Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.983
Filter
1.
PLoS One ; 19(5): e0300751, 2024.
Article in English | MEDLINE | ID: mdl-38717999

ABSTRACT

Transcriptional response to changes in oxygen concentration is mainly controlled by hypoxia-inducible transcription factors (HIFs). Besides regulation of hypoxia-responsible gene expression, HIF-3α has recently been shown to be involved in lung development and in the metabolic process of fat tissue. However, the precise mechanism for such properties of HIF-3α is still largely unknown. To this end, we generated HIF3A gene-disrupted mice by means of genome editing technology to explore the pleiotropic role of HIF-3α in development and physiology. We obtained adult mice carrying homozygous HIF3A gene mutations with comparable body weight and height to wild-type mice. However, the number of litters and ratio of homozygous mutation carriers born from the mating between homozygous mutant mice was lower than expected due to sporadic deaths on postnatal day 1. HIF3A gene-disrupted mice exhibited abnormal configuration of the lung such as a reduced number of alveoli and thickened alveolar walls. Transcriptome analysis showed, as well as genes associated with lung development, an upregulation of stearoyl-Coenzyme A desaturase 1, a pivotal enzyme for fatty acid metabolism. Analysis of fatty acid composition in the lung employing gas chromatography indicated an elevation in palmitoleic acid and a reduction in oleic acid, suggesting an imbalance in distribution of fatty acid, a constituent of lung surfactant. Accordingly, administration of glucocorticoid injections during pregnancy resulted in a restoration of normal alveolar counts and a decrease in neonatal mortality. In conclusion, these observations provide novel insights into a pivotal role of HIF-3α in the preservation of critically important structure and function of alveoli beyond the regulation of hypoxia-mediated gene expression.


Subject(s)
Animals, Newborn , Pulmonary Alveoli , Animals , Mice , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Female , Repressor Proteins/genetics , Repressor Proteins/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Male , Fatty Acids/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Apoptosis Regulatory Proteins
2.
Part Fibre Toxicol ; 21(1): 25, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760786

ABSTRACT

Exposure to indoor air pollutants (IAP) has increased recently, with people spending more time indoors (i.e. homes, offices, schools and transportation). Increased exposures of IAP on a healthy population are poorly understood, and those with allergic respiratory conditions even less so. The objective of this study, therefore, was to implement a well-characterised in vitro model of the human alveolar epithelial barrier (A549 + PMA differentiated THP-1 incubated with and without IL-13, IL-5 and IL-4) to determine the effects of a standardised indoor particulate (NIST 2583) on both a healthy lung model and one modelling a type-II (stimulated with IL-13, IL-5 and IL-4) inflammatory response (such as asthma).Using concentrations from the literature, and an environmentally appropriate exposure we investigated 232, 464 and 608ng/cm2 of NIST 2583 respectively. Membrane integrity (blue dextran), viability (trypan blue), genotoxicity (micronucleus (Mn) assay) and (pro-)/(anti-)inflammatory effects (IL-6, IL-8, IL-33, IL-10) were then assessed 24 h post exposure to both models. Models were exposed using a physiologically relevant aerosolisation method (VitroCell Cloud 12 exposure system).No changes in Mn frequency or membrane integrity in either model were noted when exposed to any of the tested concentrations of NIST 2583. A significant decrease (p < 0.05) in cell viability at the highest concentration was observed in the healthy model. Whilst cell viability in the "inflamed" model was decreased at the lower concentrations (significantly (p < 0.05) after 464ng/cm2). A significant reduction (p < 0.05) in IL-10 and a significant increase in IL-33 was seen after 24 h exposure to NIST 2583 (464, 608ng/cm2) in the "inflamed" model.Collectively, the results indicate the potential for IAP to cause the onset of a type II response as well as exacerbating pre-existing allergic conditions. Furthermore, the data imposes the importance of considering unhealthy individuals when investigating the potential health effects of IAP. It also highlights that even in a healthy population these particles have the potential to induce this type II response and initiate an immune response following exposure to IAP.


Subject(s)
Air Pollution, Indoor , Cell Survival , Particulate Matter , Humans , Air Pollution, Indoor/adverse effects , Particulate Matter/toxicity , Cell Survival/drug effects , A549 Cells , Cytokines/metabolism , THP-1 Cells , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Air Pollutants/toxicity , Inflammation/chemically induced , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology
3.
Exp Cell Res ; 438(1): 114030, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38583855

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a serious lung condition that often leads to hospitalization in intensive care units and a high mortality rate. Sevoflurane is a volatile anesthetic with growing interest for sedation in ventilated patients with ARDS. It has been shown to have potential lung-protective effects, such as reduced inflammation and lung edema, or improved arterial oxygenation. In this study, we investigated the effects of sevoflurane on lung injury in cultured human carcinoma-derived lung alveolar epithelial (A549) cells. We found that sevoflurane was associated with improved wound healing after exposure to inflammatory cytokines, with preserved cell proliferation but no effect on cell migration properties. Sevoflurane exposure was also associated with enhanced cell viability and active autophagy in A549 cells exposed to cytokines. These findings suggest that sevoflurane may have beneficial effects on lung epithelial injury by promoting alveolar epithelial wound healing and by influencing the survival and proliferation of A549 epithelial cells in vitro. Further research is needed to confirm these findings and to investigate the key cellular mechanisms explaining sevoflurane's potential effects on lung epithelial injury.


Subject(s)
Cell Proliferation , Cell Survival , Respiratory Distress Syndrome , Sevoflurane , Wound Healing , Sevoflurane/pharmacology , Humans , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/pathology , Wound Healing/drug effects , Cell Survival/drug effects , A549 Cells , Cell Proliferation/drug effects , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Cell Movement/drug effects , Anesthetics, Inhalation/pharmacology , Cytokines/metabolism , Autophagy/drug effects , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology
4.
FASEB J ; 38(8): e23612, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648494

ABSTRACT

Considerable progress has been made in understanding the function of alveolar epithelial cells in a quiescent state and regeneration mechanism after lung injury. Lung injury occurs commonly from severe viral and bacterial infections, inhalation lung injury, and indirect injury sepsis. A series of pathological mechanisms caused by excessive injury, such as apoptosis, autophagy, senescence, and ferroptosis, have been studied. Recovery from lung injury requires the integrity of the alveolar epithelial cell barrier and the realization of gas exchange function. Regeneration mechanisms include the participation of epithelial progenitor cells and various niche cells involving several signaling pathways and proteins. While alveoli are damaged, alveolar type II (AT2) cells proliferate and differentiate into alveolar type I (AT1) cells to repair the damaged alveolar epithelial layer. Alveolar epithelial cells are surrounded by various cells, such as fibroblasts, endothelial cells, and various immune cells, which affect the proliferation and differentiation of AT2 cells through paracrine during alveolar regeneration. Besides, airway epithelial cells also contribute to the repair and regeneration process of alveolar epithelium. In this review, we mainly discuss the participation of epithelial progenitor cells and various niche cells involving several signaling pathways and transcription factors.


Subject(s)
Alveolar Epithelial Cells , Lung Injury , Regeneration , Humans , Regeneration/physiology , Animals , Lung Injury/metabolism , Lung Injury/pathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Stem Cells/metabolism , Stem Cells/physiology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/metabolism , Signal Transduction , Cell Differentiation
5.
Toxicol Appl Pharmacol ; 485: 116886, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38452946

ABSTRACT

Despite extensive preclinical testing, cancer therapeutics can result in unanticipated toxicity to non-tumor tissue in patients. These toxicities may pass undetected in preclinical experiments due to modeling limitations involving poor biomimicry of 2-dimensional in vitro cell cultures and due to lack of interspecies translatability in in vivo studies. Instead, primary cells can be grown into miniature 3-dimensional structures that recapitulate morphological and functional aspects of native tissue, termed "organoids." Here, human bronchioalveolar organoids grown from primary alveolar epithelial cells were employed to model lung epithelium and investigate off-target toxicities associated with antibody-drug conjugates (ADCs). ADCs with three different linker-payload combinations (mafodotin, vedotin, and deruxtecan) were tested in bronchioalveolar organoids generated from human, rat, and nonhuman primate lung cells. Organoids demonstrated antibody uptake and changes in viability in response to ADC exposure that model in vivo drug sensitivity. RNA sequencing identified inflammatory activation in bronchioalveolar cells in response to deruxtecan. Future studies will explore specific cell populations involved in interstitial lung disease and incorporate immune cells to the culture.


Subject(s)
Immunoconjugates , Organoids , Organoids/drug effects , Organoids/pathology , Animals , Immunoconjugates/toxicity , Humans , Rats , Drug Evaluation, Preclinical/methods , Macaca fascicularis , Cells, Cultured , Toxicity Tests/methods , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology
6.
Biomed Pharmacother ; 174: 116447, 2024 May.
Article in English | MEDLINE | ID: mdl-38518606

ABSTRACT

Sepsis-induced acute respiratory distress syndrome (ARDS) causes significant fatalities worldwide and lacks pharmacological intervention. Alveolar fluid clearance (AFC) plays a pivotal role in the remission of ARDS and is markedly impaired in the pathogenesis of ARDS. Here, we demonstrated that erythropoietin could effectively ameliorate lung injury manifestations and lethality, restore lung function and promote AFC in a rat model of lipopolysaccharide (LPS)-induced ARDS. Moreover, it was proven that EPO-induced restoration of AFC occurs through triggering the total protein expression of ENaC and Na,K-ATPase channels, enhancing their protein abundance in the membrane, and suppressing their ubiquitination for degeneration. Mechanistically, the data indicated the possible involvement of EPOR/JAK2/STAT3/SGK1/Nedd4-2 signaling in this process, and the pharmacological inhibition of the pathway markedly eliminated the stimulating effects of EPO on ENaC and Na,K-ATPase, and subsequently reversed the augmentation of AFC by EPO. Consistently, in vitro studies of alveolar epithelial cells paralleled with that EPO upregulated the expression of ENaC and Na,K-ATPase, and patch-clamp studies further demonstrated that EPO substantially strengthened sodium ion currents. Collectively, EPO could effectively promote AFC by improving ENaC and Na,K-ATPase protein expression and abundance in the membrane, dependent on inhibition of ENaC and Na,K-ATPase ubiquitination, and resulting in diminishing LPS-associated lung injuries.


Subject(s)
Epithelial Sodium Channels , Erythropoietin , Rats, Sprague-Dawley , Respiratory Distress Syndrome , Sepsis , Sodium-Potassium-Exchanging ATPase , Ubiquitination , Animals , Epithelial Sodium Channels/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Erythropoietin/pharmacology , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Ubiquitination/drug effects , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/metabolism , Male , Rats , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Lipopolysaccharides , Signal Transduction/drug effects , Disease Models, Animal
8.
Respir Res ; 25(1): 37, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238778

ABSTRACT

Acute respiratory distress syndrome (ARDS) alters the dynamics of lung inflation during mechanical ventilation. Repetitive alveolar collapse and expansion (RACE) predisposes the lung to ventilator-induced lung injury (VILI). Two broad approaches are currently used to minimize VILI: (1) low tidal volume (LVT) with low-moderate positive end-expiratory pressure (PEEP); and (2) open lung approach (OLA). The LVT approach attempts to protect already open lung tissue from overdistension, while simultaneously resting collapsed tissue by excluding it from the cycle of mechanical ventilation. By contrast, the OLA attempts to reinflate potentially recruitable lung, usually over a period of seconds to minutes using higher PEEP used to prevent progressive loss of end-expiratory lung volume (EELV) and RACE. However, even with these protective strategies, clinical studies have shown that ARDS-related mortality remains unacceptably high with a scarcity of effective interventions over the last two decades. One of the main limitations these varied interventions demonstrate to benefit is the observed clinical and pathologic heterogeneity in ARDS. We have developed an alternative ventilation strategy known as the Time Controlled Adaptive Ventilation (TCAV) method of applying the Airway Pressure Release Ventilation (APRV) mode, which takes advantage of the heterogeneous time- and pressure-dependent collapse and reopening of lung units. The TCAV method is a closed-loop system where the expiratory duration personalizes VT and EELV. Personalization of TCAV is informed and tuned with changes in respiratory system compliance (CRS) measured by the slope of the expiratory flow curve during passive exhalation. Two potentially beneficial features of TCAV are: (i) the expiratory duration is personalized to a given patient's lung physiology, which promotes alveolar stabilization by halting the progressive collapse of alveoli, thereby minimizing the time for the reopened lung to collapse again in the next expiration, and (ii) an extended inspiratory phase at a fixed inflation pressure after alveolar stabilization gradually reopens a small amount of tissue with each breath. Subsequently, densely collapsed regions are slowly ratcheted open over a period of hours, or even days. Thus, TCAV has the potential to minimize VILI, reducing ARDS-related morbidity and mortality.


Subject(s)
Respiratory Distress Syndrome , Ventilator-Induced Lung Injury , Humans , Respiration, Artificial/methods , Lung/pathology , Pulmonary Alveoli/pathology , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/pathology , Continuous Positive Airway Pressure/methods , Tidal Volume , Ventilator-Induced Lung Injury/prevention & control , Ventilator-Induced Lung Injury/pathology
10.
Indian J Pathol Microbiol ; 66(4): 834-838, 2023.
Article in English | MEDLINE | ID: mdl-38084542

ABSTRACT

Introduction: The predominant pathology noted in the lungs of patients dying of COVID-19 is reported to be diffuse alveolar damage (DAD). Other studies have identified microthrombi to be a prominent finding of lung injury in patients affected by COVID-19. We describe the lung histopathological findings in fifteen cases of COVID-19 who died from the disease with the aim of reporting the microscopic changes in the lungs of patients dying from this disease. Materials and Methods: Lung tissues from fifteen consecutive autopsy cases of COVID-19 were studied for gross and microscopic features. The case history of the deaths was noted, and the information was analyzed. The lung damage seen was graded on a semiquantitative scale on the basis of the percentage of tissue involved. Results: Gross examination of the lungs showed multiple foci of consolidation mainly in the lower lobes of the lungs as the most commonly encountered finding. The other significant pattern was congested and edematous lungs with areas of consolidation. Microscopic assessment of lung sections showed 8 out of the 15 cases showing changes of the exudative phase of diffuse alveolar damage, whereas two cases were in the proliferative phase. Hyaline membranes were one of the common findings along with intra-alveolar edema and interstitial edema. Four cases showed changes in organizing phase. Other findings were microthrombi formation, fungal abscesses, dilated and collapsed alveoli, intra-alveolar hemorrhage, and acute neutrophilic pneumonia. Conclusion: DADand interstitial pneumonitis were the most striking features in our autopsy study. Features of different phases of diffuse alveolar damage were seen to coexist in the same patient indicating the temporal heterogeneity of the ongoing lung injury in these patients.


Subject(s)
COVID-19 , Lung Injury , Pneumonia , Humans , COVID-19/pathology , Lung Injury/pathology , Lung/pathology , Pulmonary Alveoli/pathology , Edema/pathology
11.
Lab Invest ; 103(11): 100233, 2023 11.
Article in English | MEDLINE | ID: mdl-37567389

ABSTRACT

Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal congenital lung disorder that presents shortly after birth with respiratory failure and therapy-resistant pulmonary hypertension. It is associated with heterozygous point mutations and genomic deletions that involve the FOXF1 gene or its upstream regulatory region. Patients are unresponsive to the intensive treatment regimens and suffer unnecessarily because ACDMPV is not always timely recognized and histologic diagnosis is invasive and time consuming. Here, we demonstrate the usefulness of a noninvasive, fast genetic test for FOXF1 variants that we previously developed to rapidly diagnose ACDMPV and reduce the time of hospitalization.


Subject(s)
Persistent Fetal Circulation Syndrome , Pulmonary Alveoli/abnormalities , Infant, Newborn , Humans , Persistent Fetal Circulation Syndrome/diagnosis , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , Clinical Relevance , Pulmonary Alveoli/pathology , Forkhead Transcription Factors/genetics
12.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37581936

ABSTRACT

Secondary lung infection by inhaled Staphylococcus aureus (SA) is a common and lethal event for individuals infected with influenza A virus (IAV). How IAV disrupts host defense to promote SA infection in lung alveoli, where fatal lung injury occurs, is not known. We addressed this issue using real-time determinations of alveolar responses to IAV in live, intact, perfused lungs. Our findings show that IAV infection blocked defensive alveolar wall liquid (AWL) secretion and induced airspace liquid absorption, thereby reversing normal alveolar liquid dynamics and inhibiting alveolar clearance of inhaled SA. Loss of AWL secretion resulted from inhibition of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel in the alveolar epithelium, and airspace liquid absorption was caused by stimulation of the alveolar epithelial Na+ channel (ENaC). Loss of AWL secretion promoted alveolar stabilization of inhaled SA, but rescue of AWL secretion protected against alveolar SA stabilization and fatal SA-induced lung injury in IAV-infected mice. These findings reveal a central role for AWL secretion in alveolar defense against inhaled SA and identify AWL inhibition as a critical mechanism of IAV lung pathogenesis. AWL rescue may represent a new therapeutic approach for IAV-SA coinfection.


Subject(s)
Coinfection , Influenza A virus , Influenza, Human , Lung Injury , Mice , Animals , Humans , Influenza, Human/pathology , Lung Injury/pathology , Coinfection/pathology , Pulmonary Alveoli/pathology , Lung/pathology
13.
Pediatr Pulmonol ; 58(10): 2746-2749, 2023 10.
Article in English | MEDLINE | ID: mdl-37401868

ABSTRACT

We report the case of a preterm infant who died at 10 months of age with severe bronchopulmonary dysplasia (sBPD) with refractory pulmonary hypertension and respiratory failure who had striking histologic features compatible with the diagnosis of alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) but without genetic confirmation of the diagnosis. We further demonstrate dramatic reductions in lung FOXF1 and TMEM100 content in sBPD, suggesting common mechanistic links between ACDMPV and sBPD with impaired FOXF1 signaling.


Subject(s)
Bronchopulmonary Dysplasia , Persistent Fetal Circulation Syndrome , Humans , Infant , Bronchopulmonary Dysplasia/diagnosis , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Forkhead Transcription Factors/genetics , Gene Expression , Infant, Premature , Lung/diagnostic imaging , Lung/pathology , Membrane Proteins/genetics , Persistent Fetal Circulation Syndrome/diagnosis , Persistent Fetal Circulation Syndrome/genetics , Pulmonary Alveoli/pathology
14.
Radiographics ; 43(7): e220176, 2023 07.
Article in English | MEDLINE | ID: mdl-37289644

ABSTRACT

Diffuse alveolar damage (DAD), which represents the pathologic changes seen after acute lung injury, is caused by damage to all three layers of the alveolar wall and can ultimately result in alveolar collapse with loss of the normal pulmonary architecture. DAD has an acute phase that predominantly manifests as airspace disease at CT owing to filling of the alveoli with cells, plasma fluids, and hyaline membranes. DAD then evolves into a heterogeneous organizing phase, with mixed airspace and interstitial disease characterized by volume loss, architectural distortion, fibrosis, and parenchymal loss. Patients with DAD have a severe clinical course and typically require prolonged mechanical ventilation, which may result in ventilator-induced lung injury. In those patients who survive DAD, the lungs will remodel over time, but most will have residual findings at chest CT. Organizing pneumonia (OP) is a descriptive term for a histologic pattern characterized by intra-alveolar fibroblast plugs. The significance and pathogenesis of OP are controversial. Some authors regard it as part of a spectrum of acute lung injury, while others consider it a marker of acute or subacute lung injury. At CT, OP manifests with various forms of airspace disease that are most commonly bilateral and relatively homogeneous in appearance at individual time points. Patients with OP most often have a mild clinical course, although some may have residual findings at CT. In patients with DAD and OP, imaging findings can be combined with clinical information to suggest the diagnosis in many cases, with biopsy reserved for difficult cases with atypical findings or clinical manifestations. To best participate in the multidisciplinary approach to patients with lung injury, radiologists must not only recognize these entities but also describe them with consistent and meaningful terminology, examples of which are emphasized in the article. © RSNA, 2023 See the invited commentary by Kligerman et al in this issue. Quiz questions for this article are available in the supplemental material.


Subject(s)
Acute Lung Injury , Pneumonia , Humans , Lung/diagnostic imaging , Pulmonary Alveoli/pathology , Disease Progression , Tomography, X-Ray Computed/methods , Acute Lung Injury/pathology
15.
Clin Respir J ; 17(6): 499-506, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37156734

ABSTRACT

BACKGROUND: Acute fibrinous and organizing pneumonia (AFOP) is a rare histological interstitial pneumonia pattern characterized by patches of "fibrin balls" distributed within the alveoli and organizing pneumonia. Currently, there is no consensus on the diagnosis and treatment of this disease. METHODS: We present the case of a 44-year-old male with AFOP secondary to Mycobacterium tuberculosis. We have further reviewed organizing pneumonia (OP) and AFOP caused by tuberculosis. CONCLUSION: Tuberculosis secondary to OP or AFOP is rare and challenging to diagnose. We need to constantly adjust the treatment plan based on the patient's symptoms, test results, and response to treatment in order to arrive at an accurate diagnosis and maximize treatment efficacy.


Subject(s)
Cryptogenic Organizing Pneumonia , Organizing Pneumonia , Pneumonia , Tuberculosis, Pulmonary , Male , Humans , Adult , Pneumonia/diagnosis , Lung/pathology , Tuberculosis, Pulmonary/complications , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/drug therapy , Pulmonary Alveoli/pathology , Cryptogenic Organizing Pneumonia/diagnostic imaging , Cryptogenic Organizing Pneumonia/drug therapy
16.
Cell Mol Life Sci ; 80(6): 145, 2023 May 11.
Article in English | MEDLINE | ID: mdl-37166489

ABSTRACT

Alveolar epithelial type II cells (AT2s) together with AT1s constitute the epithelial lining of lung alveoli. In contrast to the large flat AT1s, AT2s are cuboidal and smaller. In addition to surfactant production, AT2s also serve as prime alveolar progenitors in homeostasis and play an important role during regeneration/repair. Based on different lineage tracing strategies in mice and single-cell transcriptomic analysis, recent reports highlight the heterogeneous nature of AT2s. These studies present compelling evidence for the presence of stable or transitory AT2 subpopulations with distinct marker expression, signaling pathway activation and functional properties. Despite demonstrated progenitor potentials of AT2s in maintaining homeostasis, through self-renewal and differentiation to AT1s, the exact identity, full progenitor potential and regulation of these progenitor cells, especially in the context of human diseases remain unclear. We recently identified a novel subset of AT2 progenitors named "Injury-Activated Alveolar Progenitors" (IAAPs), which express low levels of Sftpc, Sftpb, Sftpa1, Fgfr2b and Etv5, but are highly enriched for the expression of the surface receptor programmed cell death-ligand 1 (Pd-l1). IAAPs are quiescent during lung homeostasis but activated upon injury with the potential to proliferate and differentiate into AT2s. Significantly, a similar population of PD-L1 positive cells expressing intermediate levels of SFTPC are found to be expanded in human IPF lungs. We summarize here the current understanding of this newly discovered AT2 progenitor subpopulation and also try to reconcile the relationship between different AT2 stem cell subpopulations regarding their progenitor potential, regulation, and relevance to disease pathogenesis and therapeutic interventions.


Subject(s)
B7-H1 Antigen , Lung , Mice , Humans , Animals , B7-H1 Antigen/metabolism , Lung/metabolism , Alveolar Epithelial Cells , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Cell Differentiation/physiology
17.
Virchows Arch ; 482(3): 605-613, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36849560

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a serious complication of systemic inflammatory response syndrome, and diffuse alveolar damage (DAD) is a histological manifestation of ARDS. Endothelial cell injury is mainly responsible for ARDS. Many neutrophils and macrophages/monocytes, which are inflammatory cells that play a role in innate immunity, infiltrate the lung tissue in DAD. In recent years, it has become clear that CD8 plays an important role not only in the acquired immune system, but also in the innate immune system. Non-antigen-activated bystander CD8 + T cells express the unique granzyme B (GrB) + /CD25-/programmed cell death-1 (PD-1)-phenotype. The involvement of bystander CD8 + T cells in lung tissue in DAD is an unexplored field. This study aimed to determine whether bystander CD8 is involved in DAD. Twenty-three consecutive autopsy specimens were retrieved from patients with DAD, and the phenotypes of infiltrating lymphocytes in the DAD lesions were evaluated using immunohistochemistry. In most cases, the number of CD8 + T cells was higher than that of CD4 + T cells, and many GrB + cells were also observed. However, the number of CD25 + and PD-1 + cells was low. We conclude that bystander CD8 + T cells may be involved in cell injury during the development of DAD.


Subject(s)
Pulmonary Alveoli , Respiratory Distress Syndrome , Humans , Pulmonary Alveoli/pathology , Programmed Cell Death 1 Receptor , Lung/pathology , CD8-Positive T-Lymphocytes , Respiratory Distress Syndrome/pathology
18.
Respir Res ; 24(1): 12, 2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36631871

ABSTRACT

BACKGROUND: Pulmonary hypoperfusion is common in children with congenital heart diseases (CHDs) or pulmonary hypertension (PH) and causes adult pulmonary dysplasia. Systematic reviews have shown that some children with CHDs or PH have mitigated clinical outcomes with COVID-19. Understanding the effects of pulmonary hypoperfusion on postnatal alveolar development may aid in the development of methods to improve the pulmonary function of children with CHDs or PH and improve their care during the COVID-19 pandemic, which is characterized by cytokine storm and persistent inflammation. METHODS AND RESULTS: We created a neonatal pulmonary hypoperfusion model through pulmonary artery banding (PAB) surgery at postnatal day 1 (P1). Alveolar dysplasia was confirmed by gross and histological examination at P21. Transcriptomic analysis of pulmonary tissues at P7(alveolar stage 2) and P14(alveolar stage 4) revealed that the postnatal alveolar development track had been changed due to pulmonary hypoperfusion. Under the condition of pulmonary hypoperfusion, the cell-cell communication and axon guidance, which both determine the final number of alveoli, were lost; instead, there was hyperactive cell cycle activity. The transcriptomic results were further confirmed by the examination of axon guidance and cell cycle markers. Because axon guidance controls inflammation and immune cell activation, the loss of axon guidance may explain the lack of severe COVID-19 cases among children with CHDs or PH accompanied by pulmonary hypoperfusion. CONCLUSIONS: This study suggested that promoting cell-cell communication or supplementation with guidance molecules may treat pulmonary hypoperfusion-induced alveolar dysplasia, and that COVID-19 is less likely to cause a cytokine storm in children with CHD or PH accompanied by pulmonary hypoperfusion.


Subject(s)
COVID-19 , Hypertension, Pulmonary , Child , Infant, Newborn , Humans , Axon Guidance , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/pathology , Pandemics , COVID-19/metabolism , Pulmonary Alveoli/pathology , Hypertension, Pulmonary/metabolism , Cell Communication
19.
Am J Case Rep ; 24: e938456, 2023 Jan 07.
Article in English | MEDLINE | ID: mdl-36609435

ABSTRACT

BACKGROUND Pulmonary alveolar microlithiasis (PAM) is an uncommon pulmonary disease characterized by deposition of microliths in the alveoli. In this report, we describe the first ever documented case from the Indonesian population of an adult patient who was diagnosed with PAM based on clinical and pathognomonic radiological findings. CASE REPORT A 57-year-old man with a 12-year history of progressive shortness of breath on exertion was admitted to our center. When the lungs were listened to, there were coarse crackles and wheezing during inspiration, and the vesicular sound was lower in all thoracic regions. Cardiac auscultation was unremarkable, with fingers having a clubbed drumstick appearance. Bronchoscopy revealed all patent branches of the bronchial tree. Unfortunately, the microliths were absent, and the histology findings from bronchoalveolar lavage and transbronchial lung biopsy were inconclusive. Radiologic features of a chest radiograph show the characteristic finding of multiple diffuse micronodules with a high density in both lungs. A high-resolution computed tomography (HRCT) scan corroborated the typical findings of extensive intraparenchymal calcified micronodules with diffuse ground-glass attenuation areas. Black pleural line signs were also seen. CONCLUSIONS PAM is a rare disease with a chronic clinical course and varying manifestations according to phase, but progressive deterioration may result in a poor prognosis. It is particularly important for clinicians to be able to narrow down the differential diagnosis of multiple diffuse micronodules of the lungs. When a non-invasive method of diagnosis is preferred, chest X-rays and, even better, HRCT should be used to find the characteristic features of alveolar microlithiasis.


Subject(s)
Lung Diseases , Adult , Humans , Male , Middle Aged , Lung/diagnostic imaging , Lung/pathology , Lung Diseases/diagnostic imaging , Lung Diseases/pathology , Pulmonary Alveoli/diagnostic imaging , Pulmonary Alveoli/pathology
20.
JCI Insight ; 8(1)2023 01 10.
Article in English | MEDLINE | ID: mdl-36454643

ABSTRACT

Dysfunction of alveolar epithelial type 2 cells (AEC2s), the facultative progenitors of lung alveoli, is implicated in pulmonary disease pathogenesis, highlighting the importance of human in vitro models. However, AEC2-like cells in culture have yet to be directly compared to their in vivo counterparts at single-cell resolution. Here, we performed head-to-head comparisons among the transcriptomes of primary (1°) adult human AEC2s, their cultured progeny, and human induced pluripotent stem cell-derived AEC2s (iAEC2s). We found each population occupied a distinct transcriptomic space with cultured AEC2s (1° and iAEC2s) exhibiting similarities to and differences from freshly purified 1° cells. Across each cell type, we found an inverse relationship between proliferative and maturation states, with preculture 1° AEC2s being most quiescent/mature and iAEC2s being most proliferative/least mature. Cultures of either type of human AEC2s did not generate detectable alveolar type 1 cells in these defined conditions; however, a subset of iAEC2s cocultured with fibroblasts acquired a transitional cell state described in mice and humans to arise during fibrosis or following injury. Hence, we provide direct comparisons of the transcriptomic programs of 1° and engineered AEC2s, 2 in vitro models that can be harnessed to study human lung health and disease.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Animals , Mice , Transcriptome , Alveolar Epithelial Cells/metabolism , Lung/pathology , Pulmonary Alveoli/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...