Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 494
Filter
1.
Eur Respir J ; 63(1)2024 01.
Article in English | MEDLINE | ID: mdl-37973175

ABSTRACT

RATIONALE: Whole lung lavage (WLL) is a widely accepted palliative treatment for autoimmune pulmonary alveolar proteinosis (aPAP) but does not correct myeloid cell dysfunction or reverse the pathological accumulation of surfactant. In contrast, inhaled recombinant granulocyte-macrophage colony-stimulating factor (rGM-CSF) is a promising pharmacological approach that restores alveolar macrophage functions including surfactant clearance. Here, we evaluate WLL followed by inhaled rGM-CSF (sargramostim) as therapy of aPAP. METHODS: 18 patients with moderate-to-severe aPAP were enrolled, received baseline WLL, were randomised into either the rGM-CSF group (receiving inhaled sargramostim) or control group (no scheduled therapy) and followed for 30 months after the baseline WLL. Outcome measures included additional unscheduled "rescue" WLL for disease progression, assessment of arterial blood gases, pulmonary function, computed tomography, health status, biomarkers and adverse events. Patients requiring rescue WLL were considered to have failed their assigned intervention group. RESULTS: The primary end-point of time to first rescue WLL was longer in rGM-CSF-treated patients than controls (30 versus 18 months, n=9 per group, p=0.0078). Seven control patients (78%) and only one rGM-CSF-treated patient (11%) required rescue WLL, demonstrating a 7-fold increase in relative risk (p=0.015). Compared to controls, rGM-CSF-treated patients also had greater improvement in peripheral arterial oxygen tension, alveolar-arterial oxygen tension difference, diffusing capacity of the lungs for carbon monoxide and aPAP biomarkers. One patient from each group withdrew for personal reasons. No serious adverse events were reported. CONCLUSIONS: This long-term, prospective, randomised trial demonstrated inhaled sargramostim following WLL reduced the requirement for WLL, improved lung function and was safe in aPAP patients. WLL plus inhaled sargramostim may be useful as combined therapy for aPAP.


Subject(s)
Autoimmune Diseases , Pulmonary Alveolar Proteinosis , Pulmonary Surfactants , Humans , Pulmonary Alveolar Proteinosis/drug therapy , Pulmonary Alveolar Proteinosis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor , Prospective Studies , Administration, Inhalation , Treatment Outcome , Autoimmune Diseases/drug therapy , Pulmonary Surfactants/therapeutic use , Bronchoalveolar Lavage , Oxygen/therapeutic use , Surface-Active Agents/therapeutic use , Biomarkers
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 25(10): 1089-1094, 2023 Oct 15.
Article in Chinese | MEDLINE | ID: mdl-37905769

ABSTRACT

The male patient was referred to the hospital at 44 days old due to dyspnea after birth and inability to wean off oxygen. His brother died three days after birth due to respiratory failure. The main symptoms observed were respiratory failure, dyspnea, and hypoxemia. A chest CT scan revealed characteristic reduced opacity in both lungs with a "crazy-paving" appearance. The bronchoalveolar lavage fluid (BALF) showed periodic acid-Schiff positive proteinaceous deposits. Genetic testing indicated a compound heterozygous mutation in the ABCA3 gene. The diagnosis for the infant was congenital pulmonary alveolar proteinosis (PAP). Congenital PAP is a significant cause of challenging-to-treat respiratory failure in full-term infants. Therefore, congenital PAP should be considered in infants experiencing persistently difficult-to-treat dyspnea shortly after birth. Early utilization of chest CT scans, BALF pathological examination, and genetic testing may aid in early diagnosis.


Subject(s)
Pulmonary Alveolar Proteinosis , Respiratory Insufficiency , Infant , Infant, Newborn , Humans , Male , Bronchoalveolar Lavage/adverse effects , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/etiology , Pulmonary Alveolar Proteinosis/pathology , Dyspnea/etiology
5.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1009852

ABSTRACT

The male patient was referred to the hospital at 44 days old due to dyspnea after birth and inability to wean off oxygen. His brother died three days after birth due to respiratory failure. The main symptoms observed were respiratory failure, dyspnea, and hypoxemia. A chest CT scan revealed characteristic reduced opacity in both lungs with a "crazy-paving" appearance. The bronchoalveolar lavage fluid (BALF) showed periodic acid-Schiff positive proteinaceous deposits. Genetic testing indicated a compound heterozygous mutation in the ABCA3 gene. The diagnosis for the infant was congenital pulmonary alveolar proteinosis (PAP). Congenital PAP is a significant cause of challenging-to-treat respiratory failure in full-term infants. Therefore, congenital PAP should be considered in infants experiencing persistently difficult-to-treat dyspnea shortly after birth. Early utilization of chest CT scans, BALF pathological examination, and genetic testing may aid in early diagnosis.


Subject(s)
Infant , Infant, Newborn , Humans , Male , Bronchoalveolar Lavage/adverse effects , Pulmonary Alveolar Proteinosis/pathology , Dyspnea/etiology , Respiratory Insufficiency
6.
Vnitr Lek ; 68(8): 525-531, 2022.
Article in English | MEDLINE | ID: mdl-36575071

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.


Subject(s)
Amyloidosis , Lipoid Proteinosis of Urbach and Wiethe , Pulmonary Alveolar Proteinosis , Humans , Child , Lipoid Proteinosis of Urbach and Wiethe/pathology , Lung/metabolism , Lung/pathology , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveoli/pathology
7.
Part Fibre Toxicol ; 19(1): 69, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36539793

ABSTRACT

BACKGROUND: Many studies have shown that occupational exposure to indium and its compounds could induce lung disease. Although animal toxicological studies and human epidemiological studies suggest indium exposure may cause lung injury, inflammation, pulmonary fibrosis, emphysema, pulmonary alveolar proteinosis, and even lung cancer, related data collected from humans is currently limited and confined to single workplaces, and the early effects of exposure on the lungs are not well understood. OBJECTIVES: This study combined population studies and animal experiments to examine the links of indium with pulmonary injury, as well as its mechanism of action. A cross-sectional epidemiological study of indium-exposed workers from China was conducted to evaluate associations between occupational indium exposure and serum biomarkers of early effect. This study also compares and analyzes the causal perspectives of changes in human serum biomarkers induced by indium compound exposure and indium exposure-related rat lung pathobiology, and discusses possible avenues for their recognition and prevention. METHODS: This is a study of 57 exposed (at least 6 h per day for one year) workers from an indium ingot production plant, and 63 controls. Indium concentration in serum, urine, and airborne as exposure indices were measured by inductively coupled plasma-mass spectrometry. Sixteen serum biomarkers of pulmonary injury, inflammation, and oxidative stress were measured using ELISA. The associations between serum indium and 16 serum biomarkers were analyzed to explore the mechanism of action of indium on pulmonary injury in indium-exposed workers. Animal experiments were conducted to measure inflammatory factors levels in bronchoalveolar lavage fluid (BALF) and lung tissue protein expressions in rats. Four different forms of indium compound-exposed rat models were established (intratracheal instillation twice per week, 8 week exposure, 8 week recovery). Model I: 0, 1.2, 3, and 6 mg/kg bw indium tin oxide group; Model II: 0, 1.2, 3, and 6 mg/kg bw indium oxide (In2O3) group; Model III: 0, 0.523, 1.046, and 2.614 mg/kg bw indium sulfate (In2(SO4)3) group; Model IV: 0, 0.065, 0.65, and 1.3 mg/kg bw indium trichloride (InCl3) group. Lung pathological changes were assessed by hematoxylin & eosin, periodic acid Schiff, and Masson's staining, transmission electron microscopy, and the protein changes were determined by immunohistochemistry. RESULTS: In the production workshop, the airborne indium concentration was 78.4 µg/m3. The levels of serum indium and urine indium in indium-exposed workers were 39.3 µg/L and 11.0 ng/g creatinine. Increased lung damage markers, oxidative stress markers, and inflammation markers were found in indium-exposed workers. Serum indium levels were statistically and positively associated with the serum levels of SP-A, IL-1ß, IL-6 in indium-exposed workers. Among them, SP-A showed a duration-response pattern. The results of animal experiments showed that, with an increase in dosage, indium exposure significantly increased the levels of serum indium and lung indium, as well as the BALF levels of IL­1ß, IL­6, IL­10, and TNF­α and up-regulated the protein expression of SP-A, SP-D, KL-6, GM-CSF, NF-κB p65, and HO-1 in all rat models groups. TEM revealed that In2(SO4)3 and InCl3 are soluble and that no particles were found in lung tissue, in contrast to the non-soluble compounds (ITO and In2O3). No PAS-staining positive substance was found in the lung tissue of In2(SO4)3 and InCl3 exposure groups, whereas ITO and In2O3 rat models supported findings of pulmonary alveolar proteinosis and interstitial fibrosis seen in human indium lung disease. ITO and InCl3 can accelerate interstitial fibrosis. Findings from our in vivo studies demonstrated that intra-alveolar accumulation of surfactant (immunohistochemistry) and characteristic cholesterol clefts granulomas of indium lung disease (PAS staining) were triggered by a specific form of indium (ITO and In2O3). CONCLUSIONS: In indium-exposed workers, biomarker findings indicated lung damage, oxidative stress and an inflammatory response. In rat models of the four forms of indium encountered in a workplace, the biomarkers response to all compounds overall corresponded to that in humans. In addition, pulmonary alveolar proteinosis was found following exposure to indium tin oxide and indium oxide in the rat models, and interstitial fibrosis was found following exposure to indium tin oxide and indium trichloride, supporting previous report of human disease. Serum SP-A levels were positively associated with indium exposure and may be considered a potential biomarker of exposure and effect in exposed workers.


Subject(s)
Lung Injury , Pulmonary Alveolar Proteinosis , Pulmonary Fibrosis , Humans , Rats , Animals , Pulmonary Alveolar Proteinosis/chemically induced , Pulmonary Alveolar Proteinosis/pathology , Indium/toxicity , Indium/chemistry , Cross-Sectional Studies , Rodentia , Interleukin-6 , Inflammation , Biomarkers
8.
J Coll Physicians Surg Pak ; 32(8): S183-S185, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36210689

ABSTRACT

Pulmonary alveolar proteinosis (PAP) is a rare lung disorder in which surfactant-derived lipoproteins accumulate excessively within pulmonary alveoli, causing severe respiratory distress. It is essential to gain a better understanding of the signs to clinically diagnose PAP and include PAP among the differential diagnoses of interstitial pulmonary diseases or other diseases with similar manifestations. We describe a 2.5-year patient with atopy who presented with pulmonary infiltration, recurrent wheezing, and cough despite steroid and salbutamol administration via inhalation. High-resolution computed tomography revealed crazy-paving patterns in both lungs, suggesting PAP. An open lung biopsy revealed intra-alveolar granular amphophilic material, which was strongly positive on periodic acid-Schiff staining. The results of pulmonary-associated surfactant protein B and C gene analyses were normal. However, granulocyte-macrophage colony-stimulating factor receptor beta-protein was not detected in leucocytes, and a novel mutation was identified in the CSF2RB gene. The patient was diagnosed with PAP and treated with whole-lung lavage. Key Words: Pulmonary alveolar proteinosis, Child, Atopy, Wheezing.


Subject(s)
Pulmonary Alveolar Proteinosis , Albuterol , Bronchoalveolar Lavage/methods , Child , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Mutation , Periodic Acid , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/pathology , Rare Diseases , Respiratory Sounds , Surface-Active Agents
9.
Ecotoxicol Environ Saf ; 241: 113812, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36068741

ABSTRACT

Indium-tin oxide (ITO) was previously found to have a toxic effect on lung tissues, and oxidative stress and the inflammatory response are two important mechanisms of ITO­induced lung injury. N-acetylcysteine (NAC) has been found to exhibit antioxidant and anti­inflammatory properties. The current study aimed to evaluate the possible protective effects of NAC against ITO nanoparticle (Nano-ITO)-induced pulmonary alveolar proteinosis (PAP) in adult male Sprague-Dawley rats, especially via modulation of nuclear factor-kappa B (NF-κB) signaling. For this purpose, 50 rats were randomly allocated into five groups (10 rats each) as follows: (1) control group; (2) saline group; (3) NAC (200 mg/kg) group; (4) PAP model group receiving a repeated intratracheal dose of Nano-ITO (6 mg/kg); and (5) PAP model+NF-κB inhibitor (NAC) group pre-treated intraperitoneally with NAC (200 mg/kg) twice per week before the administration of an intratracheal dose of Nano-ITO (6 mg/kg). Rats were then euthanized under anesthesia, and their lungs were removed for histopathological and biochemical investigations. A 6 mg/kg dose of Nano-ITO markedly altered the levels of some oxidative stress biomarkers. The histological examination of Nano-ITO-exposed rats demonstrated diffused alveolar damage that involved PAP, cholesterol crystals, alveolar fibrosis, pulmonary fibrosis, and alveolar emphysema. The immunohistochemical results of Nano-ITO-exposed rats revealed strongly positive NF-κB p65 and inhibitory kappa B kinase (IKK)-ß and weakly positive inhibitor of kappa-B subunit alpha (IκB-α) staining reactivity in the nuclei of cells lining the epithelium of the bronchioles and alveoli. Moreover, Nano-ITO activated the NF-κB pathway. However, pre-treatment with NAC significantly attenuated Nano-ITO-evoked alterations in the previously mentioned parameters, highlighting their antioxidant, anti-inflammatory, and anti-apoptotic potential. The results indicated that the degree of pulmonary fibrosis and proteinosis in the NAC­treated group was improved compared with that in the Nano-ITO-induced PAP model group. The level of malondialdehyde was also decreased overall in the NAC-treated group compared with that in the Nano-ITO-induced model group, indicating that the pulmonary fibrosis degree and oxidation levels were decreased. The present study also demonstrated that NAC increased the activity of antioxidant enzyme superoxide dismutase and total antioxidant capacity, indicating that it could alleviate oxidative stress in the lung tissue of Nano-ITO­exposed rats. In addition, NAC reduced the production of pro­inflammatory cytokines interleukin (IL)­1ß, IL­6, and tumor necrosis factor (TNF)­α, and increased the levels of anti­inflammatory factor IL­10. The current study demonstrated that NAC can effectively attenuate Nano-ITO­induced lung injury by reducing oxidative damage and the inflammatory response.


Subject(s)
Lung Injury , Nanoparticles , Pulmonary Alveolar Proteinosis , Pulmonary Fibrosis , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/metabolism , Indium/toxicity , Lung , Lung Injury/chemically induced , Lung Injury/drug therapy , Lung Injury/metabolism , Male , NF-kappa B/metabolism , Nanoparticles/toxicity , Pulmonary Alveolar Proteinosis/chemically induced , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Tin Compounds , Tumor Necrosis Factor-alpha/metabolism
10.
Am J Respir Crit Care Med ; 205(9): 1016-1035, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35227171

ABSTRACT

Autoimmune pulmonary alveolar proteinosis (PAP) is a rare disease characterized by myeloid cell dysfunction, abnormal pulmonary surfactant accumulation, and innate immune deficiency. It has a prevalence of 7-10 per million; occurs in individuals of all races, geographic regions, sex, and socioeconomic status; and accounts for 90% of all patients with PAP syndrome. The most common presentation is dyspnea of insidious onset with or without cough, production of scant white and frothy sputum, and diffuse radiographic infiltrates in a previously healthy adult, but it can also occur in children as young as 3 years. Digital clubbing, fever, and hemoptysis are not typical, and the latter two indicate that intercurrent infection may be present. Low prevalence and nonspecific clinical, radiological, and laboratory findings commonly lead to misdiagnosis as pneumonia and substantially delay an accurate diagnosis. The clinical course, although variable, usually includes progressive hypoxemic respiratory insufficiency and, in some patients, secondary infections, pulmonary fibrosis, respiratory failure, and death. Two decades of research have raised autoimmune PAP from obscurity to a paradigm of molecular pathogenesis-based diagnostic and therapeutic development. Pathogenesis is driven by GM-CSF (granulocyte/macrophage colony-stimulating factor) autoantibodies, which are present at high concentrations in blood and tissues and form the basis of an accurate, commercially available diagnostic blood test with sensitivity and specificity of 100%. Although whole-lung lavage remains the first-line therapy, inhaled GM-CSF is a promising pharmacotherapeutic approach demonstrated in well-controlled trials to be safe, well tolerated, and efficacious. Research has established GM-CSF as a pulmonary regulatory molecule critical to surfactant homeostasis, alveolar stability, lung function, and host defense.


Subject(s)
Autoimmune Diseases , Pulmonary Alveolar Proteinosis , Adult , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Bronchoalveolar Lavage , Child , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/therapy
11.
Med J Malaysia ; 77(2): 261-263, 2022 03.
Article in English | MEDLINE | ID: mdl-35338641

ABSTRACT

Pulmonary alveolar proteinosis (PAP) is a rare disease and its prognosis can be improved by whole lung lavage (WLL). Herein, we present three cases with idiopathic PAP treated successfully with either single or double WLL in the same setting. All three of them presented with exertional dyspnoea with radiographic findings of pulmonary infiltrates. They showed a marked clinical and physiologic improvement post WLL. Two of them were in remission. These three cases were diagnosed using different lung biopsy modalities, including video-assisted thoracoscopic lung biopsy, computed tomography-guided percutaneous transthoracic tru-cut needle lung biopsy, and transbronchial forceps lung biopsy (TBLB), respectively. The current cases have shown that TBLB may provide adequate diagnostic yield, and the invasive surgical lung biopsy may not be necessary to achieve a definitive diagnosis.


Subject(s)
Pulmonary Alveolar Proteinosis , Biopsy , Bronchoalveolar Lavage , Humans , Lung/pathology , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/therapy , Tomography, X-Ray Computed
12.
Int J Nanomedicine ; 17: 713-731, 2022.
Article in English | MEDLINE | ID: mdl-35210771

ABSTRACT

PURPOSE: The main objective of this study was to clarify the biodistribution and in vivo toxicological effects of indium-tin oxide nanoparticles (Nano-ITO) in male rats. METHODS: Dose-response (three divided doses) and time-course studies (six exposure durations) were performed to examine Nano­ITO-induced pulmonary and systemic toxicity. At the end of the experiment, hematology and serum biochemical parameters were determined, and cytokines levels and oxidative stress were analyzed in the bronchoalveolar lavage fluid. In addition, indium biodistribution following Nano­ITO exposure was determined using inductively coupled plasma mass spectrometer to measure indium concentration in the lung, spleen, brain, liver, kidney, and testis. Rat lung tissues were also harvested for staining with hematoxylin and eosin, periodic acid Schiff stain, Masson's trichrome, and Sirius red. RESULTS: Relative lung weights were significantly increased in all Nano-ITO-exposed groups. All organs exhibited a statistically significant difference in indium levels. Rat exposure to Nano­ITO resulted in a dose-response increase in acute systemic inflammation and injury. BALF analysis revealed significantly elevated levels of lung oxidative stress, pulmonary injury, and inflammatory markers across most groups. Serum biochemistry results showed that Nano-ITO could affect the liver and renal functions of rats when exposed for 3 days. Compared with the control group, significant inflammatory responses or pathological changes were observed in the liver, kidney, and testis of rats at different sampling times and three doses examined. Histopathologically, foci of slight-to-severe pulmonary inflammatory response along with acute inflammatory, pulmonary fibrosis and alveolar proteinosis were detected, and the severity of these lesions worsened in a dose- and time-dependent manner. DISCUSSION: These findings provide novel evidence that enhanced progressive massive pulmonary fibrosis, diffuse interstitial fibrosis, and collagen accumulation play a role in the development of pulmonary alveolar proteinosis following Nano-ITO exposure.


Subject(s)
Nanoparticles , Pulmonary Alveolar Proteinosis , Animals , Indium/toxicity , Lung , Male , Nanoparticles/toxicity , Pulmonary Alveolar Proteinosis/chemically induced , Pulmonary Alveolar Proteinosis/pathology , Rats , Tin Compounds , Tissue Distribution
13.
Am J Respir Cell Mol Biol ; 66(1): 64-75, 2022 01.
Article in English | MEDLINE | ID: mdl-34586974

ABSTRACT

Tissue-resident macrophages are of vital importance as they preserve tissue homeostasis in all mammalian organs. Nevertheless, appropriate cell culture models are still limited. Here, we propose a novel culture model to study and expand murine primary alveolar macrophages (AMs), the tissue-resident macrophages of the lung, in vitro over several months. By providing a combination of granulocyte-macrophage colony-stimulating factor, TGFß, and the PPARγ activator rosiglitazone, we maintain and expand mouse ex vivo cultured AMs (mexAMs) over several months. MexAMs maintain typical morphologic features and stably express primary AM surface markers throughout in vitro culture. They respond to microbial ligands and exhibit an AM-like transcriptional profile, including the expression of AM-specific transcription factors. Furthermore, when transferred into AM-deficient mice, mexAMs efficiently engraft in the lung and fulfill key macrophage functions, leading to a significantly reduced surfactant load in those mice. Altogether, mexAMs provide a novel, simple, and versatile tool to study AM behavior in homeostasis and disease settings.


Subject(s)
Macrophages, Alveolar/metabolism , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Liver/metabolism , Lung/pathology , Lung/physiopathology , Macrophages, Alveolar/pathology , Mice, Inbred C57BL , Phenotype , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/physiopathology , Transcription, Genetic
14.
F1000Res ; 11: 1439, 2022.
Article in English | MEDLINE | ID: mdl-38779463

ABSTRACT

Background: Pulmonary alveolar proteinosis is a very rare diffuse lung disease characterized by the accumulation of amorphous and periodic acid Schiff-positive lipoproteinaceous material in the alveolar spaces due to impaired surfactant clearance by alveolar macrophages. Three main types were identified: Autoimmune, secondary and congenital. Pulmonary alveolar proteinosis has been previously reported to be associated with several systemic auto-immune diseases. Accordingly, we present the first case report of pulmonary alveolar proteinosis associated with myasthenia gravis. Case: A 27-year-old female patient, ex-smoker, developed a dyspnea on exertion in 2020. The chest X-ray detected diffuse symmetric alveolar opacities. Pulmonary infection was ruled out, particularly COVID-19 infection. The chest scan revealed the "crazy paving" pattern. The bronchoalveolar lavage showed a rosy liquid with granular acellular eosinophilic material Periodic acid-Schiff positive. According to the lung biopsy results, she was diagnosed with pulmonary alveolar proteinosis. The granulocyte macrophage colony-stimulating factor autoantibodies were negative. Nine months later, she was diagnosed with bulbar seronegative myasthenia gravis, confirmed with the electroneuromyography with repetitive nerve stimulation showing significant amplitude decrement of the trapezius and spinal muscles. She was treated with pyridostigmine, oral corticosteroids and azathioprine. Given the worsening respiratory condition of the patient, a bilateral whole lung lavage was performed with a partial resolution of symptoms. Thus, this previously unreported association was treated successfully with rituximab, including improvement of dyspnea, diplopia and muscle fatigability at six months of follow-up. Conclusions: This case emphasizes on the possible association of auto-immune disease to PAP, which could worsen the disease course, as the specific treatment does not exist yet. Hence, further studies are needed to establish clear-cut guidelines for PAP management, particularly when associated to auto-immune diseases.


Subject(s)
Myasthenia Gravis , Pulmonary Alveolar Proteinosis , Humans , Pulmonary Alveolar Proteinosis/therapy , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/complications , Female , Adult , Myasthenia Gravis/complications , Myasthenia Gravis/pathology
15.
Chest ; 160(4): e343-e346, 2021 10.
Article in English | MEDLINE | ID: mdl-34625181

ABSTRACT

CASE PRESENTATION: A 30-year-old woman was referred with increasing shortness of breath and cough in the setting of GATA2 deficiency. She initially presented 9 years previously with recurrent episodes of pneumonia and sinusitis. Genetic testing revealed a heterozygous GATA2 mutation (c.988C>T). She has since had multiple infections that have included necrotizing fasciitis of the right thumb, recurrent pilonidal infections (which required 23 procedures), esophageal candidiasis, and human papillomavirus-positive high-grade squamous intraepithelial lesion of the cervix. Serial bone marrow biopsy specimens showed persistent hypocellularity (20% to 60%) with intermittent erythroid atypia and variable detection of trisomy 8, which were concerning for evolving myelodysplastic syndrome. One year before the current admission, she was diagnosed with disseminated Mycobacterium avium complex and was treated with rifabutin, ethambutol, and azithromycin. She was taking voriconazole, acyclovir, and trimethoprim-sulfamethoxazole prophylaxis.


Subject(s)
Cough/physiopathology , Dyspnea/physiopathology , GATA2 Deficiency/physiopathology , Pulmonary Alveolar Proteinosis/diagnosis , Adult , Biopsy , Bronchoalveolar Lavage , Female , GATA2 Deficiency/complications , GATA2 Deficiency/therapy , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Deficiency Syndromes/physiopathology , Lung/pathology , Pulmonary Alveolar Proteinosis/etiology , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/physiopathology , Thoracoscopy , Tomography, X-Ray Computed
17.
Int J Mol Sci ; 22(7)2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33804918

ABSTRACT

Macrophages act as immune scavengers and are important cell types in the homeostasis of various tissues. Given the multiple roles of macrophages, these cells can also be found as tissue resident macrophages tightly integrated into a variety of tissues in which they fulfill crucial and organ-specific functions. The lung harbors at least two macrophage populations: interstitial and alveolar macrophages, which occupy different niches and functions. In this review, we provide the latest insights into the multiple roles of alveolar macrophages while unraveling the distinct factors which can influence the ontogeny and function of these cells. Furthermore, we will highlight pulmonary diseases, which are associated with dysfunctional macrophages, concentrating on congenital diseases as well as pulmonary infections and impairment of immunological pathways. Moreover, we will provide an overview about different treatment approaches targeting macrophage dysfunction. Improved knowledge of the role of macrophages in the onset of pulmonary diseases may provide the basis for new pharmacological and/or cell-based immunotherapies and will extend our understanding to other macrophage-related disorders.


Subject(s)
Macrophages, Alveolar/metabolism , Pulmonary Alveolar Proteinosis/metabolism , Animals , Homeostasis , Humans , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Surfactants/metabolism
18.
Nat Commun ; 12(1): 1032, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33589587

ABSTRACT

Pulmonary alveolar proteinosis (PAP) is a devastating lung disease caused by abnormal surfactant homeostasis, with a prevalence of 6-7 cases per million population worldwide. While mutations causing hereditary PAP have been reported, the genetic basis contributing to autoimmune PAP (aPAP) has not been thoroughly investigated. Here, we conducted a genome-wide association study of aPAP in 198 patients and 395 control participants of Japanese ancestry. The common genetic variant, rs138024423 at 6p21, in the major-histocompatibility-complex (MHC) region was significantly associated with disease risk (Odds ratio [OR] = 5.2; P = 2.4 × 10-12). HLA fine-mapping revealed that the common HLA class II allele, HLA-DRB1*08:03, strongly drove this signal (OR = 4.8; P = 4.8 × 10-12), followed by an additional independent risk allele at HLA-DPß1 amino acid position 8 (OR = 0.28; P = 3.4 × 10-7). HLA-DRB1*08:03 was also associated with an increased level of anti-GM-CSF antibody, a key driver of the disease (ß = 0.32; P = 0.035). Our study demonstrated a heritable component of aPAP, suggesting an underlying genetic predisposition toward an abnormal antibody production.


Subject(s)
Autoantibodies/genetics , Autoimmune Diseases/genetics , Genetic Predisposition to Disease , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , HLA-DRB1 Chains/genetics , Pulmonary Alveolar Proteinosis/genetics , Adult , Aged , Alleles , Asian People , Autoantibodies/biosynthesis , Autoimmune Diseases/ethnology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Case-Control Studies , Chromosomes, Human, Pair 6 , Female , Gene Expression , Gene Frequency , Genome-Wide Association Study , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , HLA-DRB1 Chains/immunology , Humans , Japan , Male , Middle Aged , Odds Ratio , Protein Isoforms/genetics , Pulmonary Alveolar Proteinosis/ethnology , Pulmonary Alveolar Proteinosis/immunology , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Surfactants/immunology , Pulmonary Surfactants/metabolism , Risk
20.
J Appl Toxicol ; 40(12): 1636-1646, 2020 12.
Article in English | MEDLINE | ID: mdl-32608070

ABSTRACT

Concerns have been raised over the safety and health of industrial workers exposed to indium oxide nanoparticles (IO-NPs) when working. IO-NPs were previously shown in vitro and in vivo to be cytotoxic, but the mechanism of pathogenesis was unclear. In this study, the effects of IO-NPs on lung cells associated with respiratory and immune barriers and the toxic effects of intercellular cascades were studied. Here IO-NPs had acute toxicity to Wistar rats over a time course (5 days post-intratracheal instillation). Following treatment epithelial cells (16HBE) or macrophages (RAW264.7) with IO-NPs or IO fine particles (IO-FPs), the damage of 16HBE cells caused by IO-NPs was serious, mainly in the mitochondrial and rough endoplasmic reticulum. The lactate dehydrogenase level also showed that cytotoxicity in vitro was more serious for IO-NPs compared with IO-FPs. The level of In3+ (examined by inductively coupled plasma mass spectrometry) in 16HBE cells was 10 times higher than that in RAW cells. In3+ , releasing from IO-NPs absorbed by 16HBE cells, could not only significantly inhibit the phagocytosis and migration of macrophages (P < .0001), but also stimulate RAW cells to secrete high levels of inflammatory cytokines. IO-NPs can directly damage pulmonary epithelial cells. The In3+ released by epithelial cells affect the phagocytosis and migration of macrophages, which may be a new point for the decrease in the clearance of alveolar surfactants and the development of IO-related pulmonary alveolar proteinosis.


Subject(s)
Epithelial Cells/drug effects , Indium/toxicity , Macrophages/drug effects , Metal Nanoparticles/toxicity , Pulmonary Alveolar Proteinosis/chemically induced , Pulmonary Alveoli/drug effects , Animals , Cell Movement/drug effects , Cytokines/metabolism , Endoplasmic Reticulum, Rough/drug effects , Endoplasmic Reticulum, Rough/metabolism , Endoplasmic Reticulum, Rough/ultrastructure , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Humans , Inflammation Mediators/metabolism , Macrophages/metabolism , Macrophages/ultrastructure , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Phagocytosis/drug effects , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/ultrastructure , RAW 264.7 Cells , Rats, Wistar , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...