1
|
Wang Y, Ma C, Hao X, Wang W, Luo H, Li M. Identification of Mycoplasma pneumoniae proteins interacting with NOD2 and their role in macrophage inflammatory response. Front Microbiol 2024; 15:1391453. [PMID: 38863748 PMCID: PMC11165193 DOI: 10.3389/fmicb.2024.1391453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024] Open
Abstract
Mycoplasma pneumoniae (M. pneumoniae, Mp) is a cell wall-deficient microorganism known to cause chronic respiratory infections in both children and adults. Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) is an intracellular pattern recognition receptor primarily responsible for identifying muramyl dipeptide (MDP) found in bacterial cell walls. Previous experiments have demonstrated that Mycoplasma ovipneumoniae induces macrophage autophagy through NOD2. In this study, we conducted RNA-seq analysis on macrophages infected with M. pneumoniae and observed an up-regulation in the expression of genes associated with the NOD2 signaling pathway. Mechanistic investigations further revealed the involvement of the NOD2 signaling pathway in the inflammatory response of macrophages activated by M. pneumoniae. We utilized GST pull-down technology in conjunction with liquid chromatography-tandem mass spectrometry (LC-MS/MS) to pinpoint the M. pneumoniae proteins that interact with NOD2. Additionally, co-immunoprecipitation (Co-IP) and immunofluorescence co-localization techniques were used to confirm the interaction between DUF16 protein and NOD2. We found that DUF16 protein can enter macrophages and induce macrophage inflammatory response through the NOD2/RIP2/NF-κB pathway. Notably, the region spanning amino acids 13-90 was identified as a critical region necessary for DUF16-induced inflammation. This research not only broadens our comprehension of the recognition process of the intracellular receptor NOD2, but also deepens our understanding of the development of M. pneumoniae infection.
Collapse
Affiliation(s)
- Yongyu Wang
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Chunji Ma
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
- Ningxia Polytechnic College, Yinchuan, China
| | - Xiujing Hao
- Life Science School, Ningxia University, Yinchuan, China
| | - Weili Wang
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Haixia Luo
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Min Li
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| |
Collapse
|
2
|
Chen J, Wang S, Dong D, Zhang Z, Huang Y, Zhang Y. Isolation and Characterization of Mycoplasma ovipneumoniae Infecting Goats with Pneumonia in Anhui Province, China. Life (Basel) 2024; 14:218. [PMID: 38398727 PMCID: PMC10890177 DOI: 10.3390/life14020218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Mycoplasma ovipneumoniae (M. ovipneumoniae) causes a fatal infection in goats, leading to significant economic losses in the small-ruminant industry worldwide. The present study aimed to characterize the strains of M. ovipneumoniae infecting goats with pneumonia in Anhui Province, China. From November 2021 to January 2023, among 20 flocks, a total of 1320 samples (600 samples of unvaccinated blood, 400 nasal swabs, 200 samples of pleural fluid, and 120 samples of lung tissue) were obtained from goats with typical signs of pneumonia, such as a low growth rate, appetite suppression, increased temperature, discharge from the nose, and a cough. Necropsied goats showed increased pleural fluid, fibrinous pleuropneumonia, and attached localized pleural adhesions. M. ovipneumoniae isolated from the samples were subjected to an indirect hemagglutination test (IHA), PCR amplicon sequencing, phylogenetic analysis, and biochemical identification tests. The overall positivity rate of M. ovipneumoniae was 27.50%. Mycoplasmas were obtained from 80 (20.0%) nasal swabs, 21 (10.5%) pleural fluid samples, and 15 (12.5%) lung samples. PCR amplicon (288 bp) sequencing identified eight strains of M. ovipneumoniae. In a phylogenetic tree, the isolated strains were homologous to the standard strain M. ovipneumoniae Y-98 and most similar to M. ovipneumoniae FJ-SM. Local strains of M. ovipneumoniae were isolated from goats in Anhui province. The identified genomic features and population structure will promote further study of M. ovipneumoniae pathogenesis and could form the basis for vaccine and therapy development.
Collapse
Affiliation(s)
- Jiahong Chen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (S.W.); (Z.Z.); (Y.H.)
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Dingyuan 233200, China;
| | - Shijia Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (S.W.); (Z.Z.); (Y.H.)
| | - Dong Dong
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Dingyuan 233200, China;
| | - Zijun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (S.W.); (Z.Z.); (Y.H.)
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Dingyuan 233200, China;
| | - Yafeng Huang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (S.W.); (Z.Z.); (Y.H.)
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Dingyuan 233200, China;
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| |
Collapse
|
3
|
Ma C, Hao X, Gao L, Wang Y, Shi J, Luo H, Li M. Extracellular Vesicles Released from Macrophages Infected with Mycoplasma pneumoniae Stimulate Proinflammatory Response via the TLR2-NF-κB/JNK Signaling Pathway. Int J Mol Sci 2023; 24:ijms24108588. [PMID: 37239946 DOI: 10.3390/ijms24108588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Mycoplasma pneumoniae (M. pneumoniae, Mp) is an intracellular pathogen that causes pneumonia, tracheobronchitis, pharyngitis, and asthma in humans and can infect and survive in the host cells leading to excessive immune responses. Extracellular vesicles (EVs) from host cells carry components of pathogens to recipient cells and play a role in intercellular communication during infection. However, there is limited knowledge on whether EVs derived from M. pneumoniae-infected macrophages play as intercellular messengers and functional mechanisms. In this study, we establish a cell model of M. pneumoniae-infected macrophages that continuously secrete EVs to further asses their role as intercellular messengers and their functional mechanisms. Based on this model, we determined a method for isolating the pure EVs from M. pneumoniae-infected macrophages, which employs a sequence of operations, including differential centrifugation, filtering, and ultracentrifugation. We identified EVs and their purity using multiple methods, including electron microscopy, nanoparticle tracking analysis, Western blot, bacteria culture, and nucleic acid detection. EVs from M. pneumoniae-infected macrophages are pure, with a 30-200 nm diameter. These EVs can be taken up by uninfected macrophages and induce the production of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-8 through the nuclear factor (NF)-κB, and mitogen-activated protein kinases (MAPK) signals pathway. Moreover, the expression of inflammatory cytokines induced by EVs relies on TLR2-NF-κB/JNK signal pathways. These findings will help us better understand a persistent inflammatory response and cell-to-cell immune modulation in the context of M. pneumoniae infection.
Collapse
Affiliation(s)
- Chunji Ma
- Life Science School, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Xiujing Hao
- Life Science School, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Liyang Gao
- Life Science School, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Yongyu Wang
- Life Science School, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Juan Shi
- Life Science School, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Haixia Luo
- Life Science School, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| | - Min Li
- Life Science School, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan 750021, China
| |
Collapse
|
4
|
Chen X, Ishfaq M, Wang J. Baicalin ameliorates Mycoplasma gallisepticum-induced inflammatory injury via inhibiting STIM1-regulated ceramide accumulation in DF-1 cells. Poult Sci 2023; 102:102687. [PMID: 37099879 PMCID: PMC10149409 DOI: 10.1016/j.psj.2023.102687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Mycoplasma gallisepticum (MG) is dependent on its host for many nutrients due to the loss of many important metabolic pathways. Ceramide is a sphingolipid that regulates multiple cellular processes in eukaryotic cell. Several studies highlighted the crucial role of ceramide on the pathogenesis of various pathogens. This study aimed to determine whether ceramide plays a crucial role in the pathogenesis of MG. Based on an MG infection model in DF-1 cells, the results revealed that MG infection induced ceramide accumulation in DF-1 cells. Inhibiting the de novo synthesis of ceramide significantly inhibited MG proliferation and inflammatory injury caused by MG in DF-1 cells. Meanwhile, MG infection led to endoplasmic reticulum stress, and pharmacologic inhibition of endoplasmic reticulum stress prevented ceramide accumulation and MG proliferation in DF-1 cells, alleviating the inflammatory injury caused by MG. In addition, MG infection significantly promoted expression level of stromal interaction molecule 1 (STIM1), thus induced calcium overload and oxidative stress. Furthermore, inhibition of STIM1 expression partially restored calcium homeostasis and mitigated oxidative stress, thus alleviated endoplasmic reticulum stress. Importantly, the inflammatory injury caused by MG were partially ameliorated by baicalin treatment (20 µg/mL) through downregulating STIM1 expression. In summary, these results suggests that ceramide accumulation through the de novo pathway plays an important role to promote MG proliferation and baicalin can alleviate MG infection induced inflammatory injury via regulating STIM1-related oxidative stress, endoplasmic reticulum stress and ceramide accumulation in DF-1 cells.
Collapse
|
5
|
Xie Y, Hu W, Chen X, Ren P, Ye C, Wang Y, Luo J, Li X. Identification and validation of autophagy-related genes in exogenous sepsis-induced acute respiratory distress syndrome. Immun Inflamm Dis 2022; 10:e691. [PMID: 36169246 PMCID: PMC9500593 DOI: 10.1002/iid3.691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/19/2022] [Accepted: 08/05/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE To analyze the differential expression of autophagy-related genes of sepsis-induced acute respiratory distress syndrome (ARDS) as potential markers for early diagnosis. METHODS Male Sprague-Dawley rats (aged 8 weeks) were selected and randomly divided into sepsis-induced ARDS group (n = 6) and a normal control group (n = 6). Lung tissue samples were collected for high-throughput sequencing using Illumina HiSeq sequencing platform in the paired-end sequencing mode. Differentially expressed genes (DEGs) were screened by DESeq. 2 software [|log2FC | ≥1 and p < .05] and autophagy-related genes were identified using Mouse Genome Informatics. Co-expressed autophagy-related DEGs from these two datasets were filtered by construction of a Venn diagram. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on these autophagy-related DEGs and a protein interaction network was constructed using STRING and Cytoscape software to identify hub genes, which were verified by real-time quantitative polymerase chain reaction (qRT-PCR). RESULTS A total of 42 autophagy-related DEGs (26 upregulated genes and 16 downregulated genes) were identified. The GO and KEGG pathway analyses showed enrichment in 969 biological processes (BPs), three cellular components (CCs), eight molecular functions (MFs) and 27 signaling pathways. The protein interaction (PPI) network revealed 42 node proteins and 75 interacting edges, with an average node degree of 3.52, and an average local clustering coefficient of 0.509. Among the top 10 hub genes with the RNA-Seq, six hub genes (Stat3, Il10, Ifng, Hmox1, Hif1a, and Nod2) were validated by qRT-PCR (all p < .05). CONCLUSION 42 potential autophagy-related genes associated with sepsis-induced ARDS lung injury were identified and six hub genes (Stat3, Il10, Ifng, Hmox1, Hif1a, and Nod2) may affect the development of ARDS by regulating autophagy. These results expanded our understanding of ARDS and might be useful in treatment of exogenous sepsis-induced ARDS.
Collapse
Affiliation(s)
- Yongpeng Xie
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
- The Institute of Emergency Medicine of LianyungangLianyungangJiangsuChina
| | - Wenxia Hu
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
- The Institute of Emergency Medicine of LianyungangLianyungangJiangsuChina
| | - Xiaobin Chen
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
- The Institute of Emergency Medicine of LianyungangLianyungangJiangsuChina
| | - Panpan Ren
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
| | - Chongchong Ye
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
| | - Yanli Wang
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
| | - Jiye Luo
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
| | - Xiaomin Li
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical UniversityThe First People's Hospital of LianyungangLianyungangJiangsuChina
- The Institute of Emergency Medicine of LianyungangLianyungangJiangsuChina
| |
Collapse
|
6
|
Wen Y, Chen Z, Tian Y, Yang M, Dong Q, Yang Y, Ding H. Incomplete autophagy promotes the proliferation of Mycoplasma hyopneumoniae through the JNK and Akt pathways in porcine alveolar macrophages. Vet Res 2022; 53:62. [PMID: 35927699 PMCID: PMC9351181 DOI: 10.1186/s13567-022-01074-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/24/2022] [Indexed: 11/10/2022] Open
Abstract
Autophagy is an important conserved homeostatic process related to nutrient and energy deficiency and organelle damage in diverse eukaryotic cells and has been reported to play an important role in cellular responses to pathogens and bacterial replication. The respiratory bacterium Mycoplasma hyopneumoniae has been identified to enter porcine alveolar macrophages, which are considered important immune cells. However, little is known about the role of autophagy in the pathogenesis of M. hyopneumoniae infection of porcine alveolar macrophages. Our experiments demonstrated that M. hyopneumoniae infection enhanced the formation of autophagosomes in porcine alveolar macrophages but prevented the fusion of autophagosomes with lysosomes, thereby blocking autophagic flux and preventing the acidification and destruction of M. hyopneumoniae in low-pH surroundings. In addition, using different autophagy regulators to intervene in the autophagy process, we found that incomplete autophagy promoted the intracellular proliferation of M. hyopneumoniae. We also found that blocking the phosphorylation of JNK and Akt downregulated the autophagy induced by M. hyopneumoniae, but pathways related to two mitogen-activated protein kinases (Erk1/2 and p38) did not affect the process. Collectively, M. hyopneumoniae induced incomplete autophagy in porcine alveolar macrophages through the JNK and Akt signalling pathways; conversely, incomplete autophagy prevented M. hyopneumoniae from entering and degrading lysosomes to realize the proliferation of M. hyopneumoniae in porcine alveolar macrophages. These findings raise the possibility that targeting the autophagic pathway may be effective for the prevention or treatment of M. hyopneumoniae infection.
Collapse
Affiliation(s)
- Yukang Wen
- Laboratory of Veterinary Mycoplasmology, College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - Zhengkun Chen
- Laboratory of Veterinary Mycoplasmology, College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - Yaqin Tian
- Laboratory of Veterinary Mycoplasmology, College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - Mei Yang
- Laboratory of Veterinary Mycoplasmology, College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - Qingshuang Dong
- Laboratory of Veterinary Mycoplasmology, College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - Yujiao Yang
- Laboratory of Veterinary Mycoplasmology, College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - Honglei Ding
- Laboratory of Veterinary Mycoplasmology, College of Veterinary Medicine, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
7
|
Xu M, Liu Y, Mayinuer T, Lin Y, Wang Y, Gao J, Wang D, Kastelic JP, Han B. Mycoplasma bovis inhibits autophagy in bovine mammary epithelial cells via a PTEN/PI3K-Akt-mTOR-dependent pathway. Front Microbiol 2022; 13:935547. [PMID: 35958147 PMCID: PMC9360976 DOI: 10.3389/fmicb.2022.935547] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 01/18/2023] Open
Abstract
Although autophagy can eliminate some intracellular pathogens, others, e.g., Staphylococcus aureus, Salmonella, Mycoplasma bovis, can evade it. The phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway, a key regulator of autophagy, is involved in initiation and promotion of a range of pathological diseases. As the effects of M. bovis on the autophagic pathway are not well documented, our objective was to elucidate the effects of M. bovis infection on the PI3K-Akt-mTOR cellular autophagic pathway in bovine mammary epithelial cells (bMECs). Ultrastructure of bMECs infected with M. bovis was assessed with transmission electron microscopy, co-localization of LC3 puncta with M. bovis was confirmed by laser confocal microscopy, and autophagy-related indicators were quantified with Western blotting and RT-PCR. In M. bovis-infected bMECs, intracellular M. bovis was encapsulated by membrane-like structures, the expression level of LC3-II and Beclin1 protein decreased at the middle stage of infection, degradation of SQSTM1/P62 was blocked, autophagy of bMECs was inhibited, and PI3K-Akt-mTOR protein was activated by phosphorylation. Furthermore, the tumor suppressor PTEN can inhibit the PI3K-Akt signaling pathway through dephosphorylation of phosphatidylinositol 3,4,5-trisphosphate and may be important for cellular resistance to infection. In the present study, the number of intracellular M. bovis was inversely related to the change in the level of autophagy markers (e.g., LC3-II, SQSTM1/P62) within host cells induced by the low knockdown of Akt or PTEN. We concluded that M. bovis-infected bMECs alleviated cellular autophagy through a PI3K-Akt-mTOR pathway, and that PTEN acted as a protective gene regulating autophagy, a key step in controlling infection.
Collapse
Affiliation(s)
- Maolin Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yang Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Tuerdi Mayinuer
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yushan Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yue Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dong Wang
- College of Life Science, Ningxia University, Yinchuan, China
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
- *Correspondence: Bo Han,
| |
Collapse
|
8
|
Chen J, Zhou Y, Zhu E, Yang P, Li M, Zhang S, Yue J, Wen M, Wang K, Cheng Z. Mycoplasma ovipneumoniae induces caspase-8-dependent extrinsic apoptosis and p53- and ROS-dependent intrinsic apoptosis in murine alveolar macrophages. Virulence 2021; 12:2703-2720. [PMID: 34678131 PMCID: PMC8923071 DOI: 10.1080/21505594.2021.1984714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mycoplasma ovipneumoniae (MO) is a principle causative agent of chronic respiratory disease in ruminants, including sheep, goats, and deer, posing a great threat to the ruminant industry worldwide. However, the pathogenesis of MO infection still remains not well understood and needs further clarification. Here we report a time-dependent apoptosis in cultured murine alveolar macrophage (MH-S) cell lines in response to MO infection in vitro. Mechanistically, MO infection activated apoptosis in MH-S cells through caspase-8-dependent extrinsic pathway and through tumor protein 53 (p53)- and reactive oxygen species (ROS)-dependent intrinsic mitochondrial pathways. Moreover, MO infection promoted both transcription and translation of proinflammatory cytokine genes including interleukin-1β (IL-1β), IL-18, and tumor necrosis factor-α (TNF-α), in a caspase-8-, p53-, and ROS-dependent manner, implying a potential link between MO-induced inflammation and apoptotic cell death. Collectively, our results suggest that MO infection induces the activation of extrinsic and intrinsic apoptotic pathways in cultured MH-S cells, which is related to upregulated expression of proinflammatory cytokines. Our findings will contribute to the elucidation of pathogenesis in MO infection and provide valuable reference for the development of new strategies for controlling MO infection.
Collapse
Affiliation(s)
- Jing Chen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Yi Zhou
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Erpeng Zhu
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Peng Yang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Mei Li
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Shuangxiang Zhang
- The Laboratory of Veterinary Medicine, Animal Disease Prevention and Control Center of Guizhou Province, Guiyang, China
| | - Jun Yue
- The Laboratory of Veterinary Medicine, Animal Disease Prevention and Control Center of Guizhou Province, Guiyang, China
| | - Ming Wen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Kaigong Wang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Zhentao Cheng
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| |
Collapse
|
9
|
Liu Y, Deng Z, Xu S, Liu G, Lin Y, Khan S, Gao J, Qu W, Kastelic JP, Han B. Mycoplasma bovis subverts autophagy to promote intracellular replication in bovine mammary epithelial cells cultured in vitro. Vet Res 2021; 52:130. [PMID: 34649594 PMCID: PMC8515657 DOI: 10.1186/s13567-021-01002-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/30/2021] [Indexed: 01/18/2023] Open
Abstract
Mycoplasma species are the smallest prokaryotes capable of self-replication. To investigate Mycoplasma induced autophagy in mammalian cells, Mycoplasma bovis (M. bovis) and bovine mammary epithelial cells (bMEC) were used in an in vitro infection model. Initially, intracellular M. bovis was enclosed within a membrane-like structure in bMEC, as viewed with transmission electron microscopy. In infected bMEC, increased LC3II was verified by Western blotting, RT-PCR and laser confocal microscopy, confirming autophagy at 1, 3 and 6 h post-infection (hpi), with a peak at 6 hpi. However, the M. bovis-induced autophagy flux was subsequently blocked. P62 degradation in infected bMEC was inhibited at 3, 6, 12 and 24 hpi, based on Western blotting and RT-PCR. Beclin1 expression decreased at 12 and 24 hpi. Furthermore, autophagosome maturation was subverted by M. bovis. Autophagosome acidification was inhibited by M. bovis infection, based on detection of mCherry-GFP-LC3 labeled autophagosomes; the decreases in protein levels of Lamp-2a indicate that the lysosomes were impaired by infection. In contrast, activation of autophagy (with rapamycin or HBSS) overcame the M. bovis-induced blockade in phagosome maturation by increasing delivery of M. bovis to the lysosome, with a concurrent decrease in intracellular M. bovis replication. In conclusion, although M. bovis infection induced autophagy in bMEC, the autophagy flux was subsequently impaired by inhibiting autophagosome maturation. Therefore, we conclude that M. bovis subverted autophagy to promote its intracellular replication in bMEC. These findings are the impetus for future studies to further characterize interactions between M. bovis and mammalian host cells.
Collapse
Affiliation(s)
- Yang Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Zhaoju Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Siyu Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Gang Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yushan Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Sohrab Khan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Weijie Qu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - John P Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
10
|
Mousel MR, White SN, Herndon MK, Herndon DR, Taylor JB, Becker GM, Murdoch BM. Genes involved in immune, gene translation and chromatin organization pathways associated with Mycoplasma ovipneumoniae presence in nasal secretions of domestic sheep. PLoS One 2021; 16:e0247209. [PMID: 34252097 PMCID: PMC8274911 DOI: 10.1371/journal.pone.0247209] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/26/2021] [Indexed: 12/20/2022] Open
Abstract
Mycoplasma ovipneumoniae contributes to polymicrobial pneumonia in domestic sheep. Elucidation of host genetic influences of M. ovipneumoniae nasal detection has the potential to reduce the incidence of polymicrobial pneumonia in sheep through implementation of selective breeding strategies. Nasal mucosal secretions were collected from 647 sheep from a large US sheep flock. Ewes of three breeds (Polypay n = 222, Rambouillet n = 321, and Suffolk n = 104) ranging in age from one to seven years, were sampled at three different times in the production cycle (February, April, and September/October) over four years (2015 to 2018). The presence and DNA copy number of M. ovipneumoniae was determined using a newly developed species-specific qPCR. Breed (P<0.001), age (P<0.024), sampling time (P<0.001), and year (P<0.001) of collection affected log10 transformed M. ovipneumoniae DNA copy number, where Rambouillet had the lowest (P<0.0001) compared with both Polypay and Suffolk demonstrating a possible genetic component to detection. Samples from yearlings, April, and 2018 had the highest (P<0.046) detected DNA copy number mean. Sheep genomic DNA was genotyped with the Illumina OvineHD BeadChip. Principal component analysis identified most of the variation in the dataset was associated with breed. Therefore, genome wide association analysis was conducted with a mixed model (EMMAX), with principal components 1 to 6 as fixed and a kinship matrix as random effects. Genome-wide significant (P<9x10-8) SNPs were identified on chromosomes 6 and 7 in the all-breed analysis. Individual breed analysis had genome-wide significant (P<9x10-8) SNPs on chromosomes 3, 4, 7, 9, 10, 15, 17, and 22. Annotated genes near these SNPs are part of immune (ANAPC7, CUL5, TMEM229B, PTPN13), gene translation (PIWIL4), and chromatin organization (KDM2B) pathways. Immune genes are expected to have increased expression when leukocytes encounter M. ovipneumoniae which would lead to chromatin reorganization. Work is underway to narrow the range of these associated regions to identify the underlying causal mutations.
Collapse
Affiliation(s)
- Michelle R. Mousel
- U.S. Department of Agriculture, Animal Disease Research Unit, Agricultural Research Service, Pullman, WA, United States of America
- Paul G. Allen School of Global Animal Health, Washington State University, Pullman, WA, United States of America
| | - Stephen N. White
- U.S. Department of Agriculture, Animal Disease Research Unit, Agricultural Research Service, Pullman, WA, United States of America
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States of America
- Center for Reproductive Biology, Washington State University, Pullman, WA, United States of America
| | - Maria K. Herndon
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States of America
| | - David R. Herndon
- U.S. Department of Agriculture, Animal Disease Research Unit, Agricultural Research Service, Pullman, WA, United States of America
| | - J. Bret Taylor
- U.S. Department of Agriculture, Range Sheep Production Efficiency Research, Agricultural Research Service, Dubois, ID, United States of America
| | - Gabrielle M. Becker
- Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States of America
| | - Brenda M. Murdoch
- Center for Reproductive Biology, Washington State University, Pullman, WA, United States of America
- Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States of America
| |
Collapse
|