or inflammation are of high potential as biomarkers and deserve further study. Based upon current guidelines for biomarkers, recommendations for the validation process are provided.
Collapse
Affiliation(s)
- Eline C B Eskes
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Barbara Sjouke
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Amsterdam UMC, University of Amsterdam, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Gastroenterology & Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Susan M I Goorden
- Amsterdam UMC, University of Amsterdam, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Gastroenterology & Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - André B P van Kuilenburg
- Amsterdam UMC, University of Amsterdam, Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Gastroenterology & Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Johannes M F G Aerts
- Leiden Institute of Chemistry, University of Leiden, Department of Medical Biochemistry, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Carla E M Hollak
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Vaping-Induced Lung Injury: A Case of Lipoid Pneumonia Associated with E-Cigarettes Containing Cannabis. Case Rep Pulmonol 2020; 2020:7151834. [PMID: 32309002 PMCID: PMC7157807 DOI: 10.1155/2020/7151834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/02/2020] [Accepted: 03/16/2020] [Indexed: 11/18/2022] Open
Abstract
Electronic cigarette, or vaping product use-associated lung injury (EVALI), is a group of lung disorders associated with vaping and e-cigarette products that has previously been categorized as a diagnosis of exclusion and best described as an exogenous lipoid pneumonia or chemical pneumonitis. Here, we describe the onset of an exogenous cause of lipoid pneumonia in an otherwise healthy patient using cannabis-containing electronic cigarettes. We explore similarities in the clinical case, define a common clinical presentation with progression of disease, characteristic radiographic findings along with pathological diagnosis and management.
Collapse
|
32
|
Dillon K, Jochims K, Gerigk U, Jost F, Kobesen H, Bialucha R. No pathogenic responses in rat lung upon exposure to ground granulated blast furnace slag (GGBS). Inhal Toxicol 2020; 32:39-52. [PMID: 32122189 DOI: 10.1080/08958378.2020.1731023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objective: Granulated blast furnace slag (GBS) is a by-product of the manufacture of iron by thermochemical reduction in a blast furnace. Blast furnace slag is generated at temperatures above 1500 °C. If the liquid slag is quenched very rapidly with water, a glassy slag is generated (GBS). It is used - after grinding <100 µm - [ground granulated blast furnace slag (GGBS)] for cement and concrete production. A small particulate fraction of GGBS might be accessible to the pulmonary alveoli, where it could settle down and induces physiological inflammatory responses. Within the scope of the 'Registration, Evaluation, Authorization and Restriction of Chemicals' (REACH), GGBS was already tested in rats in an acute toxicity inhalation study, as well as in a dose range finding study as a predecessor study for this study. Both did not show systemic and local toxic effect in rats upon inhalation of high-dose GGBS.Material and methods: In this study, low (4.3 mg/m3), intermediate (9.5 mg/m3), and high-dose (24.9 mg/m3) repetitive exposure of GGBS to rats was tested over a period of 4 weeks with 6 h exposure per day for 5 days per week. Results and conclusion: Even at high doses, GGBS was inactive and did not induce clinically relevant phenotypic changes in rats compared to concomitant controls.Together with both the previous acute toxicity and the dose range finding study in rats, it was shown that the exposure to the tested GGBS was unable to induce any severe pathogenic responses.
Collapse
Affiliation(s)
- Kevin Dillon
- Charles River Laboratories Preclinical Services, Edinburgh, UK
| | | | | | - Franz Jost
- Stahl Holding Saar, Dillingen/Saar, Germany
| | | | - Ruth Bialucha
- FEhS - Institut für Baustoff-Forschung e.V., Duisburg, Germany
| |
Collapse
|
33
|
Thakrar PD, Boyd KP, Swanson CP, Wideburg E, Kumbhar SS. E-cigarette, or vaping, product use-associated lung injury in adolescents: a review of imaging features. Pediatr Radiol 2020; 50:338-344. [PMID: 31897566 DOI: 10.1007/s00247-019-04572-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/22/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND There has been a recent increase in recognition of lung disease related to the use of electronic cigarettes (called "vaping"). These patients present with acute respiratory illness following exposure to vaporized cannabis or nicotine products and sometimes require hospitalization and intensive care. We describe the imaging findings of this disease entity in the pediatric population. OBJECTIVE To describe the radiologic findings of lung injury associated with electronic cigarette use (vaping) in the adolescent pediatric population. MATERIALS AND METHODS We identified all adolescents with acute respiratory illness and a history of electronic cigarette use who presented at our institution within a 3-month period (June 2019 through August 2019). We excluded adolescents with potential intercurrent pulmonary disease. We reviewed the charts for symptomatology and laboratory and pathology data. In addition, we reviewed the chest radiographs and chest CTs of these adolescents. RESULTS The review group consisted of 12 teenage pediatric patients (10 boys and 2 girls; mean age 16.9 years, range 16.0-17.7 years) with acute respiratory illness found to have a temporal association with electronic cigarette use for cannabis products, nicotine, or both. Other etiologies for illness in these adolescents had been excluded by clinical and laboratory evaluation. All of the adolescents were admitted to the hospital for treatment. The clinical presentations included dyspnea, abdominal pain and constitutional symptoms. Pulmonary function testing that was performed in all patients during admission or follow-up demonstrated reduced diffusion capacity in 4/12 (33%), an obstructive ventilatory pattern in 4/12 (33%), a restrictive pattern in 1/12 (8%), and a mixed obstructive and restrictive pattern in 2/12 (17%) adolescents. Bronchoalveolar lavage studies, performed in 9 of the 12 adolescents, revealed inflammatory cells and lipid-laden macrophages. All of the patients underwent CT of the chest; the findings were notable for centrilobular ground-glass nodules (11/12; 92%) and confluent ground-glass opacities (12/12; 100%), with frequent subpleural sparing (9/12; 75%). Additionally, 6/12 (50%) adolescents demonstrated small pleural effusions; 6/12 (50%) had mild bronchial wall thickening; 9/12 (75%) had enlarged hilar or mediastinal lymph nodes; and 2/12 (17%) had a small pericardial effusion. CONCLUSION As seen in our teenage population, e-cigarette, or vaping, product use-associated lung injury (EVALI) is characterized by centrilobular ground-glass nodules and ground-glass opacities with subpleural sparing. The imaging findings are most consistent with acute lung injury resulting from toxic inhalation. Because adolescent pediatric patients might not be forthcoming with their history of electronic cigarette use, it is important for the pediatric radiologist to be aware of the imaging patterns of this disease.
Collapse
Affiliation(s)
- Pooja D Thakrar
- Department of Pediatric Radiology, Children's Hospital of Wisconsin and the Medical College of Wisconsin,, 9000 W. Wisconsin Ave., MS-721, Milwaukee, WI, 53226, USA
| | - Kevin P Boyd
- Department of Pediatric Radiology, Children's Hospital of Wisconsin and the Medical College of Wisconsin,, 9000 W. Wisconsin Ave., MS-721, Milwaukee, WI, 53226, USA
| | - Craig P Swanson
- Department of Radiology, Aurora St. Luke's Hospital, Milwaukee, WI, USA
| | - Eric Wideburg
- Department of Radiology, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sachin S Kumbhar
- Department of Pediatric Radiology, Children's Hospital of Wisconsin and the Medical College of Wisconsin,, 9000 W. Wisconsin Ave., MS-721, Milwaukee, WI, 53226, USA.
| |
Collapse
|
34
|
Jaćević V, Nepovimova E, Kuča K. Acute Toxic Injuries of Rat's Visceral Tissues Induced by Different Oximes. Sci Rep 2019; 9:16425. [PMID: 31712702 PMCID: PMC6848205 DOI: 10.1038/s41598-019-52768-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 10/23/2019] [Indexed: 12/27/2022] Open
Abstract
Certain AChE reactivators, asoxime, obidoxime, K027, K048, and K075, when taken in overdoses and sometimes even when introduced within therapeutic ranges, may injure the different organs. As a continuation of previously published data, in this study, Wistar rats have sacrificed 24 hrs and 7 days after single im application of 0.1LD50, 0.5LD50 and 1.0LD50 of each reactivator, and examinated tissue samples were obtained for pathohistological and semiquantitative analysis. A severity of tissue alteration, expressed as different tissue damage scores were evaluated. Morphological structure of examinated tissues treated with of 0.1LD50 of all reactivators was comparable with the control group of rats. Moderate injuries were seen in visceral tissues treated with 0.5LD50 of asoxime, obidoxime and K027. Acute damages were enlarged after treatment with 0.5LD50 and 1.0LD50 of all reactivators during the next 7 days. The most prominent changes were seen in rats treated with 1.0LD50 of K048 and K075 (P < 0.001 vs. control and asoxime-treated group). All reactivators given by a single, high, unitary dose regimen, have an adverse effect not only on the main visceral tissue, but on the whole rat as well, but the exact mechanism of cellular injury remains to be confirmed in further investigation.
Collapse
Affiliation(s)
- Vesna Jaćević
- National Poison Control Centre, Military Medical Academy, Belgrade, Serbia.,Faculty of Medicine of the Military Medical Academy, University of Defense, Belgrade, Serbia.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czechia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czechia
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czechia.
| |
Collapse
|
35
|
Respiratory complications of metabolic disease in the paediatric population: A review of presentation, diagnosis and therapeutic options. Paediatr Respir Rev 2019; 32:55-65. [PMID: 31101546 DOI: 10.1016/j.prrv.2019.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/21/2022]
Abstract
Inborn errors of metabolism (IEMs) whilst individually rare, as a group constitute a field which is increasingly demands on pulmonologists. With the advent of new therapies such as enzyme replacement and gene therapy, early diagnosis and treatment of these conditions can impact on long term outcome, making their timely recognition and appropriate investigation increasingly important. Conversely, with improved treatment, survival of these patients is increasing, with the emergence of previously unknown respiratory phenotypes. It is thus important that pulmonologists are aware of and appropriately monitor and manage these complications. This review aims to highlight the respiratory manifestations which can occur. It isdivided into conditions resulting primarily in obstructive airway and lung disease, restrictive lung disease such as interstitial lung disease or pulmonary alveolar proteinosis and pulmonary hypertension, whilst acknowledging that some diseases have the potential to cause all three. The review focuses on general phenotypes of IEMs, their known respiratory complications and the basic metabolic investigations which should be performed where an IEM is suspected.
Collapse
|
36
|
Armstrong DA, Chen Y, Dessaint JA, Aridgides DS, Channon JY, Mellinger DL, Christensen BC, Ashare A. DNA Methylation Changes in Regional Lung Macrophages Are Associated with Metabolic Differences. Immunohorizons 2019; 3:274-281. [PMID: 31356157 PMCID: PMC6686200 DOI: 10.4049/immunohorizons.1900042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 12/21/2022] Open
Abstract
A number of pulmonary diseases occur with upper lobe predominance, including cystic fibrosis and smoking-related chronic obstructive pulmonary disease. In the healthy lung, several physiologic and metabolic factors exhibit disparity when comparing the upper lobe of the lung to lower lobe, including differences in oxygenation, ventilation, lymphatic flow, pH, and blood flow. In this study, we asked whether these regional differences in the lung are associated with DNA methylation changes in lung macrophages that could potentially lead to altered cell responsiveness upon subsequent environmental challenge. All analyses were performed using primary lung macrophages collected via bronchoalveolar lavage from healthy human subjects with normal pulmonary function. Epigenome-wide DNA methylation was examined via Infinium MethylationEPIC (850K) array and validated by targeted next-generation bisulfite sequencing. We observed 95 CpG loci with significant differential methylation in lung macrophages, comparing upper lobe to lower lobe (all false discovery rate < 0.05). Several of these genes, including CLIP4, HSH2D, NR4A1, SNX10, and TYK2, have been implicated as participants in inflammatory/immune-related biological processes. Functionally, we identified phenotypic differences in oxygen use, comparing upper versus lower lung macrophages. Our results support a hypothesis that epigenetic changes, specifically DNA methylation, at a multitude of gene loci in lung macrophages are associated with metabolic differences regionally in lung.
Collapse
Affiliation(s)
- David A Armstrong
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756;
| | - Youdinghuan Chen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - John A Dessaint
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Daniel S Aridgides
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Jacqueline Y Channon
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756; and
| | - Diane L Mellinger
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756.,Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756.,Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Alix Ashare
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756; .,Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756; and
| |
Collapse
|
37
|
Capron T, Trigui Y, Gautier C, Puech B, Chanez P, Reynaud-Gaubert M. Respiratory impairment in Niemann-Pick B disease: Two case reports and review for the pulmonologist. Respir Med Res 2019; 76:13-18. [PMID: 31254945 DOI: 10.1016/j.resmer.2019.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/04/2019] [Accepted: 05/06/2019] [Indexed: 11/18/2022]
Abstract
Acid sphingomyelinase deficiency (ASMD), also called Niemann-Pick disease, is a storage disorder with pulmonary involvement but few respiratory symptoms in adults. However, the disease may evolve towards clinically relevant respiratory symptoms with referral to the pulmonologist for management and care. Based on two case reports illustrating respiratory impairment, the aim of this work was to review clinical features, diagnosis, respiratory prognostic and therapeutics for the pulmonologist. Overall, storage disorder should be suspected in the presence of hepatosplenomegaly and interstitial lung disease. Concomitant thrombopenia or hyperlipidemia should also draw attention. Following recent consensus guidelines, diagnosis is based on enzyme assay for ASM activity in blood, with subsequent gene sequencing once the biochemical diagnosis has been confirmed. Disease is slowly progressive and the main causes of death are respiratory and liver failure. Presence of emphysema lesions or worsening of respiratory symptoms should call for the intensification of treatment. Though enzyme replacement therapy is a promising way of development, lung transplantation might be considered for these patients in the absence of contraindication.
Collapse
Affiliation(s)
- T Capron
- Centre de compétences national maladies pulmonaires rares, équipe de transplantation pulmonaire, Aix-Marseille université, CHU Nord, Assistance publique-Hôpitaux de Marseille, 13915 Marseille, France.
| | - Y Trigui
- Clinique des bronches, allergies et sommeil, Aix-Marseille université, CHU Nord, Assistance publique-Hôpitaux de Marseille, 13915 Marseille, France
| | - C Gautier
- Centre de compétences national maladies pulmonaires rares, équipe de transplantation pulmonaire, Aix-Marseille université, CHU Nord, Assistance publique-Hôpitaux de Marseille, 13915 Marseille, France
| | - B Puech
- Service de radiologie, Aix-Marseille université, CHU Nord, Assistance publique-Hôpitaux de Marseille, 13915 Marseille, France
| | - P Chanez
- Clinique des bronches, allergies et sommeil, Aix-Marseille université, CHU Nord, Assistance publique-Hôpitaux de Marseille, 13915 Marseille, France
| | - M Reynaud-Gaubert
- Centre de compétences national maladies pulmonaires rares, équipe de transplantation pulmonaire, Aix-Marseille université, CHU Nord, Assistance publique-Hôpitaux de Marseille, 13915 Marseille, France
| |
Collapse
|
38
|
Molecular Pathways and Respiratory Involvement in Lysosomal Storage Diseases. Int J Mol Sci 2019; 20:ijms20020327. [PMID: 30650529 PMCID: PMC6359090 DOI: 10.3390/ijms20020327] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/25/2022] Open
Abstract
Lysosomal storage diseases (LSD) include a wide range of different disorders with variable degrees of respiratory system involvement. The purpose of this narrative review is to treat the different types of respiratory manifestations in LSD, with particular attention being paid to the main molecular pathways known so far to be involved in the pathogenesis of the disease. A literature search was conducted using the Medline/PubMed and EMBASE databases to identify studies, from 1968 through to November 2018, that investigated the respiratory manifestations and molecular pathways affected in LSD. Pulmonary involvement includes interstitial lung disease in Gaucher’s disease and Niemann-Pick disease, obstructive airway disease in Fabry disease and ventilatory disorders with chronic respiratory failure in Pompe disease due to diaphragmatic and abdominal wall muscle weakness. In mucopolysaccharidosis and mucolipidoses, respiratory symptoms usually manifest early in life and are secondary to anatomical malformations, particularly of the trachea and chest wall, and to accumulation of glycosaminoglycans in the upper and lower airways, causing, for example, obstructive sleep apnea syndrome. Although the molecular pathways involved vary, ranging from lipid to glycogen and glycosaminoglycans accumulation, some clinical manifestations and therapeutic approaches are common among diseases, suggesting that lysosomal storage and subsequent cellular toxicity are the common endpoints.
Collapse
|
39
|
Abstract
Idiopathic pulmonary fibrosis (IPF) is an extremely aggressive lung disease that develops almost exclusively in older individuals, carries a very poor prognosis, and lacks any truly effective therapies. The current conceptual model is that IPF develops because of an age-related decline in the ability of the lung epithelium to regenerate after injury, largely due to death or senescence of epithelial progenitor cells in the distal airways. This loss of regenerative capacity is thought to initiate a chronic and ineffective wound-healing response, characterized by persistent, low-grade lung inflammation and sustained production of collagen and other extracellular matrix materials. Despite recent advances in our understanding of IPF pathobiology, there remains a pressing need to further delineate underlying mechanisms to develop more effective therapies for this disease. In this review, we build the case that many of the manifestations of IPF result from a failure of cells to effectively manage their proteome. We propose that epithelial progenitor cells, as well as immune cells and fibroblasts, become functionally impaired, at least in part, because of an accumulation or a loss in the expression of various crucial proteins. Further, we propose that central to this defect is the dysregulation of the ubiquitin-proteasome system (UPS), which is the major protein-degradation system in eukaryotic cells. Lastly, borrowing concepts from other fields, we discuss how targeting the UPS system could be employed as a novel treatment for IPF and perhaps for other fibrotic lung diseases as well.
Collapse
Affiliation(s)
- Willy Roque
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Freddy Romero
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
40
|
Dorfmüller P, Cavazza A. Lung pathology for the clinician: a comprehensive approach. Eur Respir Rev 2017; 26:26/144/170041. [PMID: 28659505 DOI: 10.1183/16000617.0041-2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/29/2017] [Indexed: 11/05/2022] Open
Affiliation(s)
- Peter Dorfmüller
- Dept of Pathology, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Alberto Cavazza
- Pathology Unit, Arcispedale S. Maria Nuova/IRCCS, Reggio Emilia, Italy
| |
Collapse
|