Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Stem Cells ; 41(8): 809-820, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37468433

ABSTRACT

Single-cell RNA sequencing is a valuable tool for dissecting cellular heterogeneity in complex systems. However, it is still challenging to estimate the proliferation and differentiation potentials of subpopulations within dormant tissue stem cells. Here, we established a new single-cell analysis method for profiling the organoid-forming capacity and differentiation potential of tissue stem cells to disclose stem cell subpopulations by integrating single-cell morphometrics, organoid-forming assay, and RNA sequencing, a method named scMORN. To explore lung epithelial stem cells, we initially developed feeder-free culture system, which could expand all major lung stem cells, including basal, club, and alveolar type 2 (AT2) cells, and found that club cells contained a subpopulation, which showed better survival rate and high proliferation capacity and could differentiate into alveolar cells. Using the scMORN method, we discovered a club cell subpopulation named Muc5b+ and large club (ML-club) cells that efficiently formed organoids than other club or AT2 cells in our feeder-free organoid culture and differentiated into alveolar cells in vitro. Single-cell transcriptome profiling and immunohistochemical analysis revealed that ML-club cells localized at the intrapulmonary proximal airway and distinct from known subpopulations of club cells such as BASCs. Furthermore, we identified CD14 as a cell surface antigen of ML-club cells and showed that purified CD14+ club cells engrafted into injured mouse lungs had better engraftment rate and expansion than other major lung stem cells, reflecting the observations in organoid culture systems. The scMORN method could be adapted to different stem cell tissues to discover useful stem-cell subpopulations.


Subject(s)
Lung , Transcriptome , Animals , Mice , Transcriptome/genetics , Stem Cells/metabolism , Organoids/metabolism , Gene Expression Profiling , Cell Differentiation
2.
Am J Respir Cell Mol Biol ; 69(3): 255-265, 2023 09.
Article in English | MEDLINE | ID: mdl-37315312

ABSTRACT

Targeted delivery of transgenes to tissue-resident stem cells and related niches offers avenues for interrogating pathways and editing endogenous alleles for therapeutic interventions. Here, we survey multiple adeno-associated virus (AAV) serotypes, administered via intranasal and retroorbital routes in mice, to target lung alveolar stem cell niches. We found that AAV5, AAV4, and AAV8 efficiently and preferentially transduce alveolar type-2 stem cells (AT2s), endothelial cells, and PDGFRA+ fibroblasts, respectively. Notably, some AAVs show different cell tropisms depending on the route of administration. Proof-of-concept experiments reveal the versatility of AAV5-mediated transgenesis for AT2-lineage labeling, clonal cell tracing after cell ablation, and conditional gene inactivation in both postnatal and adult mouse lungs in vivo. AAV6, but not AAV5, efficiently transduces both mouse and human AT2s in alveolar organoid cultures. Furthermore, AAV5 and AAV6 can be used to deliver guide RNAs and transgene cassettes for homologous recombination in vivo and ex vivo, respectively. Using this system coupled with clonal derivation of AT2 organoids, we demonstrate efficient and simultaneous editing of multiple loci, including targeted insertion of a payload cassette in AT2s. Taken together, our studies highlight the powerful utility of AAVs for interrogating alveolar stem cells and other specific cell types both in vivo and ex vivo.


Subject(s)
Dependovirus , Endothelial Cells , Mice , Animals , Humans , Dependovirus/genetics , Transduction, Genetic , Genetic Vectors , Gene Transfer Techniques , Stem Cells
3.
Proc Natl Acad Sci U S A ; 115(28): E6622-E6629, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29941581

ABSTRACT

The pathophysiological changes that occur in lungs infected with influenza viruses are poorly understood. Here we established an in vivo imaging system that combines two-photon excitation microscopy and fluorescent influenza viruses of different pathogenicity. This approach allowed us to monitor and correlate several parameters and physiological changes including the spread of infection, pulmonary permeability, pulmonary perfusion speed, number of recruited neutrophils in infected lungs, and neutrophil motion in the lungs of live mice. Several physiological changes were larger and occurred earlier in mice infected with a highly pathogenic H5N1 influenza virus compared with those infected with a mouse-adapted human strain. These findings demonstrate the potential of our in vivo imaging system to provide novel information about the pathophysiological consequences of virus infections.


Subject(s)
Influenza A Virus, H5N1 Subtype/metabolism , Lung , Microscopy, Fluorescence, Multiphoton , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Animals , Influenza A Virus, H5N1 Subtype/genetics , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Orthomyxoviridae Infections/genetics
4.
J Infect Dis ; 222(7): 1155-1164, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32433769

ABSTRACT

The avian influenza A(H7N9) virus has caused high mortality rates in humans, especially in the elderly; however, little is known about the mechanistic basis for this. In the current study, we used nonhuman primates to evaluate the effect of aging on the pathogenicity of A(H7N9) virus. We observed that A(H7N9) virus infection of aged animals (defined as age 20-26 years) caused more severe symptoms than infection of young animals (defined as age 2-3 years). In aged animals, lung inflammation was weak and virus infection was sustained. Although cytokine and chemokine expression in the lungs of most aged animals was lower than that in the lungs of young animals, 1 aged animal showed severe symptoms and dysregulated proinflammatory cytokine and chemokine production. These results suggest that attenuated or dysregulated immune responses in aged animals are responsible for the severe symptoms observed among elderly patients infected with A(H7N9) virus.


Subject(s)
Aging , Influenza A Virus, H7N9 Subtype , Lung/pathology , Orthomyxoviridae Infections/virology , Animals , Cytokines/immunology , Disease Models, Animal , Female , Lung/immunology , Lung/virology , Macaca fascicularis , Orthomyxoviridae Infections/immunology , Virus Replication
5.
Development ; 144(6): 986-997, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28292845

ABSTRACT

Lungs are composed of a system of highly branched tubes that bring air into the alveoli, where gas exchange takes place. The proximal and distal regions of the lung contain epithelial cells specialized for different functions: basal, secretory and ciliated cells in the conducting airways and type II and type I cells lining the alveoli. Basal, secretory and type II cells can be grown in three-dimensional culture, with or without supporting stromal cells, and under these conditions they give rise to self-organizing structures known as organoids. This Review summarizes the different methods for generating organoids from cells isolated from human and mouse lungs, and compares their final structure and cellular composition with that of the airways or alveoli of the adult lung. We also discuss the potential and limitations of organoids for addressing outstanding questions in lung biology and for developing new drugs for disorders such as cystic fibrosis and asthma.


Subject(s)
Lung/cytology , Organoids/cytology , Alveolar Epithelial Cells/cytology , Animals , Humans , Stem Cells/cytology
6.
Nature ; 501(7468): 551-5, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23842494

ABSTRACT

Avian influenza A viruses rarely infect humans; however, when human infection and subsequent human-to-human transmission occurs, worldwide outbreaks (pandemics) can result. The recent sporadic infections of humans in China with a previously unrecognized avian influenza A virus of the H7N9 subtype (A(H7N9)) have caused concern owing to the appreciable case fatality rate associated with these infections (more than 25%), potential instances of human-to-human transmission, and the lack of pre-existing immunity among humans to viruses of this subtype. Here we characterize two early human A(H7N9) isolates, A/Anhui/1/2013 (H7N9) and A/Shanghai/1/2013 (H7N9); hereafter referred to as Anhui/1 and Shanghai/1, respectively. In mice, Anhui/1 and Shanghai/1 were more pathogenic than a control avian H7N9 virus (A/duck/Gunma/466/2011 (H7N9); Dk/GM466) and a representative pandemic 2009 H1N1 virus (A/California/4/2009 (H1N1pdm09); CA04). Anhui/1, Shanghai/1 and Dk/GM466 replicated well in the nasal turbinates of ferrets. In nonhuman primates, Anhui/1 and Dk/GM466 replicated efficiently in the upper and lower respiratory tracts, whereas the replicative ability of conventional human influenza viruses is typically restricted to the upper respiratory tract of infected primates. By contrast, Anhui/1 did not replicate well in miniature pigs after intranasal inoculation. Critically, Anhui/1 transmitted through respiratory droplets in one of three pairs of ferrets. Glycan arrays showed that Anhui/1, Shanghai/1 and A/Hangzhou/1/2013 (H7N9) (a third human A(H7N9) virus tested in this assay) bind to human virus-type receptors, a property that may be critical for virus transmissibility in ferrets. Anhui/1 was found to be less sensitive in mice to neuraminidase inhibitors than a pandemic H1N1 2009 virus, although both viruses were equally susceptible to an experimental antiviral polymerase inhibitor. The robust replicative ability in mice, ferrets and nonhuman primates and the limited transmissibility in ferrets of Anhui/1 suggest that A(H7N9) viruses have pandemic potential.


Subject(s)
Influenza A virus , Influenza, Human/virology , Orthomyxoviridae Infections/virology , Virus Replication , Animals , Antiviral Agents/pharmacology , Cells, Cultured , Chickens/virology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Dogs , Enzyme Inhibitors/pharmacology , Female , Ferrets/virology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A virus/chemistry , Influenza A virus/drug effects , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Influenza, Human/drug therapy , Macaca fascicularis/virology , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Monkey Diseases/pathology , Monkey Diseases/virology , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/transmission , Quail/virology , Swine/virology , Swine, Miniature/virology , Virus Replication/drug effects
7.
Nature ; 486(7403): 420-8, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22722205

ABSTRACT

Highly pathogenic avian H5N1 influenza A viruses occasionally infect humans, but currently do not transmit efficiently among humans. The viral haemagglutinin (HA) protein is a known host-range determinant as it mediates virus binding to host-specific cellular receptors. Here we assess the molecular changes in HA that would allow a virus possessing subtype H5 HA to be transmissible among mammals. We identified a reassortant H5 HA/H1N1 virus-comprising H5 HA (from an H5N1 virus) with four mutations and the remaining seven gene segments from a 2009 pandemic H1N1 virus-that was capable of droplet transmission in a ferret model. The transmissible H5 reassortant virus preferentially recognized human-type receptors, replicated efficiently in ferrets, caused lung lesions and weight loss, but was not highly pathogenic and did not cause mortality. These results indicate that H5 HA can convert to an HA that supports efficient viral transmission in mammals; however, we do not know whether the four mutations in the H5 HA identified here would render a wholly avian H5N1 virus transmissible. The genetic origin of the remaining seven viral gene segments may also critically contribute to transmissibility in mammals. Nevertheless, as H5N1 viruses continue to evolve and infect humans, receptor-binding variants of H5N1 viruses with pandemic potential, including avian-human reassortant viruses as tested here, may emerge. Our findings emphasize the need to prepare for potential pandemics caused by influenza viruses possessing H5 HA, and will help individuals conducting surveillance in regions with circulating H5N1 viruses to recognize key residues that predict the pandemic potential of isolates, which will inform the development, production and distribution of effective countermeasures.


Subject(s)
Adaptation, Physiological/genetics , Ferrets/virology , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Reassortant Viruses/pathogenicity , Respiratory System/virology , Animals , Bioterrorism/prevention & control , Birds/virology , Body Fluids/virology , Cell Line , Dogs , Evolution, Molecular , Female , HEK293 Cells , HeLa Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hot Temperature , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/physiology , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/prevention & control , Influenza, Human/transmission , Influenza, Human/virology , Molecular Epidemiology/methods , Pandemics , Population Surveillance/methods , Protein Stability , Reassortant Viruses/genetics , Reassortant Viruses/isolation & purification , Reassortant Viruses/physiology , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Respiratory System/anatomy & histology , Security Measures , Zoonoses/transmission , Zoonoses/virology
8.
PLoS Pathog ; 11(6): e1004856, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26046528

ABSTRACT

Influenza viruses present major challenges to public health, evident by the 2009 influenza pandemic. Highly pathogenic influenza virus infections generally coincide with early, high levels of inflammatory cytokines that some studies have suggested may be regulated in a strain-dependent manner. However, a comprehensive characterization of the complex dynamics of the inflammatory response induced by virulent influenza strains is lacking. Here, we applied gene co-expression and nonlinear regression analysis to time-course, microarray data developed from influenza-infected mouse lung to create mathematical models of the host inflammatory response. We found that the dynamics of inflammation-associated gene expression are regulated by an ultrasensitive-like mechanism in which low levels of virus induce minimal gene expression but expression is strongly induced once a threshold virus titer is exceeded. Cytokine assays confirmed that the production of several key inflammatory cytokines, such as interleukin 6 and monocyte chemotactic protein 1, exhibit ultrasensitive behavior. A systematic exploration of the pathways regulating the inflammatory-associated gene response suggests that the molecular origins of this ultrasensitive response mechanism lie within the branch of the Toll-like receptor pathway that regulates STAT1 phosphorylation. This study provides the first evidence of an ultrasensitive mechanism regulating influenza virus-induced inflammation in whole lungs and provides insight into how different virus strains can induce distinct temporal inflammation response profiles. The approach developed here should facilitate the construction of gene regulatory models of other infectious diseases.


Subject(s)
Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/immunology , Animals , Blotting, Western , Female , Flow Cytometry , Inflammation/genetics , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/genetics , Transcriptome , Virulence
9.
J Virol ; 89(22): 11337-46, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26339046

ABSTRACT

UNLABELLED: We previously reported that an H5N1 virus carrying the Venus reporter gene, which was inserted into the NS gene segment from the A/Puerto Rico/8/1934(H1N1) virus (Venus-H5N1 virus), became more lethal to mice, and the reporter gene was stably maintained after mouse adaptation compared with the wild-type Venus-H5N1 (WT-Venus-H5N1) virus. However, the basis for this difference in virulence and Venus stability was unclear. Here, we investigated the molecular determinants behind this virulence and reporter stability by comparing WT-Venus-H5N1 virus with a mouse-adapted Venus-H5N1 (MA-Venus-H5N1) virus. To determine the genetic basis for these differences, we used reverse genetics to generate a series of reassortants of these two viruses. We found that reassortants with PB2 from MA-Venus-H5N1 (MA-PB2), MA-PA, or MA-NS expressed Venus more stably than did WT-Venus-H5N1 virus. We also found that a single mutation in PB2 (V25A) or in PA (R443K) increased the virulence of the WT-Venus-H5N1 virus in mice and that the presence of both of these mutations substantially enhanced the pathogenicity of the virus. Our results suggest roles for PB2 and PA in the stable maintenance of a foreign protein as an NS1 fusion protein in influenza A virus. IMPORTANCE: The ability to visualize influenza viruses has far-reaching benefits in influenza virus research. Previously, we reported that an H5N1 virus bearing the Venus reporter gene became more pathogenic to mice and that its reporter gene was more highly expressed and more stably maintained after mouse adaptation. Here, we investigated the molecular determinants behind this enhanced virulence and reporter stability. We found that mutations in PB2 (V25A) and PA (R443K) play crucial roles in the stable maintenance of a foreign protein as an NS1 fusion protein in influenza A virus and in the virulence of influenza virus in mice. Our findings further our knowledge of the pathogenicity of influenza virus in mammals and will help advance influenza virus-related live-imaging studies in vitro and in vivo.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Reassortant Viruses/pathogenicity , Viral Nonstructural Proteins/genetics , Virus Replication/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Dogs , Female , Genes, Reporter/genetics , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Reassortant Viruses/genetics , Sequence Analysis, RNA , Virulence/genetics
10.
J Virol ; 88(22): 13410-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25210171

ABSTRACT

UNLABELLED: Streptococcus pneumoniae is a major causative pathogen in community-acquired pneumonia; together with influenza virus, it represents an important public health burden. Although vaccination is the most effective prophylaxis against these infectious agents, no single vaccine simultaneously provides protective immunity against both S. pneumoniae and influenza virus. Previously, we demonstrated that several replication-incompetent influenza viruses efficiently elicit IgG in serum and IgA in the upper and lower respiratory tracts. Here, we generated a replication-incompetent hemagglutinin knockout (HA-KO) influenza virus possessing the sequence for the antigenic region of pneumococcal surface protein A (PspA). Although this virus (HA-KO/PspA virus) could replicate only in an HA-expressing cell line, it infected wild-type cells and expressed both viral proteins and PspA. PspA- and influenza virus-specific antibodies were detected in nasal wash and bronchoalveolar lavage fluids and in sera from mice intranasally inoculated with HA-KO/PspA virus, and mice inoculated with HA-KO/PspA virus were completely protected from lethal challenge with either S. pneumoniae or influenza virus. Further, bacterial colonization of the nasopharynx was prevented in mice immunized with HA-KO/PspA virus. These results indicate that HA-KO/PspA virus is a promising bivalent vaccine candidate that simultaneously confers protective immunity against both S. pneumoniae and influenza virus. We believe that this strategy offers a platform for the development of bivalent vaccines, based on replication-incompetent influenza virus, against pathogens that cause respiratory infectious diseases. IMPORTANCE: Streptococcus pneumoniae and influenza viruses cause contagious diseases, but no single vaccine can simultaneously provide protective immunity against both pathogens. Here, we used reverse genetics to generate a replication-incompetent influenza virus carrying the sequence for the antigenic region of pneumococcal surface protein A and demonstrated that mice immunized with this virus were completely protected from lethal doses of infection with either influenza virus or Streptococcus pneumoniae. We believe that this strategy, which is based on a replication-incompetent influenza virus possessing the antigenic region of other respiratory pathogens, offers a platform for the development of bivalent vaccines.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Pneumococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Carrier State/prevention & control , Disease Models, Animal , Female , Gene Knockout Techniques , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A virus/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Nasopharynx/microbiology , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/immunology , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/genetics , Streptococcus pneumoniae/genetics , Survival Analysis , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
11.
J Virol ; 86(8): 4123-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22301144

ABSTRACT

Vaccination is the primary form of protection from influenza virus infection. We recently developed a replication-incompetent PB2-knockout (PB2-KO) influenza virus that possesses a reporter gene (the green fluorescent protein gene) in the coding region of the PB2 segment. This virus replicated to high titers in PB2-expressing, but not unmodified, cells, suggesting its potential safety and feasibility as a vaccine. Here, we tested its efficacy in a murine model. The levels of IgG and IgA antibodies against influenza virus in sera, nasal washes, and bronchoalveolar lavage fluids of mice immunized with the PB2-KO virus were higher than those induced by a conventional inactivated vaccine. All PB2-KO virus-immunized mice survived challenges with lethal doses of influenza virus. Moreover, importantly, mice immunized with the PB2-KO virus produced antibodies against the reporter protein, suggesting that the PB2-KO virus has potential as a multivalent vaccine to combat infection with not only influenza virus but also other pathogens.


Subject(s)
Influenza A virus/genetics , Influenza A virus/immunology , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Virus Replication/genetics , Administration, Intranasal , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody Specificity/immunology , Body Weight , Dogs , Female , Genetic Vectors , Humans , Influenza Vaccines/administration & dosage , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/prevention & control , Turbinates/virology
12.
J Virol ; 86(3): 1405-10, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22090129

ABSTRACT

Vaccination is one of the most effective preventive measures to combat influenza. Prospectively, cell culture-based influenza vaccines play an important role for robust vaccine production in both normal settings and urgent situations, such as during the 2009 pandemic. African green monkey Vero cells are recommended by the World Health Organization as a safe substrate for influenza vaccine production for human use. However, the growth of influenza vaccine seed viruses is occasionally suboptimal in Vero cells, which places limitations on their usefulness for enhanced vaccine production. Here, we present a strategy for the development of vaccine seed viruses with enhanced growth in Vero cells by changing an amino acid residue in the stem region of the HA2 subunit of the hemagglutinin (HA) molecule. This mutation optimized the pH for HA-mediated membrane fusion in Vero cells and enhanced virus growth 100 to 1,000 times in the cell line, providing a promising strategy for cell culture-based influenza vaccines.


Subject(s)
Hydrogen-Ion Concentration , Influenza A virus/growth & development , Influenza Vaccines/biosynthesis , Membrane Fusion , Adaptation, Physiological , Animals , Chlorocebus aethiops , Dogs , Influenza A virus/immunology , Influenza A virus/physiology , Vero Cells
13.
Nat Commun ; 14(1): 4956, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37653024

ABSTRACT

The molecular etiology of idiopathic pulmonary fibrosis (IPF) has been extensively investigated to identify new therapeutic targets. Although anti-inflammatory treatments are not effective for patients with IPF, damaged alveolar epithelial cells play a critical role in lung fibrogenesis. Here, we establish an organoid-based lung fibrosis model using mouse and human lung tissues to assess the direct communication between damaged alveolar type II (AT2)-lineage cells and lung fibroblasts by excluding immune cells. Using this in vitro model and mouse genetics, we demonstrate that bleomycin causes DNA damage and activates p53 signaling in AT2-lineage cells, leading to AT2-to-AT1 transition-like state with a senescence-associated secretory phenotype (SASP). Among SASP-related factors, TGF-ß plays an exclusive role in promoting lung fibroblast-to-myofibroblast differentiation. Moreover, the autocrine TGF-ß-positive feedback loop in AT2-lineage cells is a critical cellular system in non-inflammatory lung fibrogenesis. These findings provide insights into the mechanism of IPF and potential therapeutic targets.


Subject(s)
Idiopathic Pulmonary Fibrosis , Transforming Growth Factor beta , Humans , Animals , Mice , Feedback , Alveolar Epithelial Cells , Idiopathic Pulmonary Fibrosis/genetics , Cell Differentiation
14.
PLoS Pathog ; 6(8): e1001079, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20865125

ABSTRACT

Influenza viruses resistant to antiviral drugs emerge frequently. Not surprisingly, the widespread treatment in many countries of patients infected with 2009 pandemic influenza A (H1N1) viruses with the neuraminidase (NA) inhibitors oseltamivir and zanamivir has led to the emergence of pandemic strains resistant to these drugs. Sporadic cases of pandemic influenza have been associated with mutant viruses possessing a histidine-to-tyrosine substitution at position 274 (H274Y) in the NA, a mutation known to be responsible for oseltamivir resistance. Here, we characterized in vitro and in vivo properties of two pairs of oseltaimivir-sensitive and -resistant (possessing the NA H274Y substitution) 2009 H1N1 pandemic viruses isolated in different parts of the world. An in vitro NA inhibition assay confirmed that the NA H274Y substitution confers oseltamivir resistance to 2009 H1N1 pandemic viruses. In mouse lungs, we found no significant difference in replication between oseltamivir-sensitive and -resistant viruses. In the lungs of mice treated with oseltamivir or even zanamivir, 2009 H1N1 pandemic viruses with the NA H274Y substitution replicated efficiently. Pathological analysis revealed that the pathogenicities of the oseltamivir-resistant viruses were comparable to those of their oseltamivir-sensitive counterparts in ferrets. Further, the oseltamivir-resistant viruses transmitted between ferrets as efficiently as their oseltamivir-sensitive counterparts. Collectively, these data indicate that oseltamivir-resistant 2009 H1N1 pandemic viruses with the NA H274Y substitution were comparable to their oseltamivir-sensitive counterparts in their pathogenicity and transmissibility in animal models. Our findings highlight the possibility that NA H274Y-possessing oseltamivir-resistant 2009 H1N1 pandemic viruses could supersede oseltamivir-sensitive viruses, as occurred with seasonal H1N1 viruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Oseltamivir/pharmacology , Animals , Female , Ferrets , Male , Mice , Mice, Inbred BALB C , Mutation , Neuraminidase/genetics , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/transmission , Pandemics , Virus Replication/drug effects
15.
Dev Biol ; 341(2): 459-63, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20227405

ABSTRACT

The left-right (LR) axis is essential for the proper function of internal organs. In mammals and fish, left-sided Nodal expression governs LR patterning. Here, we show that the Polycomb group protein Ezh1, which is highly conserved from fish to human, participates in LR patterning. Knockdown of olezh1, a medaka homologue of Ezh1, led to LR reversal of internal organs. It was shown that OLEZH1 acts in silencing the expression of Spaw (a medaka homolog of Nodal) via a previously unknown pathway. Furthermore, coimmunoprecipitation showed physical interaction of Ezh1 with FoxH1, a Nodal regulator. This represents a novel mechanism for LR patterning and implies that Ezh1 has developmental importance.


Subject(s)
Body Patterning , Fish Proteins/genetics , Nodal Protein/genetics , Oryzias/embryology , Repressor Proteins/metabolism , Animals , Gene Expression Regulation, Developmental , Humans , Oryzias/metabolism , Polycomb-Group Proteins
16.
Am J Transl Res ; 13(11): 12318-12337, 2021.
Article in English | MEDLINE | ID: mdl-34956455

ABSTRACT

Metformin, an AMP-activated protein kinase activator used to treat diabetes mellitus, has recently attracted attention as a promising anti-fibrotic agent. However, its anti-fibrotic effects on pleural fibroelastosis remain unknown. We induced mouse pleural fibroelastosis by intra-pleural coadministration of bleomycin and carbon and evaluated its validity as a preclinical model for human pleural fibrosis. We assessed the expression of the myofibroblast surface marker CD90 in the fibrotic pleura and the effects of metformin in vivo and in vitro. Finally, we evaluated the effects of metformin on human pleural mesothelial cells stimulated by transforming growth factor ß1 (TGFß1). The fibrotic pleura in mice had collagen and elastin fiber deposition similar to that seen in human fibrotic pleura. Moreover, CD90-positive myofibroblasts were detected in and successfully isolated from the fibrotic pleura. Metformin significantly suppressed the deposition of collagen and elastic fibers in the fibrotic pleura and decreased the expression of extracellular matrix (ECM)-related genes, including Col1a1, Col3a1, Fn1, and Eln, in pleural CD90-positive myofibroblasts. In human pleural mesothelial cells, metformin decreased TGFß1-induced upregulation of ECM-related genes and SNAI1. Overall, metformin suppresses pleural fibroelastosis by inhibition of ECM production by pleural myofibroblasts, suggesting that this drug has therapeutic potential against human pleural fibrosis, including pleuroparenchymal fibroelastosis.

17.
Nat Cell Biol ; 22(8): 934-946, 2020 08.
Article in English | MEDLINE | ID: mdl-32661339

ABSTRACT

Stem cells undergo dynamic changes in response to injury to regenerate lost cells. However, the identity of transitional states and the mechanisms that drive their trajectories remain understudied. Using lung organoids, multiple in vivo repair models, single-cell transcriptomics and lineage tracing, we find that alveolar type-2 epithelial cells undergoing differentiation into type-1 cells acquire pre-alveolar type-1 transitional cell state (PATS) en route to terminal maturation. Transitional cells undergo extensive stretching during differentiation, making them vulnerable to DNA damage. Cells in the PATS show an enrichment of TP53, TGFß, DNA-damage-response signalling and cellular senescence. Gain and loss of function as well as genomic binding assays revealed a direct transcriptional control of PATS by TP53 signalling. Notably, accumulation of PATS-like cells in human fibrotic lungs was observed, suggesting persistence of the transitional state in fibrosis. Our study thus implicates a transient state associated with senescence in normal epithelial tissue repair and its abnormal persistence in disease conditions.


Subject(s)
Alveolar Epithelial Cells , Cell Differentiation , Pulmonary Fibrosis/pathology , Adult Stem Cells/pathology , Alveolar Epithelial Cells/pathology , Animals , Cell Lineage , Cell Shape , Cellular Senescence , DNA Damage , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Organoids , Pulmonary Fibrosis/genetics , Signal Transduction , Tumor Suppressor Protein p53/metabolism
18.
Cell Stem Cell ; 27(6): 890-904.e8, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33128895

ABSTRACT

Coronavirus infection causes diffuse alveolar damage leading to acute respiratory distress syndrome. The absence of ex vivo models of human alveolar epithelium is hindering an understanding of coronavirus disease 2019 (COVID-19) pathogenesis. Here, we report a feeder-free, scalable, chemically defined, and modular alveolosphere culture system for the propagation and differentiation of human alveolar type 2 cells/pneumocytes derived from primary lung tissue. Cultured pneumocytes express the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor angiotensin-converting enzyme receptor type-2 (ACE2) and can be infected with virus. Transcriptome and histological analysis of infected alveolospheres mirror features of COVID-19 lungs, including emergence of interferon (IFN)-mediated inflammatory responses, loss of surfactant proteins, and apoptosis. Treatment of alveolospheres with IFNs recapitulates features of virus infection, including cell death. In contrast, alveolospheres pretreated with low-dose IFNs show a reduction in viral replication, suggesting the prophylactic effectiveness of IFNs against SARS-CoV-2. Human stem cell-based alveolospheres, thus, provide novel insights into COVID-19 pathogenesis and can serve as a model for understanding human respiratory diseases.


Subject(s)
Adult Stem Cells/virology , Alveolar Epithelial Cells/drug effects , COVID-19 Drug Treatment , Interferons/pharmacology , SARS-CoV-2/immunology , Adult , Adult Stem Cells/drug effects , Adult Stem Cells/enzymology , Aged , Aged, 80 and over , Alveolar Epithelial Cells/enzymology , Alveolar Epithelial Cells/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/physiopathology , Cell Culture Techniques , Cell Differentiation , Female , Humans , Inflammation , Male , Mice , Receptors, Coronavirus/metabolism , Transcriptome , Virus Replication
19.
Stem Cell Reports ; 12(4): 657-666, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30930244

ABSTRACT

Inflammatory responses are known to facilitate tissue recovery following injury. However, the precise mechanisms that enhance lung alveolar regeneration remain unclear. Here, using an organoid-based screening assay, we find that interleukin-1 (IL-1) and tumor necrosis factor α (TNFα) enhance the proliferation of AEC2s while maintaining their differentiation capacity. Furthermore, we find that expression of IL-1ß and TNFα are induced in the AEC2 niche following influenza-induced injury in vivo, and lineage tracing analysis revealed that surviving AEC2s around the damaged area contribute to alveolar regeneration. Through genetic and pharmacological modulation of multiple components of the IL-1-nuclear factor κB (NF-κB) signaling axis, we show that cell-intrinsic as well as stromal mediated IL-1 signaling are essential for AEC2 mediated lung regeneration. Taken together, we propose that the IL-1/TNFα-NF-κB signaling axis functions as a component of an inflammation-associated niche to regulate proliferation of surviving AEC2s and promote lung regeneration.


Subject(s)
Cellular Microenvironment , Interleukin-1/metabolism , Pulmonary Alveoli/physiology , Regeneration , Tumor Necrosis Factor-alpha/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cellular Microenvironment/genetics , Cytokines/metabolism , Fluorescent Antibody Technique , Humans , Inflammation Mediators , Influenza, Human , NF-kappa B/metabolism , Regeneration/genetics , Signal Transduction
20.
Sci Rep ; 6: 19933, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26847414

ABSTRACT

Influenza viruses that express reporter proteins are useful tools, but are often attenuated. Recently, we found that an influenza virus encoding the Venus fluorescent protein acquired two mutations in its PB2 and HA proteins upon mouse adaptation. Here, we demonstrate that the enhanced viral replication and virulence in mice of this Venus-expressing influenza virus are primarily conferred by the PB2-E712D mutation, with only a minor contribution by the HA-T380A mutation.


Subject(s)
Hemagglutinins, Viral/genetics , Luminescent Proteins/metabolism , Orthomyxoviridae/metabolism , Viral Proteins/genetics , Amino Acid Substitution , Animals , Dogs , Female , Genes, Reporter , HEK293 Cells , Hemagglutinins, Viral/chemistry , Hemagglutinins, Viral/metabolism , Humans , Hydrogen-Ion Concentration , Luminescent Proteins/genetics , Madin Darby Canine Kidney Cells , Membrane Fusion , Mice , Mice, Inbred C57BL , Orthomyxoviridae/growth & development , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Viral Proteins/chemistry , Viral Proteins/metabolism , Virulence/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL