1
|
Vázquez Marrero VR, Dresler M, Haggadone MD, Lu A, Shin S. GM-CSF engages multiple signaling pathways to enhance pro-inflammatory cytokine responses in human monocytes during Legionella infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627084. [PMID: 39713445 PMCID: PMC11661061 DOI: 10.1101/2024.12.05.627084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The proinflammatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) is required for host defense against a wide range of pathogens. We previously found that GM-CSF enhances inflammatory cytokine production in murine monocytes and is required for in vivo control of the intracellular bacterial pathogen Legionella pneumophila . It is unclear whether GM-CSF similarly augments cytokine production in human monocytes during bacterial infection. Here, we find that GM-CSF enhances inflammatory cytokine expression in Legionella- infected human monocytes by engaging multiple signaling pathways. Legionella - and TLR-dependent NF-𝜅B signaling is a prerequisite signal for GM-CSF to promote cytokine expression. Then, GM-CSF-driven JAK2/STAT5 signaling is required to augment cytokine expression in Legionella -infected human monocytes. We also found a role for PI-3K/Akt/mTORC1 signaling in GM-CSF-dependent upregulation of cytokine expression. Finally, glycolysis and amino acid metabolism are also critical for GM-CSF to boost cytokine gene expression in human monocytes during infection. Our findings show that GM-CSF-mediated enhancement of cytokine expression in infected human monocytes is regulated by multiple signaling pathways, thereby allowing the host to fine tune antibacterial immunity.
Collapse
|
2
|
Hansakon A, Khampoongern R, Schiller L, Jeerawattanawart S, Angkasekwinai P. Effect of intranasal administration of Granulocyte-Macrophage Colony-Stimulating Factor on pulmonary Cryptococcus gattii infection. Int Immunopharmacol 2024; 142:113259. [PMID: 39332096 DOI: 10.1016/j.intimp.2024.113259] [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: 05/14/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Cryptococcosis, caused by infections with C. neoformans and C. gattii, presents a serious threat to global health and necessitates effective treatment strategies. Granulocyte-Macrophage Colony-Stimulating Factor, GM-CSF, is an immune-modulating cytokine that has been utilized clinically to improve host defense against infection; however, the impact of GM-CSF treatment in C. gattii infection has not been elucidated. Our current study aimed to investigate the effect of GM-CSF treatment on pulmonary immune response during C. gattii infection. In response to C. gattii infection, GM-CSF-expressing T helper cells and CD11b+ myeloid were enhanced in the lungs. The intranasal administration of GM-CSF during C. gattii infection significantly reduced pulmonary cryptococcal load, promoted an increase in pulmonary Th17 cells, as well as neutrophil infiltration in the lungs. Exposure of neutrophils to C. gattii in the presence of GM-CSF resulted in an increased neutrophil phagocytosis and fungal killing capacity, generation of reactive oxygen species (ROS), and upregulation of inflammatory cytokines and anti-microbial peptides. Although GM-CSF treatment in C. neoformans-infected mice had a comparable impact on the reduction of lung fungal burden, it resulted in the enhancement of Th1-type cytokine IFN-γ and the activation of M1 macrophages. Altogether, this study demonstrated that the intranasal delivery of GM-CSF has distinct effects on promoting the protection against C. gattii and C. neoformans by activating neutrophil/type-17 immune response and stimulating M1 macrophage/type-1 immunity, respectively.
Collapse
Affiliation(s)
- Adithap Hansakon
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120 Thailand; Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani 12120 Thailand
| | - Rungwadee Khampoongern
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani 12120 Thailand
| | - Lauritz Schiller
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani 12120 Thailand; Carl Von Ossietzky University of Oldenburg, Faculty VI - Medicine and Health Sciences, Oldenburg, Germany
| | - Siranart Jeerawattanawart
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani 12120 Thailand
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani 12120 Thailand; Research Unit in Molecular Pathogenesis and Immunology of Infectious Diseases, Thammasat University, Pathum Thani 12120, Thailand.
| |
Collapse
|
3
|
Obi ON, Saketkoo LA, Maier LA, Baughman RP. Developmental drugs for sarcoidosis. J Autoimmun 2024; 149:103179. [PMID: 38548579 DOI: 10.1016/j.jaut.2024.103179] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/04/2023] [Accepted: 02/08/2024] [Indexed: 12/15/2024]
Abstract
Sarcoidosis is a multi-organ granulomatous inflammatory disease of unknown etiology. Over 50% of patients will require treatment at some point in their disease and 10%-30% will develop a chronic progressive disease with pulmonary fibrosis leading to significant morbidity and mortality. Recently published guidelines recommend immunosuppressive therapy for sarcoidosis patients at risk of increased disease-related morbidity and mortality, and in whom disease has negatively impacted quality of life. Prednisone the currently recommended first line therapy is associated with significant toxicity however none of the other guideline recommended steroid sparing therapy is approved by regulatory agencies for use in sarcoidosis, and data in support of their use is weak. For patients with severe refractory disease requiring prolonged therapy, treatment options are limited. The need for expanding treatment options in sarcoidosis has been emphasized. Well conducted large, randomized trials evaluating currently available therapeutic options as well as novel pathways for targeting disease are necessary to better guide treatment decisions. These trials will not be without significant challenges. Sarcoidosis is a rare disease with heterogenous presentation and variable progression and clinical outcome. There are no universally agreed upon biomarkers of disease activity and measurement of outcomes is confounded by the need to balance patient centric measures and objective measures of disease activity. Our paper provides an update on developmental drugs in sarcoidosis and outlines several novel pathways that may be targeted for future drug development. Currently available trials are highlighted and ongoing challenges to drug development and clinical trial design are briefly discussed.
Collapse
Affiliation(s)
- Ogugua Ndili Obi
- Division of Pulmonary Critical Care and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| | - Lesley Ann Saketkoo
- New Orleans Scleroderma and Sarcoidosis Patient Care and Research Center, New Orleans, USA; University Medical Center - Comprehensive Pulmonary Hypertension Center and Interstitial Lung Disease Clinic Programs, New Orleans, USA; Louisiana State University School of Medicine, Section of Pulmonary Medicine, New Orleans, LA, USA; Tulane University School of Medicine, Undergraduate Honors Department, New Orleans, LA, USA
| | - Lisa A Maier
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA; Division of Pulmonary and Critical Care Sciences, Department of Medicine, University of Colorado School of Medicine, Denver, CO, USA
| | - Robert P Baughman
- Emeritus Professor of Medicine, Department of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
4
|
Von Stemann JH, Dungu AM, Laguarda MV, Ryrsø CK, Hegelund MH, Faurholt-Jepsen D, Krogh-Madsen R, Hansen MB, Lindegaard B, Ostrowski SR. Autoantibodies targeting interferons and GM-CSF are associated with adverse outcome risk, comorbidities, and pathogen in community-acquired pneumonia. Front Immunol 2024; 15:1459616. [PMID: 39606243 PMCID: PMC11598332 DOI: 10.3389/fimmu.2024.1459616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/04/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Cytokine autoantibodies (c-aAb) have been associated with pulmonary diseases, including severe novel coronavirus disease 2019 (COVID-19) and pulmonary alveolar proteinosis. This study aimed to determine c-aAb association with community-acquired pneumonia (CAP) etiology (SARS-CoV-2, influenza, or bacteria) and c-aAb associations with CAP-related clinical outcomes and pulmonary comorbidities. Methods In a cohort of 665 patients hospitalized with CAP, c-aAb targeting interferon α (IFNα), IFNβ, IFNγ, interleukin-1α (IL-1α), IL-6, IL-10, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were measured in plasma samples. Associations between c-aAb and baseline characteristics, pulmonary comorbidities, pathogen, intensive care unit (ICU) transferal, time to clinical stability, and mortality were estimated, with results stratified by sex. Results More men infected with SARS-CoV-2 were had high-titer type 1 IFN c-aAb compared to other pathogens. Among patients with CAP, asthma and bronchiectasis comorbidities were associated with high-titer GM-CSF c-aAb in men, and men with high-titer IFNβ c-aAb had increased odds for ICU transferal. High-titer IL-10 c-aAb were associated with faster clinical stability in women. Conclusion In men with CAP, various c-aAb-including type 1 IFN and GM-CSF c-aAb-were associated with adverse clinical events and comorbidities, whereas c-aAb targeting an autoinflammatory cytokine were associated with a positive outcome in women. This suggests that the potentially immunomodulatory effects of c-aAb depend on pathogen, autoantibody specificity, comorbidity, and sex.
Collapse
Affiliation(s)
- Jakob Hjorth Von Stemann
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Arnold Matovu Dungu
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
| | - Maria Vispe Laguarda
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Camilla Koch Ryrsø
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Centre for Physical Activity Research, Copenhagen University Hospital, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Maria Hein Hegelund
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Daniel Faurholt-Jepsen
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Rikke Krogh-Madsen
- Centre for Physical Activity Research, Copenhagen University Hospital, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Bagge Hansen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Lindegaard
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
- Centre for Physical Activity Research, Copenhagen University Hospital, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| |
Collapse
|
5
|
Dharamdasani Detaram H, Nguyen PH, Wong VC, Loh H, Mansberg R. Nocardia masquerading as pulmonary malignancy in a patient with adult-onset immunodeficiency on 18F-FDG PET/CT. Radiol Case Rep 2024; 19:4517-4521. [PMID: 39188623 PMCID: PMC11345279 DOI: 10.1016/j.radcr.2024.07.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 08/28/2024] Open
Abstract
A 77-year-old man with a history of left nephrectomy for renal cell carcinoma and partial hepatectomy for cholangiocarcinoma underwent 18F-FDG PET/CT for assessment of an irregular lung lesion. FDG-PET demonstrated development of an intensely avid spiculated left lower lobe pulmonary lesion and intensely avid left pulmonary hilar nodes, raising suspicion for a malignancy. Eleven days following the PET study, the patient was admitted to hospital with an altered mental state. CT brain revealed diffuse round hyperdensities within the brain parenchyma. Microbiology of the lung lesion was positive for Nocardia Beijingensis and he was subsequently diagnosed with disseminated nocardiosis.
Collapse
Affiliation(s)
| | | | - Veronica C. Wong
- Nuclear Medicine and PET, Nepean Hospital, Penrith
- Sydney Medical School, The University of Sydney, Sydney
| | - Han Loh
- Nuclear Medicine and PET, Nepean Hospital, Penrith
- Sydney Medical School, The University of Sydney, Sydney
| | - Robert Mansberg
- Nuclear Medicine and PET, Nepean Hospital, Penrith
- Sydney Medical School, The University of Sydney, Sydney
| |
Collapse
|
6
|
Hamaguchi T, Uchida N, Fujita-Nakata M, Nakanishi M, Tsuchido Y, Nagao M, Iinuma Y, Asahina M. Autochthonous Cryptococcus gattii genotype VGIIb infection in a Japanese patient with anti-granulocyte-macrophage colony-stimulating factor antibodies. J Infect Chemother 2024; 30:1069-1075. [PMID: 38479572 DOI: 10.1016/j.jiac.2024.03.009] [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: 11/24/2023] [Revised: 03/01/2024] [Accepted: 03/09/2024] [Indexed: 03/17/2024]
Abstract
A 31-year-old Japanese man presented with cerebral and pulmonary cryptococcosis. Cryptococcus gattii (C. gattii) genotype VGIIb was detected in the patient's sputum and cerebrospinal fluid specimens. The serum levels of anti-granulocyte-macrophage colony-stimulating factor (GM-CSF) antibodies were elevated in this patient, which has been associated with pulmonary alveolar proteinosis and is considered a risk factor for C. gattii infection. After undergoing >12 months of antifungal treatments, the patient showed improvements in symptoms and findings on brain and lung imaging. Several Japanese patients who develop C. gattii infection have also been reported; however, most of these patients have been infected outside Japan, as C. gattii infection is rare in Japan. Only one patient with C. gattii genotype VGIIb infection has been reported in Japan, and it is believed that this patient contracted the infection in China. In the present case, our patient has never been outside Japan, indicating that the infection originated in Japan. Our findings suggest that C. gattii might be spreading in Japan. Therefore, patients with positive serum anti-GM-CSF antibodies should be thoroughly monitored for C. gattii infection, even those living in Japan.
Collapse
Affiliation(s)
| | - Nobuaki Uchida
- Department of Neurology, Kanazawa Medical University, Ishikawa, Japan.
| | | | - Megumi Nakanishi
- Department of Neurology, Kanazawa Medical University, Ishikawa, Japan.
| | - Yasuhiro Tsuchido
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Yoshitsugu Iinuma
- Department of Infectious Diseases, Kanazawa Medical University, Ishikawa, Japan.
| | - Masato Asahina
- Department of Neurology, Kanazawa Medical University, Ishikawa, Japan.
| |
Collapse
|
7
|
Wołoszczak J, Wrześniewska M, Hrapkowicz A, Janowska K, Szydziak J, Gomułka K. A Comprehensive Outlook on Pulmonary Alveolar Proteinosis-A Review. Int J Mol Sci 2024; 25:7092. [PMID: 39000201 PMCID: PMC11241585 DOI: 10.3390/ijms25137092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is an ultra-rare disease caused by impaired pulmonary surfactant clearance due to the dysfunction of alveolar macrophages or their signaling pathways. PAP is categorized into autoimmune, congenital, and secondary PAP, with autoimmune PAP being the most prevalent. This article aims to present a comprehensive review of PAP classification, pathogenesis, clinical presentation, diagnostics, and treatment. The literature search was conducted using the PubMed database and a total of 67 articles were selected. The PAP diagnosis is usually based on clinical symptoms, radiological imaging, and bronchoalveolar lavage, with additional GM-CSF antibody tests. The gold standard for PAP treatment is whole-lung lavage. This review presents a summary of the most recent findings concerning pulmonary alveolar proteinosis, pointing out specific features that require further investigation.
Collapse
Affiliation(s)
- Julia Wołoszczak
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Martyna Wrześniewska
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Aleksandra Hrapkowicz
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Kinga Janowska
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Joanna Szydziak
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
8
|
Brugnoli B, Salvati L, Di Lauria N, Botta A, Tozzetti C, Biscarini A, Capone M, Ferrentino F, Naldi C, Ascione G, Mazzoni A, Maggi L, Campo I, Carey B, Trapnell B, Liotta F, Cosmi L, Bartoloni A, Annunziato F, Parronchi P, Palterer B. Disseminated nocardiosis and anti-GM-CSF antibodies. Eur J Clin Microbiol Infect Dis 2024; 43:1003-1007. [PMID: 38379052 DOI: 10.1007/s10096-024-04785-z] [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: 07/10/2023] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
Infections that are unusually severe or caused by opportunistic pathogens are a hallmark of primary immunodeficiency (PID). Anti-cytokine autoantibodies (ACA) are an emerging cause of acquired immunodeficiency mimicking PID. Nocardia spp. are Gram-positive bacteria generally inducing disseminated infections in immunocompromised patients, but seldom also occurring in apparently immunocompetent hosts. Anti-GM-CSF autoantibodies are associated with autoimmune pulmonary alveolar proteinosis (PAP). In those patients, an increased incidence of disseminated nocardiosis and cryptococcosis has been observed. It is unclear whether the PAP or the autoantibodies predispose to the infection. We report an apparently immunocompetent woman presenting with disseminated nocardiosis without any evidence of PAP. Clinical data and radiological images were retrospectively collected. Lymphocyte populations were analyzed by flow cytometry. Anti-GM-CSF autoantibodies were measured by ELISA. A 55-year-old otherwise healthy woman presented with cerebral and pulmonary abscesses. Personal and familial history of infections or autoimmunity were negative. After extensive examinations, a final diagnosis of disseminated nocardiosis was made. Immunologic investigations including neutrophilic function and IFN-γ/IL-12 circuitry failed to identify a PID. Whole-exome sequencing did not find pathogenic variants associated with immunodeficiency. Serum anti-GM-CSF autoantibodies were positive. There were no clinical or instrumental signs of PAP. Trimethoprim-sulfamethoxazole and imipenem were administered, with progressive improvement and recovery of the infectious complication. We identified anti-GM-CSF autoantibodies as the cause of disseminated nocardiosis in a previously healthy and apparently immunocompetent adult. This case emphasizes the importance of including ACA in the differential diagnosis of PID, especially in previously healthy adults. Importantly, anti-GM-CSF autoantibodies can present with disseminated nocardiosis without PAP.
Collapse
Affiliation(s)
- Barbara Brugnoli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Nicoletta Di Lauria
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Annarita Botta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Camilla Tozzetti
- Internal Medicine Unit 3, Careggi University Hospital, Florence, Italy
| | - Alessandro Biscarini
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence-Careggi University Hospital, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Chiara Naldi
- Internal Medicine Unit 3, Careggi University Hospital, Florence, Italy
| | - Giovanni Ascione
- Internal Medicine Unit 3, Careggi University Hospital, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Ilaria Campo
- Pneumology Unit, Internal Medicine and Infectious Diseases Department, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Brenna Carey
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bruce Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Paola Parronchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Boaz Palterer
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
9
|
Casanova JL, Peel J, Donadieu J, Neehus AL, Puel A, Bastard P. The ouroboros of autoimmunity. Nat Immunol 2024; 25:743-754. [PMID: 38698239 DOI: 10.1038/s41590-024-01815-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/13/2024] [Indexed: 05/05/2024]
Abstract
Human autoimmunity against elements conferring protective immunity can be symbolized by the 'ouroboros', a snake eating its own tail. Underlying infection is autoimmunity against three immunological targets: neutrophils, complement and cytokines. Autoantibodies against neutrophils can cause peripheral neutropenia underlying mild pyogenic bacterial infections. The pathogenic contribution of autoantibodies against molecules of the complement system is often unclear, but autoantibodies specific for C3 convertase can enhance its activity, lowering complement levels and underlying severe bacterial infections. Autoantibodies neutralizing granulocyte-macrophage colony-stimulating factor impair alveolar macrophages, thereby underlying pulmonary proteinosis and airborne infections, type I interferon viral diseases, type II interferon intra-macrophagic infections, interleukin-6 pyogenic bacterial diseases and interleukin-17A/F mucocutaneous candidiasis. Each of these five cytokine autoantibodies underlies a specific range of infectious diseases, phenocopying infections that occur in patients with the corresponding inborn errors. In this Review, we analyze this ouroboros of immunity against immunity and posit that it should be considered as a factor in patients with unexplained infection.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France.
- Paris Cité University, Imagine Institute, Paris, France.
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
| | - Jessica Peel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, New York, NY, USA
| | - Jean Donadieu
- Trousseau Hospital for Sick Children, Centre de référence des neutropénies chroniques, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| |
Collapse
|
10
|
Bastard P, Gervais A, Le Voyer T, Philippot Q, Cobat A, Rosain J, Jouanguy E, Abel L, Zhang SY, Zhang Q, Puel A, Casanova JL. Human autoantibodies neutralizing type I IFNs: From 1981 to 2023. Immunol Rev 2024; 322:98-112. [PMID: 38193358 PMCID: PMC10950543 DOI: 10.1111/imr.13304] [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] [Indexed: 01/10/2024]
Abstract
Human autoantibodies (auto-Abs) neutralizing type I IFNs were first discovered in a woman with disseminated shingles and were described by Ion Gresser from 1981 to 1984. They have since been found in patients with diverse conditions and are even used as a diagnostic criterion in patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1). However, their apparent lack of association with viral diseases, including shingles, led to wide acceptance of the conclusion that they had no pathological consequences. This perception began to change in 2020, when they were found to underlie about 15% of cases of critical COVID-19 pneumonia. They have since been shown to underlie other severe viral diseases, including 5%, 20%, and 40% of cases of critical influenza pneumonia, critical MERS pneumonia, and West Nile virus encephalitis, respectively. They also seem to be associated with shingles in various settings. These auto-Abs are present in all age groups of the general population, but their frequency increases with age to reach at least 5% in the elderly. We estimate that at least 100 million people worldwide carry auto-Abs neutralizing type I IFNs. Here, we briefly review the history of the study of these auto-Abs, focusing particularly on their known causes and consequences.
Collapse
Affiliation(s)
- Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistante Publique-Hôpitaux de Paris (AP-HP), Paris, France, EU
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Qian Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, APHP, Paris, France, EU
| |
Collapse
|
11
|
Dougan M, Nguyen LH, Buchbinder EI, Lazarus HM. Sargramostim for Prophylactic Management of Gastrointestinal Immune-Related Adverse Events of Immune Checkpoint Inhibitor Therapy for Cancer. Cancers (Basel) 2024; 16:501. [PMID: 38339253 PMCID: PMC10854719 DOI: 10.3390/cancers16030501] [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: 12/20/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy improves outcomes in several cancers. Unfortunately, many patients experience grade 3-4 treatment-related adverse events, including gastrointestinal (GI) toxicities which are common. These GI immune-related adverse events (irAEs) induced by ICIs present significant clinical challenges, require prompt intervention, and result in treatment delays or discontinuations. The treatment for these potentially severe and even fatal GI irAEs which include enterocolitis, severe diarrhea, and hepatitis may interfere with the anti-cancer approach. Sargramostim (glycosylated, yeast-derived, recombinant human GM-CSF) is an agent that has been used in clinical practice for more than 30 years with a well-recognized safety profile and has been studied in many therapeutic areas. The mechanism of action of sargramostim may treat moderate-to-severe GI irAEs without impairing the anti-cancer therapy. Some early data also suggest a potential survival benefit. Through the differentiation/maturation of monocytes, macrophages, and neutrophils and induction of anti-inflammatory T cell responses, GM-CSF aids in GI homeostasis, mucosal healing, and mucosal immunity. GM-CSF knockout mice are susceptible to severe colitis which was prevented with murine GM-CSF administration. For some patients with GI mucosa and immune cell function impairment, e.g., Crohn's disease, sargramostim reduces disease severity. In a prospective, randomized study (ECOG 1608), advanced melanoma patients had a reduction in grade 3-5 GI irAEs and less frequent colonic perforation in the sargramostim plus ipilimumab arm compared to ipilimumab alone. Sargramostim continues to be studied with ICIs for the prophylactic management of irAEs while also potentially providing a survival benefit.
Collapse
Affiliation(s)
- Michael Dougan
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; (M.D.); (E.I.B.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Long H. Nguyen
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA;
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Elizabeth I. Buchbinder
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; (M.D.); (E.I.B.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Hillard M. Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
12
|
Mabo A, Borie R, Wemeau-Stervinou L, Uzunhan Y, Gomez E, Prevot G, Reynaud-Gaubert M, Traclet J, Bergot E, Cadranel J, Marchand-Adam S, Bergeron A, Blanchard E, Bondue B, Bonniaud P, Bourdin A, Burgel PR, Hirschi S, Marquette CH, Quétant S, Nunes H, Chenivesse C, Crestani B, Guirriec Y, Monnier D, Ménard C, Tattevin P, Cottin V, Luque Paz D, Jouneau S. Infections in autoimmune pulmonary alveolar proteinosis: a large retrospective cohort. Thorax 2023; 79:68-74. [PMID: 37758458 DOI: 10.1136/thorax-2023-220040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Autoimmune pulmonary alveolar proteinosis (aPAP) is a rare disease, predisposing to an increased risk of infection. A complete picture of these infections is lacking. RESEARCH QUESTION Describe the characteristics and clinical outcomes of patients diagnosed with aPAP, and to identify risk factors associated with opportunistic infections. METHODS We conducted a retrospective cohort including all patients diagnosed with aPAP between 2008 and 2018 in France and Belgium. Data were collected using a standardised questionnaire including demographics, comorbidities, imaging features, outcomes and microbiological data. RESULTS We included 104 patients, 2/3 were men and median age at diagnosis was 45 years. With a median follow-up of 3.4 years (IQR 1.7-6.6 years), 60 patients (58%), developed at least one infection, including 23 (22%) with opportunistic infections. Nocardia spp was the main pathogen identified (n=10). Thirty-five (34%) patients were hospitalised due to infection. In univariate analysis, male gender was associated with opportunistic infections (p=0.04, OR=3.88; 95% CI (1.02 to 22.06)). Anti-granulocyte macrophage colony-stimulating factor antibody titre at diagnosis was significantly higher among patients who developed nocardiosis (1058 (316-1591) vs 580 (200-1190), p=0.01). Nine patients had died (9%), but only one death was related to infection. INTERPRETATION Patients with aPAP often presented with opportunistic infections, especially nocardiosis, which highlights the importance of systematic search for slow-growing bacteria in bronchoalveolar lavage or whole lung lavage.
Collapse
Affiliation(s)
- Axelle Mabo
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Raphael Borie
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie A, Hopital Bichat, APHP, Paris, France
| | - Lidwine Wemeau-Stervinou
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Institut Cœur-Poumon, Service de Pneumologie et Immuno-Allergologie, CHRU Lille, Lille, France
| | - Yurdagül Uzunhan
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie, Hôpital Avicenne, APHP, Bobigny, France
| | - Emmanuel Gomez
- Centre de Compétence pour les Maladies Pulmonaires Rares, Département de Pneumologie, Hôpitaux de Brabois, CHRU de Nancy, Vandoeuvre-les Nancy, France
| | - Gregoire Prevot
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Larry, CHU Toulouse, Toulouse, France
| | - Martine Reynaud-Gaubert
- Service de Pneumologie et Transplantation Pulmonaire, Centre de Compétences des Maladies Rares Pulmonaires et de l'Hypertension Pulmonaire, CHU Nord de Marseille, AP-HM, Aix Marseille Université, Marseille, France
| | - Julie Traclet
- Service de Pneumologie, Centre National Coordonnateur de Référence des Maladies Pulmonaires Rares, Hôpital Louis-Pradel, Hospices Civils de Lyon (HCL), UMR754, INRAE, Université Lyon 1, ERN-LUNG, Lyon, France
| | - Emmanuel Bergot
- Centre de Compétence pour les Maladies Pulmonaires Rares de l'Adulte, Service de Pneumologie et Oncologie Thoracique, Hôpital Côte de Nacre, CHU de Caen, Caen, France
| | - Jacques Cadranel
- Service de Pneumologie et Oncologie Thoracique, Centre Constitutif Maladies Pulmonaires Rares, Hôpital Tenon, APHP, Sorbonne Université, Paris, France
| | - Sylvain Marchand-Adam
- Service de Pneumologie, CHRU de Tours, Université François Rabelais de Tours, INSERM U1100, Tours, France
| | - Anne Bergeron
- Service de Pneumologie, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Elodie Blanchard
- Service de Pneumologie, centre de compétence pour les maladies pulmonaires rares, CHU de Bordeaux, Pessac, France
| | - Benjamin Bondue
- Service de Pneumologie, CUB Hôpital Erasme, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Philippe Bonniaud
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence Constitutif des Maladies Pulmonaires Rares de l'Adulte, CHU Dijon-Bourgogne, Inserm U123, Université de Bourgogne, Dijon, France
| | - Arnaud Bourdin
- Service de Pneumologie, CHU Montpellier, Université de Montpellier, Inserm U1046, Montpellier, France
| | - Pierre Regis Burgel
- Service de Pneumologie, Hopital Cochin Pneumologie, AP-HP, Université Paris Cité Paris, Paris, France
| | - Sandrine Hirschi
- Service de Pneumologie et Transplantation Pulmonaire, Hopitaux universitaires de Strasbourg, Strasbourg, France
| | - Charles Hugo Marquette
- Service de Pneumologie, CHU Nice, Fédération Hospitalo-Universitaire OncoAge, Nice, France
| | - Sébastien Quétant
- Service Hospitalo-Universitaire de Pneumologie et Physiologie, Pôle Thorax et Vaisseaux, CHU de Grenoble-Alpes, La Tronche, Grenoble, France
| | - Hilario Nunes
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie, Hôpital Avicenne, APHP, Bobigny, France
| | - Cécile Chenivesse
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Institut Cœur-Poumon, Service de Pneumologie et Immuno-Allergologie, CHRU Lille, Lille, France
| | - Bruno Crestani
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie A, Hopital Bichat, APHP, Paris, France
| | - Yoann Guirriec
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Delphine Monnier
- Service d'Immunologie, Laboratoire de Biologie Médicale de Référence Lipoprotéinose Alvéolaire, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Cédric Ménard
- Service d'Immunologie, Laboratoire de Biologie Médicale de Référence Lipoprotéinose Alvéolaire, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Pierre Tattevin
- Service de Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, Inserm U1230, Université de Rennes, Rennes, France
| | - Vincent Cottin
- Service de Pneumologie, Centre National Coordonnateur de Référence des Maladies Pulmonaires Rares, Hôpital Louis-Pradel, Hospices Civils de Lyon (HCL), UMR754, INRAE, Université Lyon 1, ERN-LUNG, Lyon, France
| | - David Luque Paz
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- Service de Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, Inserm U1230, Université de Rennes, Rennes, France
| | - Stéphane Jouneau
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- Inserm UMR1085 IRSET, Université de Rennes, EHESP, Rennes, France
| |
Collapse
|
13
|
Abstract
Pulmonary surfactant is a critical component of lung function in healthy individuals. It functions in part by lowering surface tension in the alveoli, thereby allowing for breathing with minimal effort. The prevailing thinking is that low surface tension is attained by a compression-driven squeeze-out of unsaturated phospholipids during exhalation, forming a film enriched in saturated phospholipids that achieves surface tensions close to zero. A thorough review of past and recent literature suggests that the compression-driven squeeze-out mechanism may be erroneous. Here, we posit that a surfactant film enriched in saturated lipids is formed shortly after birth by an adsorption-driven sorting process and that its composition does not change during normal breathing. We provide biophysical evidence for the rapid formation of an enriched film at high surfactant concentrations, facilitated by adsorption structures containing hydrophobic surfactant proteins. We examine biophysical evidence for and against the compression-driven squeeze-out mechanism and propose a new model for surfactant function. The proposed model is tested against existing physiological and pathophysiological evidence in neonatal and adult lungs, leading to ideas for biophysical research, that should be addressed to establish the physiological relevance of this new perspective on the function of the mighty thin film that surfactant provides.
Collapse
Affiliation(s)
- Fred Possmayer
- Department of Biochemistry, Western University, London, Ontario N6A 3K7, Canada
- Department of Obstetrics/Gynaecology, Western University, London, Ontario N6A 3K7, Canada
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manon, Honolulu, Hawaii 96822, United States
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96826, United States
| | - Ruud A W Veldhuizen
- Department of Physiology & Pharmacology, Western University, London, Ontario N6A 5C1, Canada
- Department of Medicine, Western University, London, Ontario N6A 3K7, Canada
- Lawson Health Research Institute, London, Ontario N6A 4V2, Canada
| | - Nils O Petersen
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada
| |
Collapse
|
14
|
Chang CY, Armstrong D, Corry DB, Kheradmand F. Alveolar macrophages in lung cancer: opportunities challenges. Front Immunol 2023; 14:1268939. [PMID: 37822933 PMCID: PMC10562548 DOI: 10.3389/fimmu.2023.1268939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
Alveolar macrophages (AMs) are critical components of the innate defense mechanism in the lung. Nestled tightly within the alveoli, AMs, derived from the yolk-sac or bone marrow, can phagocytose foreign particles, defend the host against pathogens, recycle surfactant, and promptly respond to inhaled noxious stimuli. The behavior of AMs is tightly dependent on the environmental cues whereby infection, chronic inflammation, and associated metabolic changes can repolarize their effector functions in the lungs. Several factors within the tumor microenvironment can re-educate AMs, resulting in tumor growth, and reducing immune checkpoint inhibitors (ICIs) efficacy in patients treated for non-small cell lung cancer (NSCLC). The plasticity of AMs and their critical function in altering tumor responses to ICIs make them a desirable target in lung cancer treatment. New strategies have been developed to target AMs in solid tumors reprograming their suppressive function and boosting the efficacy of ICIs. Here, we review the phenotypic and functional changes in AMs in response to sterile inflammation and in NSCLC that could be critical in tumor growth and metastasis. Opportunities in altering AMs' function include harnessing their potential function in trained immunity, a concept borrowed from memory response to infections, which could be explored therapeutically in managing lung cancer treatment.
Collapse
Affiliation(s)
- Cheng-Yen Chang
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Dominique Armstrong
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - David B. Corry
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, United States
| | - Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, United States
| |
Collapse
|
15
|
Pahari S, Arnett E, Simper J, Azad A, Guerrero-Arguero I, Ye C, Zhang H, Cai H, Wang Y, Lai Z, Jarvis N, Lumbreras M, Maselli DJ, Peters J, Torrelles JB, Martinez-Sobrido L, Schlesinger LS. A new tractable method for generating human alveolar macrophage-like cells in vitro to study lung inflammatory processes and diseases. mBio 2023; 14:e0083423. [PMID: 37288969 PMCID: PMC10470505 DOI: 10.1128/mbio.00834-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 06/09/2023] Open
Abstract
Alveolar macrophages (AMs) are unique lung resident cells that contact airborne pathogens and environmental particulates. The contribution of human AMs (HAMs) to pulmonary diseases remains poorly understood due to the difficulty in accessing them from human donors and their rapid phenotypic change during in vitro culture. Thus, there remains an unmet need for cost-effective methods for generating and/or differentiating primary cells into a HAM phenotype, particularly important for translational and clinical studies. We developed cell culture conditions that mimic the lung alveolar environment in humans using lung lipids, that is, Infasurf (calfactant, natural bovine surfactant) and lung-associated cytokines (granulocyte macrophage colony-stimulating factor, transforming growth factor-β, and interleukin 10) that facilitate the conversion of blood-obtained monocytes to an AM-like (AML) phenotype and function in tissue culture. Similar to HAM, AML cells are particularly susceptible to both Mycobacterium tuberculosis and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. This study reveals the importance of alveolar space components in the development and maintenance of HAM phenotype and function and provides a readily accessible model to study HAM in infectious and inflammatory disease processes, as well as therapies and vaccines. IMPORTANCE Millions die annually from respiratory disorders. Lower respiratory track gas-exchanging alveoli maintain a precarious balance between fighting invaders and minimizing tissue damage. Key players herein are resident AMs. However, there are no easily accessible in vitro models of HAMs, presenting a huge scientific challenge. Here, we present a novel model for generating AML cells based on differentiating blood monocytes in a defined lung component cocktail. This model is non-invasive, significantly less costly than performing a bronchoalveolar lavage, yields more AML cells than HAMs per donor, and retains their phenotype in culture. We have applied this model to early studies of M. tuberculosis and SARS-CoV-2. This model will significantly advance respiratory biology research.
Collapse
Affiliation(s)
- Susanta Pahari
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Eusondia Arnett
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jan Simper
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Department of Molecular Immunology and Microbiology, UT Health San Antonio, San Antonio, Texas, USA
| | - Abul Azad
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Israel Guerrero-Arguero
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Chengjin Ye
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Hong Cai
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Yufeng Wang
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Zhao Lai
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | - Natalie Jarvis
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Department of Molecular Immunology and Microbiology, UT Health San Antonio, San Antonio, Texas, USA
| | - Miranda Lumbreras
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Diego Jose Maselli
- Division of Pulmonary and Critical Care Medicine, UT Health Science Center, San Antonio, Texas, USA
| | - Jay Peters
- Division of Pulmonary and Critical Care Medicine, UT Health Science Center, San Antonio, Texas, USA
| | - Jordi B. Torrelles
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Luis Martinez-Sobrido
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Larry S. Schlesinger
- Host Pathogen Interactions and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
16
|
Kappagoda S, Deresinski S. Anticytokine Autoantibodies and Fungal Infections. J Fungi (Basel) 2023; 9:782. [PMID: 37623553 PMCID: PMC10455114 DOI: 10.3390/jof9080782] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Anticytokine autoantibodies (ACAAs) can cause adult onset immunodeficiencies which mimic primary immunodeficiencies and can present as refractory and severe fungal infections. This paper provides an overview of the role of innate immunity, including key cytokines, in fungal infections and then describes four clinical scenarios where ACAAs are associated with severe presentations of a fungal infection: (1) Talaromyces marneffei infection and anti-interferon-γ, (2) histoplasmosis and anti-interferon-γ, (3) Cryptococcus gattii infection and anti-GM-CSF, and (4) mucocutaneous candidiasis and anti-IL-17A/F (IL-22). Testing for ACAAs and potential therapeutic options are discussed.
Collapse
Affiliation(s)
- Shanthi Kappagoda
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA 94304, USA;
| | | |
Collapse
|
17
|
Garavaglia ML, Bodega F, Porta C, Milzani A, Sironi C, Dalle-Donne I. Molecular Impact of Conventional and Electronic Cigarettes on Pulmonary Surfactant. Int J Mol Sci 2023; 24:11702. [PMID: 37511463 PMCID: PMC10380520 DOI: 10.3390/ijms241411702] [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: 06/20/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The alveolar epithelium is covered by a non-cellular layer consisting of an aqueous hypophase topped by pulmonary surfactant, a lipo-protein mixture with surface-active properties. Exposure to cigarette smoke (CS) affects lung physiology and is linked to the development of several diseases. The macroscopic effects of CS are determined by several types of cell and molecular dysfunction, which, among other consequences, lead to surfactant alterations. The purpose of this review is to summarize the published studies aimed at uncovering the effects of CS on both the lipid and protein constituents of surfactant, discussing the molecular mechanisms involved in surfactant homeostasis that are altered by CS. Although surfactant homeostasis has been the topic of several studies and some molecular pathways can be deduced from an analysis of the literature, it remains evident that many aspects of the mechanisms of action of CS on surfactant homeostasis deserve further investigation.
Collapse
Affiliation(s)
| | - Francesca Bodega
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Cristina Porta
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Aldo Milzani
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milan, Italy
| | - Chiara Sironi
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Isabella Dalle-Donne
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
18
|
El-Atoum M, Hargarten JC, Park YD, Ssebambulidde K, Ding L, Chittiboina P, Hammoud DA, Anjum SH, Glassman SR, Merchant S, Williamson PR, Hu JC. Persistent neurological symptoms and elevated intracranial pressures in a previously healthy host with cryptococcal meningitis. BMC Infect Dis 2023; 23:407. [PMID: 37316806 DOI: 10.1186/s12879-023-08349-y] [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: 01/06/2023] [Accepted: 05/25/2023] [Indexed: 06/16/2023] Open
Abstract
Cryptococcal meningoencephalitis can occur in both previously healthy and immunocompromised hosts. Here, we describe a 55 year-old HIV-negative male with no known prior medical problems, who presented with three months of worsening headaches, confusion, and memory changes without fever. Magnetic resonance imaging of the brain demonstrated bilateral enlargement/enhancement of the choroid plexi, with hydrocephalus, temporal and occipital horn entrapments, as well as marked periventricular transependymal cerebrospinal fluid (CSF) seepage. CSF analysis yielded a lymphocytic pleocytosis and cryptococcal antigen titer of 1:160 but sterile fungal cultures. Despite standard antifungal therapy and CSF drainage, the patient had worsening confusion and persistently elevated intracranial pressures. External ventricular drainage led to improved mental status but only with valve settings at negative values. Ventriculoperitoneal shunt placement could thus not be considered due to a requirement for drainage into the positive pressure venous system. Due to this persistent CSF inflammation and cerebral circulation obstruction, the patient required transfer to the National Institute of Health. He was treated for cryptococcal post-infectious inflammatory response syndrome with pulse-taper corticosteroid therapy, with resultant reductions in CSF pressures along with decreased protein and obstructive material, allowing successful shunt placement. After tapering of corticosteroids, the patient recovered without sequelae. This case highlights (1) the necessity to consider cryptococcal meningitis as a rare cause of neurological deterioration in the absence of fever even in apparently immunocompetent individuals and (2) the potential for obstructive phenomena from inflammatory sequelae and the prompt response to corticosteroid therapy.
Collapse
Affiliation(s)
- Mohammad El-Atoum
- Department of Medicine, Good Samaritan Hospital, SSM Health Medical Group, Mount Vernon, IL, USA.
| | - Jessica C Hargarten
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Yoon-Dong Park
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Kenneth Ssebambulidde
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Li Ding
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Seher H Anjum
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Seth R Glassman
- Department of Medicine, Division of Infectious Diseases, University at Buffalo, 955 Main Street, Buffalo, NY, 14203, USA
| | - Shehzad Merchant
- Department of Medicine, Division of Infectious Diseases, University at Buffalo, 955 Main Street, Buffalo, NY, 14203, USA
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - John C Hu
- Department of Medicine, Division of Infectious Diseases, University at Buffalo, 955 Main Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
19
|
Chen S, Saeed AFUH, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L, Duo Y. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther 2023; 8:207. [PMID: 37211559 DOI: 10.1038/s41392-023-01452-1] [Citation(s) in RCA: 353] [Impact Index Per Article: 353.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023] Open
Abstract
Macrophages exist in various tissues, several body cavities, and around mucosal surfaces and are a vital part of the innate immune system for host defense against many pathogens and cancers. Macrophages possess binary M1/M2 macrophage polarization settings, which perform a central role in an array of immune tasks via intrinsic signal cascades and, therefore, must be precisely regulated. Many crucial questions about macrophage signaling and immune modulation are yet to be uncovered. In addition, the clinical importance of tumor-associated macrophages is becoming more widely recognized as significant progress has been made in understanding their biology. Moreover, they are an integral part of the tumor microenvironment, playing a part in the regulation of a wide variety of processes including angiogenesis, extracellular matrix transformation, cancer cell proliferation, metastasis, immunosuppression, and resistance to chemotherapeutic and checkpoint blockade immunotherapies. Herein, we discuss immune regulation in macrophage polarization and signaling, mechanical stresses and modulation, metabolic signaling pathways, mitochondrial and transcriptional, and epigenetic regulation. Furthermore, we have broadly extended the understanding of macrophages in extracellular traps and the essential roles of autophagy and aging in regulating macrophage functions. Moreover, we discussed recent advances in macrophages-mediated immune regulation of autoimmune diseases and tumorigenesis. Lastly, we discussed targeted macrophage therapy to portray prospective targets for therapeutic strategies in health and diseases.
Collapse
Affiliation(s)
- Shanze Chen
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Abdullah F U H Saeed
- Department of Cancer Biology, Beckman Research Institute of City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen University, Shenzhen, 518052, China
| | - Qiong Jiang
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Haizhao Xu
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Department of Respiratory, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Gary Guishan Xiao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian, China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Yanhong Duo
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
20
|
Quiros-Roldan E, Sottini A, Signorini SG, Serana F, Tiecco G, Imberti L. Autoantibodies to Interferons in Infectious Diseases. Viruses 2023; 15:v15051215. [PMID: 37243300 DOI: 10.3390/v15051215] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Anti-cytokine autoantibodies and, in particular, anti-type I interferons are increasingly described in association with immunodeficient, autoimmune, and immune-dysregulated conditions. Their presence in otherwise healthy individuals may result in a phenotype characterized by a predisposition to infections with several agents. For instance, anti-type I interferon autoantibodies are implicated in Coronavirus Disease 19 (COVID-19) pathogenesis and found preferentially in patients with critical disease. However, autoantibodies were also described in the serum of patients with viral, bacterial, and fungal infections not associated with COVID-19. In this review, we provide an overview of anti-cytokine autoantibodies identified to date and their clinical associations; we also discuss whether they can act as enemies or friends, i.e., are capable of acting in a beneficial or harmful way, and if they may be linked to gender or immunosenescence. Understanding the mechanisms underlying the production of autoantibodies could improve the approach to treating some infections, focusing not only on pathogens, but also on the possibility of a low degree of autoimmunity in patients.
Collapse
Affiliation(s)
- Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, ASST Spedali Civili, Brescia and University of Brescia, 25123 Brescia, Italy
| | - Alessandra Sottini
- Clinical Chemistry Laboratory, ASST Spedali Civili of Brescia, 25123 Brescia, Italy
| | | | - Federico Serana
- Clinical Chemistry Laboratory, ASST Spedali Civili of Brescia, 25123 Brescia, Italy
| | - Giorgio Tiecco
- Department of Infectious and Tropical Diseases, ASST Spedali Civili, Brescia and University of Brescia, 25123 Brescia, Italy
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, P. le Spedali Civili, 1, 25123 Brescia, Italy
| |
Collapse
|
21
|
Pahari S, Arnett E, Simper J, Azad A, Guerrero-Arguero I, Ye C, Zhang H, Cai H, Wang Y, Lai Z, Jarvis N, Lumbreras M, Maselli-Caceres DJ, Peters J, Torrelles JB, Martinez-Sobrido L, Schlesinger LS. A new tractable method for generating Human Alveolar Macrophage Like cells in vitro to study lung inflammatory processes and diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.05.535806. [PMID: 37066199 PMCID: PMC10104118 DOI: 10.1101/2023.04.05.535806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Alveolar macrophages (AMs) are unique lung resident cells that contact airborne pathogens and environmental particulates. The contribution of human AMs (HAM) to pulmonary diseases remains poorly understood due to difficulty in accessing them from human donors and their rapid phenotypic change during in vitro culture. Thus, there remains an unmet need for cost-effective methods for generating and/or differentiating primary cells into a HAM phenotype, particularly important for translational and clinical studies. We developed cell culture conditions that mimic the lung alveolar environment in humans using lung lipids, i.e. , Infasurf (calfactant, natural bovine surfactant) and lung-associated cytokines (GM-CSF, TGF-β, and IL-10) that facilitate the conversion of blood-obtained monocytes to an AM-Like (AML) phenotype and function in tissue culture. Similar to HAM, AML cells are particularly susceptible to both Mycobacterium tuberculosis and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. This study reveals the importance of alveolar space components in the development and maintenance of HAM phenotype and function, and provides a readily accessible model to study HAM in infectious and inflammatory disease processes, as well as therapies and vaccines. IMPORTANCE Millions die annually from respiratory disorders. Lower respiratory track gas-exchanging alveoli maintain a precarious balance between fighting invaders and minimizing tissue damage. Key players herein are resident AMs. However, there are no easily accessible in vitro models of HAMs, presenting a huge scientific challenge. Here we present a novel model for generating AML cells based on differentiating blood monocytes in a defined lung component cocktail. This model is non-invasive, significantly less costly than performing a bronchoalveolar lavage, yields more AML cells than HAMs per donor and retains their phenotype in culture. We have applied this model to early studies of M. tuberculosis and SARS-CoV-2. This model will significantly advance respiratory biology research.
Collapse
Affiliation(s)
- Susanta Pahari
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Eusondia Arnett
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jan Simper
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Molecular Immunology and Microbiology, UT Health San Antonio, San Antonio, TX, USA
| | - Abul Azad
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Israel Guerrero-Arguero
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chengjin Ye
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious diseases, University of Texas at San Antonio, San Antonio, TX, USA
| | - Hong Cai
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious diseases, University of Texas at San Antonio, San Antonio, TX, USA
| | - Yufeng Wang
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious diseases, University of Texas at San Antonio, San Antonio, TX, USA
| | - Zhao Lai
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Natalie Jarvis
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Molecular Immunology and Microbiology, UT Health San Antonio, San Antonio, TX, USA
| | - Miranda Lumbreras
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jay Peters
- Division of Pulmonary and Critical Care Medicine, UT Health Science Center, San Antonio, TX 78207
| | - Jordi B Torrelles
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Luis Martinez-Sobrido
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Larry S Schlesinger
- Host Pathogen Interactions and Population Health programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
22
|
Mondini L, Salton F, Trotta L, Bozzi C, Pozzan R, Barbieri M, Tavano S, Lerda S, Hughes M, Confalonieri M, Confalonieri P, Ruaro B. Host-Based Treatments for Severe COVID-19. Curr Issues Mol Biol 2023; 45:3102-3121. [PMID: 37185727 PMCID: PMC10136924 DOI: 10.3390/cimb45040203] [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: 03/03/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
COVID-19 has been a global health problem since 2020. There are different spectrums of manifestation of this disease, ranging from asymptomatic to extremely severe forms requiring admission to intensive care units and life-support therapies, mainly due to severe pneumonia. The progressive understanding of this disease has allowed researchers and clinicians to implement different therapeutic alternatives, depending on both the severity of clinical involvement and the causative molecular mechanism that has been progressively explored. In this review, we analysed the main therapeutic options available to date based on modulating the host inflammatory response to SARS-CoV-2 infection in patients with severe and critical illness. Although current guidelines are moving toward a personalised treatment approach titrated on the timing of presentation, disease severity, and laboratory parameters, future research is needed to identify additional biomarkers that can anticipate the disease course and guide targeted interventions on an individual basis.
Collapse
Affiliation(s)
- Lucrezia Mondini
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Francesco Salton
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Liliana Trotta
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Chiara Bozzi
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Riccardo Pozzan
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Mariangela Barbieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Stefano Tavano
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Selene Lerda
- Graduate School, University of Milan, 20149 Milano, Italy
| | - Michael Hughes
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester M6 8HD, UK
| | - Marco Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Paola Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Barbara Ruaro
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
23
|
Chen Y, Li F, Hua M, Liang M, Song C. Role of GM-CSF in lung balance and disease. Front Immunol 2023; 14:1158859. [PMID: 37081870 PMCID: PMC10111008 DOI: 10.3389/fimmu.2023.1158859] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor originally identified as a stimulus that induces the differentiation of bone marrow progenitor cells into granulocytes and macrophages. GM-CSF is now considered to be a multi-origin and pleiotropic cytokine. GM-CSF receptor signals activate JAK2 and induce nuclear signals through the JAK-STAT, MAPK, PI3K, and other pathways. In addition to promoting the metabolism of pulmonary surfactant and the maturation and differentiation of alveolar macrophages, GM-CSF plays a key role in interstitial lung disease, allergic lung disease, alcoholic lung disease, and pulmonary bacterial, fungal, and viral infections. This article reviews the latest knowledge on the relationship between GM-CSF and lung balance and lung disease, and indicates that there is much more to GM-CSF than its name suggests.
Collapse
Affiliation(s)
- Yingzi Chen
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
| | - Fan Li
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
| | - Mengqing Hua
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
| | - Meng Liang
- Department of Biotechnology, School of Life Science, Bengbu Medical College, Anhui, China
- *Correspondence: Chuanwang Song, ; Meng Liang,
| | - Chuanwang Song
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
- *Correspondence: Chuanwang Song, ; Meng Liang,
| |
Collapse
|
24
|
Of Mycelium and Men: Inherent Human Susceptibility to Fungal Diseases. Pathogens 2023; 12:pathogens12030456. [PMID: 36986378 PMCID: PMC10058615 DOI: 10.3390/pathogens12030456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
In medical mycology, the main context of disease is iatrogenic-based disease. However, historically, and occasionally, even today, fungal diseases affect humans with no obvious risk factors, sometimes in a spectacular fashion. The field of “inborn errors of immunity” (IEI) has deduced at least some of these previously enigmatic cases; accordingly, the discovery of single-gene disorders with penetrant clinical effects and their immunologic dissection have provided a framework with which to understand some of the key pathways mediating human susceptibility to mycoses. By extension, they have also enabled the identification of naturally occurring auto-antibodies to cytokines that phenocopy such susceptibility. This review provides a comprehensive update of IEI and autoantibodies that inherently predispose humans to various fungal diseases.
Collapse
|
25
|
Bai JW, Huang JN, Shi SY, Ge A, Lu HW, Sun XL, Gu SY, Liang S, Cheng KB, Tian XL, Xiao YL, Xu KF, Xu JF. Updated severity and prognosis score of pulmonary alveolar proteinosis: A multi-center cohort study in China. Front Med (Lausanne) 2023; 10:1058001. [PMID: 36824611 PMCID: PMC9941621 DOI: 10.3389/fmed.2023.1058001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/19/2023] [Indexed: 02/10/2023] Open
Abstract
Background The high-resolution computed tomography (HRCT) score is an important component of the severity and prognosis score of pulmonary alveolar proteinosis (SPSP). However, the HRCT score in SPSP only considers the extent of opacity, which is insufficient. Methods We retrospectively evaluated HRCT scores for 231 patients with autoimmune pulmonary alveolar proteinosis (APAP) from three centers of the China Alliance for Rare Diseases. The SPSPII was created based on the overall density and extent, incorporating the SPSP. The severity of APAP patients was assessed using disease severity scores (DSS), SPSP, and SPSPII to determine the strengths and weaknesses of the different assessment methods. We then prospectively applied the SPSPII to patients before treatment, and the curative effect was assessed after 3 months. Results The HRCT overall density and extent scores in our retrospective analysis were higher than the extent scores in all patients and every original extent score severity group, as well as higher related to arterial partial oxygen pressure (PaO2) than extent scores. The mild patients accounted for 61.9% based on DSS 1-2, 20.3% based on SPSP 1-3, and 20.8% based on SPSPII 1-3. Based on SPSP or SPSPII, the number of severe patients deteriorating was higher in the mild and moderate groups. When applied prospectively, arterial PaO2 differed between any two SPSPII severity groups. The alveolar-arterial gradient in PaO2 (P[A-a]O2), % predicted carbon monoxide diffusing capacity of the lung (DLCO), and HRCT score were higher in the severe group than in the mild and moderate groups. After diagnosis, mild patients received symptomatic treatment, moderate patients received pure whole lung lavage (WLL) or granulocyte-macrophage colony-stimulating factor (GM-CSF) therapy, and severe patients received WLL and GM-CSF therapy. Importantly, the SPSPII in mild and severe groups were lower than baseline after 3 months. Conclusion The HRCT density and extent scores of patients with APAP were better than the extent score. The SPSPII score system based on smoking status, symptoms, PaO2, predicted DLCO, and overall HRCT score was better than DSS and SPSP for assessing the severity and efficacy and predicting the prognosis. Trial registration ClinicalTrial.gov, identifier: NCT04516577.
Collapse
Affiliation(s)
- Jiu-Wu Bai
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian-nan Huang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shen-yun Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, China
| | - Ai Ge
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hai-wen Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiao-li Sun
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shu-yi Gu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuo Liang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ke-bin Cheng
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin-lun Tian
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yong-long Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, China,*Correspondence: Yong-long Xiao,
| | - Kai-feng Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China,Kai-feng Xu,
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China,Jin-Fu Xu,
| |
Collapse
|
26
|
Kadota N, Nakahira N, Miyauchi M, Naruse K, Takeuchi E, Shinohara T. Usefulness of bronchoalveolar lavage (BAL) in the diagnosis of pulmonary alveolar proteinosis. QJM 2022; 115:767-768. [PMID: 35809073 DOI: 10.1093/qjmed/hcac168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/04/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- N Kadota
- Department of Pulmonary Medicine, National Hospital Organization Kochi Hospital, 1-2-25 Asakuranishimachi, Kochi 780-8077, Japan
| | - N Nakahira
- Department of Clinical Laboratory, National Hospital Organization Kochi Hospital, 1-2-25 Asakuranishimachi, Kochi 780-8077, Japan
| | - M Miyauchi
- Department of Clinical Laboratory, National Hospital Organization Kochi Hospital, 1-2-25 Asakuranishimachi, Kochi 780-8077, Japan
| | - K Naruse
- Department of Pathology, National Hospital Organization Kochi Hospital, 1-2-25 Asakuranishimachi, Kochi 780-8077, Japan
| | - E Takeuchi
- Department of Clinical Investigation, National Hospital Organization Kochi Hospital, 1-2-25 Asakuranishimachi, Kochi 780-8077, Japan
| | - T Shinohara
- Department of Clinical Investigation, National Hospital Organization Kochi Hospital, 1-2-25 Asakuranishimachi, Kochi 780-8077, Japan
- Department of Community Medicine for Respirology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
27
|
Obi ON, Saketkoo LA, Russell AM, Baughman RP. Sarcoidosis: Updates on therapeutic drug trials and novel treatment approaches. Front Med (Lausanne) 2022; 9:991783. [PMID: 36314034 PMCID: PMC9596775 DOI: 10.3389/fmed.2022.991783] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 12/04/2022] Open
Abstract
Sarcoidosis is a systemic granulomatous inflammatory disease of unknown etiology. It affects the lungs in over 90% of patients yet extra-pulmonary and multi-organ involvement is common. Spontaneous remission of disease occurs commonly, nonetheless, over 50% of patients will require treatment and up to 30% of patients will develop a chronic progressive non-remitting disease with marked pulmonary fibrosis leading to significant morbidity and death. Guidelines outlining an immunosuppressive treatment approach to sarcoidosis were recently published, however, the strength of evidence behind many of the guideline recommended drugs is weak. None of the drugs currently used for the treatment of sarcoidosis have been rigorously studied and prescription of these drugs is often based on off-label” indications informed by experience with other diseases. Indeed, only two medications [prednisone and repository corticotropin (RCI) injection] currently used in the treatment of sarcoidosis are approved by the United States Food and Drug Administration. This situation results in significant reimbursement challenges especially for the more advanced (and often more effective) drugs that are favored for severe and refractory forms of disease causing an over-reliance on corticosteroids known to be associated with significant dose and duration dependent toxicities. This past decade has seen a renewed interest in developing new drugs and exploring novel therapeutic pathways for the treatment of sarcoidosis. Several of these trials are active randomized controlled trials (RCTs) designed to recruit relatively large numbers of patients with a goal to determine the safety, efficacy, and tolerability of these new molecules and therapeutic approaches. While it is an exciting time, it is also necessary to exercise caution. Resources including research dollars and most importantly, patient populations available for trials are limited and thus necessitate that several of the challenges facing drug trials and drug development in sarcoidosis are addressed. This will ensure that currently available resources are judiciously utilized. Our paper reviews the ongoing and anticipated drug trials in sarcoidosis and addresses the challenges facing these and future trials. We also review several recently completed trials and draw lessons that should be applied in future.
Collapse
Affiliation(s)
- Ogugua Ndili Obi
- Division of Pulmonary Critical Care and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, United States,*Correspondence: Ogugua Ndili Obi,
| | - Lesley Ann Saketkoo
- New Orleans Scleroderma and Sarcoidosis Patient Care and Research Center, New Orleans, LA, United States,University Medical Center—Comprehensive Pulmonary Hypertension Center and Interstitial Lung Disease Clinic Programs, New Orleans, LA, United States,Section of Pulmonary Medicine, Louisiana State University School of Medicine, New Orleans, LA, United States,Department of Undergraduate Honors, Tulane University School of Medicine, New Orleans, LA, United States
| | - Anne-Marie Russell
- Exeter Respiratory Institute University of Exeter, Exeter, United Kingdom,Royal Devon and Exeter NHS Foundation Trust, Devon, United Kingdom,Faculty of Medicine, Imperial College and Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Robert P. Baughman
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
28
|
Nagaoka Y, Kosaku K, Yoshikawa H, Abe M, Miyawaki M, Daa T, Hiramatsu K, Sugio K, Kadota JI. Aspergillus Nodule in a Patient With Autoimmune Pulmonary Alveolar Proteinosis. Cureus 2022; 14:e29095. [PMID: 36249614 PMCID: PMC9556838 DOI: 10.7759/cureus.29095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 11/29/2022] Open
Abstract
Although autoimmune pulmonary alveolar proteinosis (APAP) is more likely to be associated with infectious diseases, clinical case-based evidence is too limited to confirm this. We describe a case of a man in his late forties diagnosed with APAP nine years prior to the current presentation. A nodule in the right upper lobe gradually increased from 8 to 12 mm over a period of 6 months and was suspicious of malignancy. The pathological analyses revealed Aspergillus nodule without any malignant features. This study aims to report a case of Aspergillus nodule with APAP and discuss the differential diagnosis of solitary lung nodule developed in APAP.
Collapse
|
29
|
Lee E, Miller C, Ataya A, Wang T. Opportunistic Infection Associated with Elevated GM-CSF Autoantibodies: A Case Series and Review of the Literature. Open Forum Infect Dis 2022; 9:ofac146. [PMID: 35531378 PMCID: PMC9070348 DOI: 10.1093/ofid/ofac146] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is known to play a key role in enhancing multiple immune functions that affect response to infectious pathogens including antigen presentation, complement- and antibody-mediated phagocytosis, microbicidal activity, and neutrophil chemotaxis. Reduced GM-CSF activity and immune response provides a mechanism for increased infection risk associated with autoimmune pulmonary alveolar proteinosis (aPAP) and other disorders involving the presence of GM-CSF autoantibodies. We present a case series of five patients with persistent or unusual pulmonary and central nervous system opportunistic infections (Cryptococcus gattii, Flavobacterium, Nocardia) and elevated GM-CSF autoantibody levels, as well as 27 cases identified on systematic review of the literature.
Collapse
Affiliation(s)
- Elinor Lee
- UCLA Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Christopher Miller
- UCLA Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ali Ataya
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Tisha Wang
- UCLA Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
30
|
Recent Advances in Influenza, HIV and SARS-CoV-2 Infection Prevention and Drug Treatment—The Need for Precision Medicine. CHEMISTRY 2022. [DOI: 10.3390/chemistry4020019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Viruses, and in particular, RNA viruses, dominate the WHO’s current list of ten global health threats. Of these, we review the widespread and most common HIV, influenza virus, and SARS-CoV-2 infections, as well as their possible prevention by vaccination and treatments by pharmacotherapeutic approaches. Beyond the vaccination, we discuss the virus-targeting and host-targeting drugs approved in the last five years, in the case of SARS-CoV-2 in the last one year, as well as new drug candidates and lead molecules that have been published in the same periods. We share our views on vaccination and pharmacotherapy, their mutually reinforcing strategic significance in combating pandemics, and the pros and cons of host and virus-targeted drug therapy. The COVID-19 pandemic has provided evidence of our limited armamentarium to fight emerging viral diseases. Novel broad-spectrum vaccines as well as drugs that could even be applied as prophylactic treatments or in early phases of the viremia, possibly through oral administration, are needed in all three areas. To meet these needs, the use of multi-data-based precision medicine in the practice and innovation of vaccination and drug therapy is inevitable.
Collapse
|
31
|
Šterclová M. Pulmonary storage. VNITRNI LEKARSTVI 2022; 68:525-531. [PMID: 36575071 DOI: 10.36290/vnl.2022.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Interstitial lung diseases (ILDs) are not just a matter of scarring or inflammation in the lung tissue. The lungs can also serve as a repository for products that can be produced in excessive amounts in the human body as a result of disease. Geneticaly based dysfunctions of lysosomal enzymes, which leads to an unefficient degradation and transport of various macromolecules from lysosomes, are considered to be storage diseases sensu stricto. ILDs were described in patients with Gaucher disease, Niemann-Pick disease and Fabry disease. In a broader context, however, the accumulation of various substances in the lung tissue is also encountered in cases of pediatric pulmonary interstitial glycogenosis (PIG), alveolar lipoproteinosis or pulmonary amyloidosis. The cause of PIG is not clear. The disease was first described in 2002 and a lung tissue sample is required to establish this diagnosis. Even though PIG usually goes well in childhood and the patients difficulties spontaneously subside over time, the long-term prognosis of the patients is unknown. Alveolar lipoproteinoses can be acquired (e.g. after massive exposure to silica dust), autoimmune, but also genetically determined. Unlike lysosomal storage diseases, in the case of pulmonary alveolar lipoproteinosis, accumulation of abnormal macromolecules occurs only in the lungs of affected individuals. Similarly, amyloidosis is not a single disease, but a group of diseases with different etiopathogenesis, as a result of which amyloid - a group of different proteins with a distinctvive conformation, which can be deposited in various organs, including the lungs - is formed. The diagnosis of pulmonary alveolar lipoproteinosis is based on the typical appearance and biochemical composition of the fluid obtained by bronchoalveolar lavage, the diagnosis of amyloidosis is histological.
Collapse
|