Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 800
Filter
1.
Vet Immunol Immunopathol ; 273: 110791, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38824909

ABSTRACT

Infectious bronchitis virus (IBV) strains of the Delmarva (DMV)/1639 genotype have been causing false layer syndrome (FLS) in the Eastern Canadian layer operations since the end of 2015. FLS is characterized by the development of cystic oviducts in layer pullets infected at an early age. Currently, there are no homologous vaccines for the control of this IBV genotype. Our previous research showed that a heterologous vaccination regimen incorporating Massachusetts (Mass) and Connecticut (Conn) IBV types protects layers against DMV/1639 genotype IBV. The aim of this study was to investigate the role of maternal antibodies conferred by breeders received the same vaccination regimen in the protection against the development of DMV/1639-induced FLS in pullets. Maternal antibody-positive (MA+) and maternal antibody-negative (MA-) female progeny chicks were challenged at 1 day of age and kept under observation for 16 weeks. Oviductal cystic formations were observed in 3 of 14 birds (21.4 %) in the MA- pullets, while the lesions were notably absent in the MA+ pullets. Milder histopathological lesions were observed in the examined tissues of the MA+ pullets. However, the maternal derived immunity failed to demonstrate protection against the damage to the tracheal ciliary activity, viral shedding, and viral tissue distribution. Overall, this study underscores the limitations of maternal derived immunity in preventing certain aspects of viral pathogenesis, emphasizing the need for comprehensive strategies to address different aspects of IBV infection.


Subject(s)
Antibodies, Viral , Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Viral Vaccines , Animals , Infectious bronchitis virus/immunology , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/virology , Chickens/immunology , Chickens/virology , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Immunity, Maternally-Acquired , Trachea/immunology , Trachea/virology , Oviducts/immunology , Oviducts/pathology , Oviducts/virology
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167216, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38718843

ABSTRACT

Studies have highlighted an upregulation of PD-1 expression in CD4+ T cells, which accelerates lung fibrosis by activating the IL-17/STAT3 pathway, leading to IL-17A and TGF-ß1 secretion. However, the relation with traumatic tracheal stenosis (TS) remains unexplored. Our analysis found significant increases in PD-1+CD4+ T cells, IL-17A, and TGF-ß1 in the TS patients (n = 10). The cellular model used CD4+ T cells co-cultured with bronchial fibroblasts while the animal model used a nylon brush to scrape the damaged tracheal mucosa. Interventions with PD-1 and STAT3 inhibitors both in vitro (n = 5) and in vivo (n = 6) showed decreased expression of TGF-ß1 and IL-17A in CD4+ T cells, decreased collagen I synthesis in vitro, and reduced tractal fibrosis in vivo. Furthermore, PD-1's modulation of the STAT3 was evident. This research unveils PD-1+CD4+ T cells' role in TS, thus suggesting a novel immunotherapeutic strategy to counteract tracheal fibrosis.


Subject(s)
CD4-Positive T-Lymphocytes , Interleukin-17 , Programmed Cell Death 1 Receptor , STAT3 Transcription Factor , Signal Transduction , Tracheal Stenosis , STAT3 Transcription Factor/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Interleukin-17/metabolism , Interleukin-17/immunology , Humans , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Tracheal Stenosis/pathology , Tracheal Stenosis/metabolism , Tracheal Stenosis/immunology , Male , Female , Adult , Middle Aged , Transforming Growth Factor beta1/metabolism , Mice , Fibrosis , Disease Models, Animal , Trachea/pathology , Trachea/metabolism , Trachea/immunology
3.
Viruses ; 16(4)2024 04 14.
Article in English | MEDLINE | ID: mdl-38675946

ABSTRACT

Infectious bronchitis virus (IBV) is a highly contagious Gammacoronavirus causing moderate to severe respiratory infection in chickens. Understanding the initial antiviral response in the respiratory mucosa is crucial for controlling viral spread. We aimed to characterize the impact of IBV Delmarva (DMV)/1639 and IBV Massachusetts (Mass) 41 at the primary site of infection, namely, in chicken tracheal epithelial cells (cTECs) in vitro and the trachea in vivo. We hypothesized that some elements of the induced antiviral responses are distinct in both infection models. We inoculated cTECs and infected young specific pathogen-free (SPF) chickens with IBV DMV/1639 or IBV Mass41, along with mock-inoculated controls, and studied the transcriptome using RNA-sequencing (RNA-seq) at 3 and 18 h post-infection (hpi) for cTECs and at 4 and 11 days post-infection (dpi) in the trachea. We showed that IBV DMV/1639 and IBV Mass41 replicate in cTECs in vitro and the trachea in vivo, inducing host mRNA expression profiles that are strain- and time-dependent. We demonstrated the different gene expression patterns between in vitro and in vivo tracheal IBV infection. Ultimately, characterizing host-pathogen interactions with various IBV strains reveals potential mechanisms for inducing and modulating the immune response during IBV infection in the chicken trachea.


Subject(s)
Chickens , Coronavirus Infections , Gene Expression Profiling , Infectious bronchitis virus , Poultry Diseases , Trachea , Animals , Trachea/virology , Trachea/immunology , Chickens/virology , Infectious bronchitis virus/physiology , Infectious bronchitis virus/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Poultry Diseases/virology , Poultry Diseases/immunology , Poultry Diseases/genetics , Epithelial Cells/virology , Epithelial Cells/immunology , Transcriptome , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Virus Replication , Specific Pathogen-Free Organisms
4.
Nature ; 626(7998): 385-391, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096903

ABSTRACT

A limitation of current SARS-CoV-2 vaccines is that they provide minimal protection against infection with current Omicron subvariants1,2, although they still provide protection against severe disease. Enhanced mucosal immunity may be required to block infection and onward transmission. Intranasal administration of current vaccines has proven inconsistent3-7, suggesting that alternative immunization strategies may be required. Here we show that intratracheal boosting with a bivalent Ad26-based SARS-CoV-2 vaccine results in substantial induction of mucosal humoral and cellular immunity and near-complete protection against SARS-CoV-2 BQ.1.1 challenge. A total of 40 previously immunized rhesus macaques were boosted with a bivalent Ad26 vaccine by the intramuscular, intranasal and intratracheal routes, or with a bivalent mRNA vaccine by the intranasal route. Ad26 boosting by the intratracheal route led to a substantial expansion of mucosal neutralizing antibodies, IgG and IgA binding antibodies, and CD8+ and CD4+ T cell responses, which exceeded those induced by Ad26 boosting by the intramuscular and intranasal routes. Intratracheal Ad26 boosting also led to robust upregulation of cytokine, natural killer, and T and B cell pathways in the lungs. After challenge with a high dose of SARS-CoV-2 BQ.1.1, intratracheal Ad26 boosting provided near-complete protection, whereas the other boosting strategies proved less effective. Protective efficacy correlated best with mucosal humoral and cellular immune responses. These data demonstrate that these immunization strategies induce robust mucosal immunity, suggesting the feasibility of developing vaccines that block respiratory viral infections.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunity, Mucosal , Immunization, Secondary , Macaca mulatta , SARS-CoV-2 , Animals , Humans , Administration, Intranasal , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Cytokines/immunology , Immunity, Mucosal/immunology , Immunization, Secondary/methods , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Injections, Intramuscular , Killer Cells, Natural/immunology , Lung/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , mRNA Vaccines/administration & dosage , mRNA Vaccines/immunology , SARS-CoV-2/classification , SARS-CoV-2/immunology , Trachea/immunology , Trachea/virology
5.
Avian Dis ; 65(3): 364-372, 2021 09.
Article in English | MEDLINE | ID: mdl-34427409

ABSTRACT

We previously reported that recombinant Newcastle disease virus LaSota (rLS) expressing infectious bronchitis virus (IBV) Arkansas (Ark)-type trimeric spike (S) ectodomain (Se; rLS/ArkSe) provides suboptimal protection against IBV challenge. We have now developed rLS expressing chicken granulocyte-macrophage colony-stimulating factor (GMCSF) and IBV Ark Se in an attempt to enhance vaccine effectiveness. In the current study, we first compared protection conferred by vaccination with rLS/ArkSe and rLS/ArkSe.GMCSF. Vaccinated chickens were challenged with virulent Ark, and protection was determined by clinical signs, viral load, and tracheal histomorphometry. Results showed that coexpression of GMCSF and the Se from rLS significantly reduced tracheal viral load and tracheal lesions compared with chickens vaccinated with rLS/ArkSe. In a second experiment, we evaluated enhancement of cross-protection of a Massachusetts (Mass) attenuated vaccine by priming or boosting with rLS/ArkSe.GMCSF. Vaccinated chickens were challenged with Ark, and protection was evaluated. Results show that priming or boosting with the recombinant virus significantly increased cross-protection conferred by Mass against Ark virulent challenge. Greater reductions of viral loads in both trachea and lachrymal fluids were observed in chickens primed with rLS/ArkSe.GMCSF and boosted with Mass. Consistently, Ark Se antibody levels measured with recombinant Ark Se protein-coated ELISA plates 14 days after boost were significantly higher in these chickens. Unexpectedly, the inverse vaccination scheme, that is, priming with Mass and boosting with the recombinant vaccine, proved somewhat less effective. We concluded that a prime and boost strategy by using rLS/ArkSe.GMCSF and the worldwide ubiquitous Mass attenuated vaccine provides enhanced cross-protection. Thus, rLS/GMCSF coexpressing the Se of regionally relevant IBV serotypes could be used in combination with live Mass to protect against regionally circulating IBV variant strains.


Protección incrementada por el virus recombinante de la enfermedad de Newcastle que expresa el ectodominio de la espícula del virus de la bronquitis infecciosa y el factor estimulante de colonias de granulocitos y macrófagos del pollo. Anteriormente se reportó que la cepa LaSota recombinante del virus de la enfermedad de Newcastle (rLS) que expresa el ectodominio de la espícula trimérica (S) de tipo Arkansas (Ark) del virus de la bronquitis infecciosa (IBV) (Se; rLS/ArkSe) proporciona una protección subóptima contra la exposición al virus de la bronquitis infecciosa. Ahora se ha desarrollado hemos desarrollado una cepa LaSota recombinante (rLS) que expresa el factor estimulante de colonias de granulocitos y macrófagos de pollo (GMCSF) y la espícula del virus de bronquitis Arkansas en un intento para mejorar la efectividad de la vacuna. En el estudio actual, primero se comparó la protección conferida por la vacunación con los virus rLS/ArkSe y rLS/ArkSe.GMCSF. Los pollos vacunados se desafiaron con un virus Arkansas virulento y la protección se determinó mediante los signos clínicos, la carga viral y la histomorfometría de la tráquea. Los resultados mostraron que la coexpresión del factor estimulante de colonias de granulocitos y macrófagos de pollo y la espícula de la cepa recombinante LaSota redujo significativamente la carga viral traqueal y las lesiones traqueales en comparación con los pollos vacunados con el virus rLS/ArkSe. En un segundo experimento, se evaluó el incremento en la protección cruzada por una vacuna atenuada de Massachusetts (Mass) mediante la primovacunación o la vacunación de refuerzo con rLS/ArkSe.GMCSF. Los pollos vacunados fueron desafiados con el virus Arkansas y se evaluó la protección. Los resultados mostraron que la primovacunación o la vacunación de refuerzo con el virus recombinante aumentó significativamente la protección cruzada conferida por el virus Massachusetts contra el desafío virulento con el virus Arkansas. Se observaron mayores reducciones de las cargas virales en los fluidos traqueales y lagrimales en pollos primovacunadoss con rLS/ArkSe.GMCSF y con vacunación de refuerzo con Massachusetts. De manera consistente, los niveles de anticuerpos Ark Se medidos con placas de ELISA recubiertas con proteína Ark Se recombinante a los 14 días después del refuerzo fueron significativamente más altos en estos pollos. De manera inesperada, el esquema de vacunación inverso, es decir, la primovacunación con Massachusetts y el refuerzo con la vacuna recombinante, resultó menos efectivo. Se concluye que una estrategia de primovacunación y refuerzo mediante el uso de rLS/ArkSe.GMCSF y la vacuna atenuada con Massachusetts usada en todo el mundo proporciona una protección cruzada aumentada. Por tanto, el virus rLS/GMCSF que coexpresa la proteína de la espícula de los serotipos regionales relevantes de bronquitis infecciosa podría usarse en combinación con una vacuna viva Massachusetts para proteger contra cepas variantes del virus de la bronquitis infecciosa que circulan regionalmente.


Subject(s)
Coronavirus Infections/veterinary , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Infectious bronchitis virus/immunology , Newcastle disease virus/genetics , Poultry Diseases/prevention & control , Spike Glycoprotein, Coronavirus/immunology , Animals , Antibodies, Viral/immunology , Chickens/genetics , Chickens/immunology , Chickens/virology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Cross Protection , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Infectious bronchitis virus/chemistry , Infectious bronchitis virus/genetics , Infectious bronchitis virus/physiology , Newcastle disease virus/metabolism , Poultry Diseases/immunology , Poultry Diseases/virology , Protein Domains , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Trachea/immunology , Trachea/virology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Load
6.
Nat Commun ; 12(1): 5152, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446707

ABSTRACT

The immunological features that distinguish COVID-19-associated acute respiratory distress syndrome (ARDS) from other causes of ARDS are incompletely understood. Here, we report the results of comparative lower respiratory tract transcriptional profiling of tracheal aspirate from 52 critically ill patients with ARDS from COVID-19 or from other etiologies, as well as controls without ARDS. In contrast to a "cytokine storm," we observe reduced proinflammatory gene expression in COVID-19 ARDS when compared to ARDS due to other causes. COVID-19 ARDS is characterized by a dysregulated host response with increased PTEN signaling and elevated expression of genes with non-canonical roles in inflammation and immunity. In silico analysis of gene expression identifies several candidate drugs that may modulate gene expression in COVID-19 ARDS, including dexamethasone and granulocyte colony stimulating factor. Compared to ARDS due to other types of viral pneumonia, COVID-19 is characterized by impaired interferon-stimulated gene (ISG) expression. The relationship between SARS-CoV-2 viral load and expression of ISGs is decoupled in patients with COVID-19 ARDS when compared to patients with mild COVID-19. In summary, assessment of host gene expression in the lower airways of patients reveals distinct immunological features of COVID-19 ARDS.


Subject(s)
COVID-19/genetics , RNA/genetics , Respiratory Distress Syndrome/genetics , Trachea/immunology , Adult , Aged , Aged, 80 and over , COVID-19/immunology , COVID-19/virology , Case-Control Studies , Cohort Studies , Critical Illness , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression Profiling , Humans , Male , Middle Aged , RNA/metabolism , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/virology , SARS-CoV-2/physiology , Sequence Analysis, RNA
7.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L466-L476, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34231389

ABSTRACT

Allergic asthma is a chronic airway inflammatory response to different triggers like inhaled allergens. Excessive ATP in fluids from patients with asthma is considered an inflammatory signal and an important autocrine/paracrine modulator of airway physiology. Here, we investigated the deleterious effect of increased extracellular ATP (eATP) concentration on the mucociliary clearance (MCC) effectiveness and determined the role of ATP releasing channels during airway inflammation in an ovalbumin (OVA)-sensitized mouse model. Our allergic mouse model exhibited high levels of eATP measured in the tracheal fluid with a luciferin-luciferase assay and reduced MCC velocity determined by microspheres tracking in the trachea ex vivo. Addition of ATP had a dual effect on MCC, where lower ATP concentration (µM) increased microspheres velocity, whereas higher concentration (mM) transiently stopped microspheres movement. Also, an augmented ethidium bromide uptake by the allergic tracheal airway epithelium suggests an increase in ATP release channel functionality during inflammatory conditions. The use of carbenoxolone, a nonspecific inhibitor of connexin and pannexin1 channels reduced the eATP concentration in the allergic mouse tracheal fluid and dye uptake by the airway epithelium, providing evidence that these ATP release channels are facilitating the net flux of ATP to the lumen during airway inflammation. However, only the specific inhibition of pannexin1 with 10Panx peptide significantly reduced eATP in bronchoalveolar lavage and decreased airway hyperresponsiveness in OVA-allergic mouse model. These data provide evidence that blocking eATP may be a pharmacological alternative to be explored in rescue therapy during episodes of airflow restriction in patients with asthma.


Subject(s)
Adenosine Triphosphate/immunology , Asthma/immunology , Carbenoxolone/pharmacology , Connexins/immunology , Nerve Tissue Proteins/immunology , Respiratory Mucosa/immunology , Trachea/immunology , Animals , Asthma/chemically induced , Asthma/drug therapy , Asthma/pathology , Connexins/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , Microspheres , Peptides/immunology , Peptides/pharmacology , Respiratory Mucosa/pathology , Trachea/pathology
8.
Cells ; 10(5)2021 05 19.
Article in English | MEDLINE | ID: mdl-34069395

ABSTRACT

Interleukin-10 plays a vital role in maintaining peripheral immunotolerance and favors a regulatory immune milieu through the suppression of T effector cells. Inflammation-induced microvascular loss has been associated with airway epithelial injury, which is a key pathological source of graft malfunctioning and subepithelial fibrosis in rejecting allografts. The regulatory immune phase maneuvers alloimmune inflammation through various regulatory modulators, and thereby promotes graft microvascular repair and suppresses the progression of fibrosis after transplantation. The present study was designed to investigate the therapeutic impact of IL-10 on immunotolerance, in particular, the reparative microenvironment, which negates airway epithelial injury, and fibrosis in a mouse model of airway graft rejection. Here, we depleted and reconstituted IL-10, and serially monitored the phase of immunotolerance, graft microvasculature, inflammatory cytokines, airway epithelium, and subepithelial collagen in rejecting airway transplants. We demonstrated that the IL-10 depletion suppresses FOXP3+ Tregs, tumor necrosis factor-inducible gene 6 protein (TSG-6), graft microvasculature, and establishes a pro-inflammatory phase, which augments airway epithelial injury and subepithelial collagen deposition while the IL-10 reconstitution facilitates FOXP3+ Tregs, TSG-6 deposition, graft microvasculature, and thereby favors airway epithelial repair and subepithelial collagen suppression. These findings establish a potential reparative modulation of IL-10-associated immunotolerance on microvascular, epithelial, and fibrotic remodeling, which could provide a vital therapeutic option to rescue rejecting transplants in clinical settings.


Subject(s)
Graft Rejection/metabolism , Interleukin-10/metabolism , Re-Epithelialization , Respiratory Mucosa/metabolism , Trachea/transplantation , Animals , Disease Models, Animal , Fibrosis , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival , Inflammation Mediators/metabolism , Interleukin-10/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , Trachea/immunology , Trachea/metabolism , Trachea/pathology , Transplantation Tolerance
9.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L102-L115, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34009050

ABSTRACT

Furin is a proprotein convertase that regulates the activation and the inactivation of multiple proteins including matrix metalloproteinases, integrins, and cytokines. It is a serine endoprotease that localizes to the plasma membrane and can be secreted into the extracellular space. The role of furin in regulating inflammation in isolated canine airway smooth muscle tissues was investigated. The treatment of airway tissues with recombinant furin (rFurin) inhibited the activation of Akt and eotaxin secretion induced by IL-13, and it prevented the IL-13-induced suppression of smooth muscle myosin heavy chain expression. rFurin promoted a differentiated phenotype by activating ß1-integrin proteins and stimulating the activation of the adhesome proteins vinculin and paxillin by talin. Activated paxillin induced the binding of Akt to ß-parvin IPP [integrin-linked kinase (ILK), PINCH, parvin] complexes, which inhibits Akt activation. Treatment of tissues with a furin inhibitor or the depletion of endogenous furin using shRNA resulted in Akt activation and inflammatory responses similar to those induced by IL-13. Furin inactivation or IL-13 caused talin cleavage and integrin inactivation, resulting in the inactivation of vinculin and paxillin. Paxillin inactivation resulted in the coupling of Akt to α-parvin IPP complexes, which catalyze Akt activation and an inflammatory response. The results demonstrate that furin inhibits inflammation in airway smooth muscle induced by IL-13 and that the anti-inflammatory effects of furin are mediated by activating integrin proteins and integrin-associated signaling complexes that regulate Akt-mediated pathways to the nucleus. Furin may have therapeutic potential for the treatment of inflammatory conditions of the lungs and airways.


Subject(s)
Furin/pharmacology , Inflammation/prevention & control , Integrins/metabolism , Interleukin-13/toxicity , Muscle, Smooth/drug effects , Trachea/drug effects , Animals , Dogs , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Integrins/genetics , Muscle, Smooth/immunology , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Signal Transduction , Trachea/immunology , Trachea/metabolism , Trachea/pathology
10.
PLoS Pathog ; 17(1): e1009195, 2021 01.
Article in English | MEDLINE | ID: mdl-33465158

ABSTRACT

SARS-CoV-2 emerged in late 2019 and resulted in the ongoing COVID-19 pandemic. Several animal models have been rapidly developed that recapitulate the asymptomatic to moderate disease spectrum. Now, there is a direct need for additional small animal models to study the pathogenesis of severe COVID-19 and for fast-tracked medical countermeasure development. Here, we show that transgenic mice expressing the human SARS-CoV-2 receptor (angiotensin-converting enzyme 2 [hACE2]) under a cytokeratin 18 promoter (K18) are susceptible to SARS-CoV-2 and that infection resulted in a dose-dependent lethal disease course. After inoculation with either 104 TCID50 or 105 TCID50, the SARS-CoV-2 infection resulted in rapid weight loss in both groups and uniform lethality in the 105 TCID50 group. High levels of viral RNA shedding were observed from the upper and lower respiratory tract and intermittent shedding was observed from the intestinal tract. Inoculation with SARS-CoV-2 resulted in upper and lower respiratory tract infection with high infectious virus titers in nasal turbinates, trachea and lungs. The observed interstitial pneumonia and pulmonary pathology, with SARS-CoV-2 replication evident in pneumocytes, were similar to that reported in severe cases of COVID-19. SARS-CoV-2 infection resulted in macrophage and lymphocyte infiltration in the lungs and upregulation of Th1 and proinflammatory cytokines/chemokines. Extrapulmonary replication of SARS-CoV-2 was observed in the cerebral cortex and hippocampus of several animals at 7 DPI but not at 3 DPI. The rapid inflammatory response and observed pathology bears resemblance to COVID-19. Additionally, we demonstrate that a mild disease course can be simulated by low dose infection with 102 TCID50 SARS-CoV-2, resulting in minimal clinical manifestation and near uniform survival. Taken together, these data support future application of this model to studies of pathogenesis and medical countermeasure development.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/genetics , COVID-19/pathology , Keratin-18/genetics , Angiotensin-Converting Enzyme 2/immunology , Animals , COVID-19/immunology , COVID-19/virology , Disease Models, Animal , Female , Humans , Keratin-18/immunology , Lung/immunology , Lung/pathology , Lymphocytes/immunology , Macrophages/immunology , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic , SARS-CoV-2/physiology , Trachea/immunology , Trachea/virology
11.
Laryngoscope ; 131(5): 967-974, 2021 05.
Article in English | MEDLINE | ID: mdl-32557663

ABSTRACT

OBJECTIVES: Laryngotracheal stenosis (LTS) is a fibrotic condition of the upper airway. Recent evidence suggests dysregulated host immunity plays a role in LTS development and progression. The programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) axis, targeted by paradigm-shifting immunotherapies for cancer treatment, has also recently been implicated in the pathogenesis of fibrotic pulmonary disease. However, a role for the PD-1/PD-L1 axis in the proximal airway fibrosis seen in LTS patients has not been explored. STUDY DESIGN: Controlled ex vivo study. METHODS: Expression of PD-1, PD-L1, CD4, and CD8 were evaluated using immunohistochemical staining of cricotracheal resection specimens from postintubation iatrogenic laryngotracheal stenosis (iLTS), idiopathic subglottic stenosis (iSGS) patients, and normal controls derived from rapid autopsy (n = 8 per group). Fibroblasts derived from iLTS scar were also treated with transforming growth factor beta 1 (TGFß1) and analyzed for PD-L1 expression by quantitative real-time polymerase chain reaction (n = 6). RESULTS: iLTS specimens exhibited increased expression of PD-1, PD-L1, and CD4 (all P < .0167) compared to controls, whereas iSGS specimens exhibited increased expression of PD-1 and CD4 (P < .0167) compared to controls. PD-1, PD-L1, and CD4 showed periepithelial patterns of expression in both disease cohorts. TGFß1 treatment of iLTS fibroblasts increased expression of PD-L1 (the cognate ligand for PD-1). CONCLUSION: Expression of both PD-1 and its ligand PD-L1 are significantly greater in patients with iLTS compared to controls, and PD-1 expression is also elevated in patients with iSGS. Given published evidence implicating the PD-1/PD-L1 axis in pulmonary fibrosis, this suggests a possible role for checkpoint inhibitors targeting the PD-1/PD-L1 axis for the treatment of LTS. LEVEL OF EVIDENCE: N/A Laryngoscope, 131:967-974, 2021.


Subject(s)
B7-H1 Antigen/metabolism , Laryngostenosis/immunology , Programmed Cell Death 1 Receptor/metabolism , Tracheal Stenosis/immunology , B7-H1 Antigen/analysis , Biopsy , Case-Control Studies , Cells, Cultured , Cricoid Cartilage/immunology , Cricoid Cartilage/pathology , Cricoid Cartilage/surgery , Female , Fibroblasts , Fibrosis , Humans , Immunohistochemistry , Laryngostenosis/pathology , Laryngostenosis/surgery , Male , Middle Aged , Primary Cell Culture , Programmed Cell Death 1 Receptor/analysis , Trachea/immunology , Trachea/pathology , Trachea/surgery , Tracheal Stenosis/pathology , Tracheal Stenosis/surgery , Tracheostomy
12.
Laryngoscope ; 131(2): E346-E353, 2021 02.
Article in English | MEDLINE | ID: mdl-33051870

ABSTRACT

OBJECTIVE: Macrophages exhibit distinct phenotypes and are dysregulated in a model of iatrogenic laryngotracheal stenosis (iLTS). Increased populations of alternatively activated or M2 macrophages have been demonstrated. However, the role of these macrophages is unknown. The aims of this study are: 1) define the macrophage population in iLTS in the context of classically activated or M1 and M2 macrophage phenotypes, and 2) characterize the effect of monocyte-derived M1 and M2 macrophages on normal airway and LTS-derived fibroblasts (FBs) in vitro. STUDY DESIGN: Comparative analysis; in vitro controlled study. METHODS: Immunohistochemical analysis of human iLTS and control specimens was performed to define the macrophage population. In vitro, M1, and M2 macrophages were polarized using M-CSF + Interferon-gamma and lipopolysaccharide or Interleukin-4, respectively. FBs isolated from laryngotracheal scar (LTS-FBs) and normal tracheal airway (NA-FBs) in eight patients with iLTS were cocultured with polarized macrophages. Fibrosis gene expression, soluble collagen production, and proliferation were assessed. RESULTS: Immunohistochemical analysis revealed increased CD11b + cells (macrophage marker) in laryngotracheal scar specimens (18.3% vs. 8.5%, P = .03) and predominant CD206 (M2) costaining versus CD86 (M1) (51.5% vs. 9.8%, n = 10, P = .001). In vitro, NA-FBs cultured with M2 macrophages demonstrated a 2.41-fold increase in collagen-1 expression (P = .05, n = 8) and an increase in soluble collagen (9.98 vs. 8.875, mean difference: 1.11 95%, confidence interval 0.024-2.192, n = 8, P = .015). CONCLUSION: Increased populations of CD11b cells are present in iLTS specimens and are predominantly CD206+, indicating an M2 phenotype. In vitro, M2 macrophages promoted collagen expression in airway FBs. Targeting macrophages may represent a therapeutic strategy for attenuating fibrosis in iLTS. LEVEL OF EVIDENCE: NA Laryngoscope, 131:E346-E353, 2021.


Subject(s)
Fibroblasts/pathology , Laryngostenosis/immunology , Macrophages/immunology , Tracheal Stenosis/immunology , Adult , CD11b Antigen/metabolism , Cell Communication/immunology , Cell Line , Collagen/metabolism , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Fibrosis , Humans , Iatrogenic Disease , Intubation, Intratracheal/adverse effects , Laryngostenosis/etiology , Laryngostenosis/pathology , Larynx/cytology , Larynx/immunology , Larynx/pathology , Macrophages/metabolism , Male , Membrane Glycoproteins/metabolism , Primary Cell Culture , Receptors, Immunologic/metabolism , Trachea/cytology , Trachea/immunology , Trachea/pathology , Tracheal Stenosis/etiology , Tracheal Stenosis/pathology
13.
Ann Otol Rhinol Laryngol ; 130(5): 441-449, 2021 May.
Article in English | MEDLINE | ID: mdl-32911959

ABSTRACT

OBJECTIVES: Reconstruction of long segmental tracheal defects is difficult because no ideal tracheal substitutes are currently available. Tracheal allotransplantation maintains cartilage and epithelium viability but requires immunosuppression because of epithelial immunogenicity. We aimed to obtain an epithelium-decellularized allograft that maintains cartilage viability and to evaluate long-term outcomes of such allografts implanted on dog backs without immunosuppressants. METHODS: Twenty-five tracheas harvested from mongrel dogs were used to explore the period of epithelium decellularization by combined use of 1% sodium dodecyl sulfate and an organ preservation solution and to assess the chondrocyte viability and immunogenicity of the tracheas after decellularization. Sixteen epithelium-decellularized tracheal allografts and 10 fresh tracheal segments (6 cm long) were implanted in 26 beagles for durations of 10 days and 1, 3, 6, and 12 months. Macroscopic and microscopic examinations were used to evaluate the morphology, viability, and immune rejection of the allografts. Safranin-O staining was used to detect glycosaminoglycans. RESULTS: The epithelium disappeared after 24 hours of decellularization. At 72 hours, almost no nuclei remained in the mucosa, while the mean survival rate of chondrocytes was 88.1%. Histological analysis demonstrated that the allograft retained intact tracheal rings and viable cartilage after heterotopic implantation for 1 year, with no immunological rejection. There were no significant differences in the glycosaminoglycan contents among the implanted epithelium-decellularized allografts. CONCLUSIONS: Epithelium-decellularized tracheal allografts with chondrocyte viability can be achieved by combined use of a detergent and organ preservation solution, which showed satisfactory cartilage viability and structural integrity after long-term heterotopic transplantation. Further studies on orthotopic transplantation are needed to assess the feasibility of allografts in reconstructing long segmental tracheal defects.


Subject(s)
Cartilage , Cell Survival , Chondrocytes/physiology , Graft Rejection/prevention & control , Plastic Surgery Procedures/methods , Trachea , Transplantation, Homologous/methods , Animals , Cartilage/immunology , Cartilage/transplantation , Disease Models, Animal , Dogs , Organ Preservation/methods , Trachea/immunology , Trachea/transplantation , Transplantation Immunology , Treatment Outcome
15.
J Allergy Clin Immunol ; 147(4): 1442-1452, 2021 04.
Article in English | MEDLINE | ID: mdl-32791164

ABSTRACT

BACKGROUND: The immunoinhibitory receptor Siglec-8 on the surface of human eosinophils and mast cells binds to sialic acid-containing ligands in the local milieu, resulting in eosinophil apoptosis, inhibition of mast cell degranulation, and suppression of inflammation. Siglec-8 ligands were found on postmortem human trachea and bronchi and on upper airways in 2 compartments, cartilage and submucosal glands, but they were surprisingly absent from the epithelium. We hypothesized that Siglec-8 ligands in submucosal glands and ducts are normally transported to the airway mucus layer, which is lost during tissue preparation. OBJECTIVE: Our aim was to identify the major Siglec-8 sialoglycan ligand on the mucus layer of human airways. METHODS: Human upper airway mucus layer proteins were recovered during presurgical nasal lavage of patients at a sinus clinic. Proteins were resolved by gel electrophoresis and blotted, and Siglec-8 ligands detected. Ligands were purified by size exclusion and affinity chromatography, identified by proteomic mass spectrometry, and validated by electrophoretic and histochemical colocalization. The affinity of Siglec-8 binding to purified human airway ligand was determined by inhibition of glycan binding. RESULTS: A Siglec-8-ligand with a molecular weight of approximately 1000 kDa was found in all patient nasal lavage samples. Purification and identification revealed deleted in malignant brain tumors 1 (DMBT1) (also known by the aliases GP340 and SALSA), a large glycoprotein with multiple O-glycosylation repeats. Immunoblotting, immunohistochemistry, and enzyme treatments confirmed that Siglec-8 ligand on the human airway mucus layer is an isoform of DMBT1 carrying O-linked sialylated keratan sulfate chains (DMBT1S8). Quantitative inhibition revealed that DMBT1S8 has picomolar affinity for Siglec-8. CONCLUSION: A distinct DMBT1 isoform, DMBT1S8, is the major high-avidity ligand for Siglec-8 on human airways.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Calcium-Binding Proteins/immunology , DNA-Binding Proteins/immunology , Lectins/immunology , Tumor Suppressor Proteins/immunology , Bronchi/immunology , Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/chemistry , Eosinophils/immunology , Humans , Ligands , Mast Cells/immunology , Nasal Lavage Fluid/immunology , Proteoglycans/immunology , Trachea/immunology , Tumor Suppressor Proteins/chemistry
16.
Vet Microbiol ; 251: 108883, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33069036

ABSTRACT

Mycoplasma gallisepticum (MG) is an important pathogen of poultry worldwide, causing chronic respiratory disease in chickens and turkeys. MG ts-304 is a GapA positive clone recovered from Vaxsafe MG (strain ts-11) that has been shown to be safe in chickens when delivered by the eye drop route to 3-week-old specific-pathogen-free chickens and to confer protection against challenge at 4 weeks after vaccination, as measured by tracheal mucosal thickness and air sac lesion scores. In this study, specific pathogen-free chickens (SPF) were vaccinated with a single dose of the MG ts-304 vaccine (106.0 colour changing units) at 3 weeks of age and experimentally challenged by aerosol with the virulent M. gallisepticum strain Ap3AS at 40, 48 and 57 weeks after vaccination. There were no significant differences in tracheal mucosal thickness 2 weeks after challenge between chickens challenged at the three time points, or between the vaccinated birds after challenge and unvaccinated/unchallenged control birds. Thus there was clear evidence that the immunity conferred by vaccination with the MG ts-304 vaccine resulted in significant protection against tracheitis in chickens that extended to, but was highly likely to exceed, 57 weeks after vaccination and that similar long term protective immunity could be expected to be conferred by a vaccine dose lower than that used in this study.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Vaccines/immunology , Mycoplasma Infections/prevention & control , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/immunology , Poultry Diseases/prevention & control , Vaccination/veterinary , Air Sacs/microbiology , Air Sacs/pathology , Animals , Bacterial Vaccines/administration & dosage , Chickens/immunology , Mucous Membrane/immunology , Mycoplasma Infections/immunology , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/immunology , Poultry Diseases/microbiology , Specific Pathogen-Free Organisms , Trachea/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
17.
Infect Immun ; 88(11)2020 10 19.
Article in English | MEDLINE | ID: mdl-32868342

ABSTRACT

Porcine circovirus type 2 (PCV2) and Streptococcus suis serotype 2 (SS2) clinical coinfection cases have been frequently detected. The respiratory epithelium plays a crucial role in host defense against a variety of inhaled pathogens. Reactive oxygen species (ROS) are involved in killing of bacteria and host immune response. The aim of this study is to assess whether PCV2 and SS2 coinfection in swine tracheal epithelial cells (STEC) affects ROS production and investigate the roles of ROS in bacterial survival and the inflammatory response. Compared to SS2 infection, PCV2/SS2 coinfection inhibited the activity of NADPH oxidase, resulting in lower ROS levels. Bacterial intracellular survival experiments showed that coinfection with PCV2 and SS2 enhanced SS2 survival in STEC. Pretreatment of STEC with N-acetylcysteine (NAC) also helps SS2 intracellular survival, indicating that PCV2/SS2 coinfection enhances the survival of SS2 in STEC through a decrease in ROS production. In addition, compared to SS2-infected STEC, PCV2/SS2 coinfection and pretreatment of STEC with NAC prior to SS2 infection both downregulated the expression of the inflammatory cytokines interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and IL-1ß. Further research found that activation of p38/MAPK promoted the expression of inflammatory cytokines in SS2-infected STEC; however, PCV2/SS2 coinfection or NAC pretreatment of STEC inhibited p38 phosphorylation, suggesting that coinfection of STEC with PCV2 and SS2 weakens the inflammatory response to SS2 infection through reduced ROS production. Collectively, coinfection of STEC with PCV2 and SS2 enhances the intracellular survival of SS2 and weakens the inflammatory response through decreased ROS production, which might exacerbate SS2 infection in the host.


Subject(s)
Circoviridae Infections/virology , Coinfection/microbiology , Reactive Oxygen Species/metabolism , Respiratory Mucosa/microbiology , Streptococcal Infections/microbiology , Swine Diseases/microbiology , Animals , Circoviridae Infections/immunology , Circoviridae Infections/metabolism , Circovirus/immunology , Circovirus/metabolism , Coinfection/immunology , Coinfection/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Streptococcus suis/immunology , Streptococcus suis/metabolism , Swine , Swine Diseases/immunology , Swine Diseases/metabolism , Trachea/immunology , Trachea/metabolism , Trachea/microbiology
18.
Metallomics ; 12(11): 1679-1692, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32910126

ABSTRACT

Dietary selenium (Se) deficiency can induce multifarious immune injury in tissues, accompanied by inflammation and a decreased expression of selenoproteins. The results of previous studies indicated that these issues are associated with Se-mediated microRNAs involved in immune regulation, although the specific mechanisms associated with these interactions have not been reported in the trachea of chickens. To explore the effects of Se deficiency in the trachea of chickens and the role of miR-196-5p, we established correlational models of tracheal injury in chickens. One hundred broilers were divided into four groups, including a control group (C group), a Se deficient group (L group), a lipopolysaccharide (LPS)-induced control group (C + LPS group) and a LPS-induced Se deficient group (L + LPS group). Light microscopy observations indicated that the infiltration of inflammatory cells was the major histopathological change caused by Se deficiency. Furthermore, ultrastructural observation of the tracheal epithelium and ciliary showed typical inflammatory signs owing to Se deficiency. We determined the targeting relationship between miR-196-5p and NFκBIA by bioinformatics analysis. In the case of Se deficiency, the changes were detected as follows: 19 selenoproteins showed different degrees of decrease (p < 0.05). Significant inhibition of both antimicrobial peptides and immunoglobulin production were observed (p < 0.05). IκB-α (NFκBIA) expression degraded with the increasing miR-196-5p (p < 0.05), and the NF-κB pathway was activated. Thereafter, we can see a significant increase in the mRNA levels of inflammatory cytokines-related genes (tumor necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, prostaglandin E (PTGE), interleukin (IL)-1ß, IL-6) and protein expression of NF-κB/iNOS pathway-related genes (NF-κB, iNOS, TNF-α, COX-2) (p < 0.05). The release of IL-2, interferon (IFN)-γ inhibited (p < 0.05) and the secretion of IL-4, IL-6 increased, suggesting the imbalance of Th1/Th2 (Th, helper T cell) cytokines. Compared to the control, the mRNA and protein expression levels of the anti-inflammatory system components with antioxidant activity (PPAR-γ/HO-1) were in an inhibitory state (p < 0.05). Antioxidases (SOD, CAT, GSH-Px) activities were suppressed. The activities of the peroxide markers (MDA, H2O2) were enhanced (p < 0.05). In addition, Se deficiency had a positive effect on the pathological changes of inflammation and the exceptional immunity in LPS-treated groups (p < 0.05). The results confirmed the relationship between miR-196-5p and NFκBIA in chickens, revealing that Se deficiency causes respiratory mucosal immune dysfunction via the miR-196-5p-NFκBIA axis, oxidative stress and inflammation. Moreover, Se deficiency exacerbates the inflammatory damage stimulated by LPS. Our work provides a theoretical basis for the prevention of tracheal injury owing to Se deficiency and can be used as a reference for comparative medicine. Furthermore, the targeted regulation of miR-196-5p and NFκBIA may contribute to the protection of the tracheal mucosa in chickens.


Subject(s)
Chickens/genetics , Chickens/immunology , MicroRNAs/metabolism , NF-KappaB Inhibitor alpha/metabolism , Selenium/deficiency , Trachea/immunology , Trachea/pathology , Animals , Antimicrobial Cationic Peptides/biosynthesis , Base Sequence , Cytokines/metabolism , Gene Expression Regulation , Heme Oxygenase-1/metabolism , Immunoglobulins/metabolism , Inflammation/genetics , Inflammation/pathology , MicroRNAs/genetics , Oxidative Stress/genetics , PPAR gamma/metabolism , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Trachea/ultrastructure
19.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-32926098

ABSTRACT

Severe COVID-19 patients develop acute respiratory distress syndrome that may progress to cytokine storm syndrome, organ dysfunction, and death. Considering that neutrophil extracellular traps (NETs) have been described as important mediators of tissue damage in inflammatory diseases, we investigated whether NETs would be involved in COVID-19 pathophysiology. A cohort of 32 hospitalized patients with a confirmed diagnosis of COVID-19 and healthy controls were enrolled. The concentration of NETs was augmented in plasma, tracheal aspirate, and lung autopsies tissues from COVID-19 patients, and their neutrophils released higher levels of NETs. Notably, we found that viable SARS-CoV-2 can directly induce the release of NETs by healthy neutrophils. Mechanistically, NETs triggered by SARS-CoV-2 depend on angiotensin-converting enzyme 2, serine protease, virus replication, and PAD-4. Finally, NETs released by SARS-CoV-2-activated neutrophils promote lung epithelial cell death in vitro. These results unravel a possible detrimental role of NETs in the pathophysiology of COVID-19. Therefore, the inhibition of NETs represents a potential therapeutic target for COVID-19.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Extracellular Traps/physiology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , A549 Cells , Adult , Angiotensin-Converting Enzyme 2 , COVID-19 , Cell Death , Coronavirus Infections/blood , Coronavirus Infections/pathology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , HeLa Cells , Humans , Male , Neutrophil Activation , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/blood , Pneumonia, Viral/pathology , SARS-CoV-2 , Serine Proteases/metabolism , Suction , Trachea/immunology
20.
Can J Physiol Pharmacol ; 98(11): 810-817, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32687729

ABSTRACT

The purpose of this study was to explore the potential contracting effect of leptin on isolated guinea pig tracheal smooth muscle (TSM), the possible mechanism, and the impact of epithelium denudation or allergen sensitization, respectively. An in vitro experiment investigated the effect of leptin at a concentration of 250-1000 nmol/L on isolated guinea pig TSM with an intact or denuded epithelium. Ovalbumin and IgE were used to test the impact of active and passive sensitization. The isolated TSM strips were incubated in Krebs solution and aerated with carbogen (95% O2 and 5% CO2) via an automated tissue organ bath system (n = 4 for each group). Isometric contractions were recorded digitally using iox2 data acquisition software. The possible mechanism of leptin-induced TSM contraction was examined by preincubation with leptin receptor (Ob-R) antagonist. Leptin had significant concentration-dependent contraction effects on guinea pig TSM (p < 0.05). Epithelium denuding and active or passive sensitization significantly increased the potency of the leptin. Preincubation with a leptin receptor (Ob-R) antagonist significantly reduced the contraction effects, suggesting an Ob-R-mediated mechanism. Leptin had a contracting effect on airway smooth muscles potentiated by either epithelium denuding or sensitization, and the Ob-R mechanism was a possible effect mediator.


Subject(s)
Asthma/physiopathology , Leptin/metabolism , Muscle Contraction/immunology , Muscle, Smooth/physiopathology , Trachea/physiopathology , Animals , Asthma/immunology , Disease Models, Animal , Guinea Pigs , Humans , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/metabolism , Trachea/drug effects , Trachea/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...