1
|
Zhu Z, Zhang Y, Chen H, Zhang H. Cell-cell crosstalk in the pathogenesis of acute lung injury and acute respiratory distress syndrome. Tissue Barriers 2025:2452082. [PMID: 39798076 DOI: 10.1080/21688370.2025.2452082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/21/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are the result of an exaggerated inflammatory response triggered by a variety of pulmonary and systemic insults. The lung tissues are comprised of a variety of cell types, including alveolar epithelial cells, pulmonary vascular endothelial cells, macrophages, neutrophils, and others. There is mounting evidence that these diverse cell populations within the lung interact to regulate lung inflammation in response to both direct and indirect stimuli. The aim of this review is to provide a summary and discussion of recent advances in the understanding of the importance of cell-cell crosstalk in the pathogenesis of ALI/ARDS, with a specific focus on the cell-cell interactions that may offer prospective therapeutic avenues for ALI/ARDS.
Collapse
Affiliation(s)
- Zhenzhen Zhu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Ying Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huan Chen
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huali Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
2
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Kleb SS, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Fibroblast-Mediated Macrophage Recruitment Supports Acute Wound Healing. J Invest Dermatol 2024:S0022-202X(24)02956-7. [PMID: 39581458 DOI: 10.1016/j.jid.2024.10.609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single-nuclei RNA sequencing, we defined changes in gene expression associated with inflammation 1 day after wounding in mouse skin. Compared with those in keratinocytes and myeloid cells, we detected enriched expression of proinflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL-33, compared with SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound-bed macrophages and monocytes during injury-induced inflammation, with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M Amuso
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - MaryEllen R Haas
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Paula O Cooper
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Sana Hafiz
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Shatha Salameh
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Miguel F Mazumder
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Violet Josephson
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Sarah S Kleb
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Khatereh Khorsandi
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Anelia Horvath
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Brett A Shook
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA; The Department of Dermatology, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA.
| |
Collapse
|
3
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Deep skin fibroblast-mediated macrophage recruitment supports acute wound healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607357. [PMID: 39149286 PMCID: PMC11326280 DOI: 10.1101/2024.08.09.607357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single nuclei RNA-sequencing, we defined changes in gene expression associated with inflammation at 1-day post-wounding (dpw) in mouse skin. Compared to keratinocytes and myeloid cells, we detected enriched expression of pro-inflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL33 compared to SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound bed macrophages and monocytes during injury-induced inflammation with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of deep skin fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M. Amuso
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - MaryEllen R. Haas
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Paula O. Cooper
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Sana Hafiz
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Shatha Salameh
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Miguel F. Mazumder
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Violet Josephson
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Khatereh Khorsandi
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Brett A. Shook
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
- Department of Dermatology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
4
|
Suresh MV, Aktay S, Yalamanchili G, Solanki S, Sathyarajan DT, Arnipalli MS, Pennathur S, Raghavendran K. Role of succinate in airway epithelial cell regulation following traumatic lung injury. JCI Insight 2023; 8:e166860. [PMID: 37737265 PMCID: PMC10561732 DOI: 10.1172/jci.insight.166860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Lung contusion and gastric aspiration (LC and GA) are major risk factors for developing acute respiratory distress following trauma. Hypoxia from lung injury is mainly regulated by hypoxia-inducible factor 1α (HIF-1α). Published data from our group indicate that HIF-1α regulation in airway epithelial cells (AEC) drives the acute inflammatory response following LC and GA. Metabolomic profiling and metabolic flux of Type II AEC following LC revealed marked increases in glycolytic and TCA intermediates in vivo and in vitro that were HIF-1α dependent. GLUT-1/4 expression was also increased in HIF-1α+/+ mice, suggesting that increased glucose entry may contribute to increased intermediates. Importantly, lactate incubation in vitro on Type II cells did not significantly increase the inflammatory byproduct IL-1β. Contrastingly, succinate had a direct proinflammatory effect on human small AEC by IL-1β generation in vitro. This effect was reversed by dimethylmalonate, suggesting an important role for succinate dehydrogenase in mediating HIF-1α effects. We confirmed the presence of the only known receptor for succinate binding, SUCNR1, on Type II AEC. These results support the hypothesis that succinate drives HIF-1α-mediated airway inflammation following LC. This is the first report to our knowledge of direct proinflammatory activation of succinate in nonimmune cells such as Type II AEC in direct lung injury models.
Collapse
|
5
|
Sun Z, Chen A, Fang H, Sun D, Huang M, Cheng E, Luo M, Zhang X, Fang H, Qian G. B cell-derived IL-10 promotes the resolution of lipopolysaccharide-induced acute lung injury. Cell Death Dis 2023; 14:418. [PMID: 37443161 PMCID: PMC10345008 DOI: 10.1038/s41419-023-05954-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
Inflammation resolution is critical for acute lung injury (ALI) recovery. Interleukin (IL)-10 is a potent anti-inflammatory factor. However, its role in ALI resolution remains unclear. We investigated the effects of IL-10 during the ALI resolution process in a murine lipopolysaccharide (LPS)-induced ALI model. Blockade of IL-10 signaling aggravates LPS-induced lung injury, as manifested by elevated pro-inflammatory factors production and increased neutrophils recruitment to the lung. Thereafter, we used IL-10 GFP reporter mice to discern the source cell of IL-10 during ALI. We found that IL-10 is predominantly generated by B cells during the ALI recovery process. Furthermore, we used IL-10-specific loss in B-cell mice to elucidate the effect of B-cell-derived IL-10 on the ALI resolution process. IL-10-specific loss in B cells leads to increased pro-inflammatory cytokine expression, persistent leukocyte infiltration, and prolonged alveolar barrier damage. Mechanistically, B cell-derived IL-10 inhibits the activation and recruitment of macrophages and downregulates the production of chemokine KC that recruits neutrophils to the lung. Moreover, we found that IL-10 deletion in B cells leads to alterations in the cGMP-PKG signaling pathway. In addition, an exogenous supply of IL-10 promotes recovery from LPS-induced ALI, and IL-10-secreting B cells are present in sepsis-related ARDS. This study highlights that B cell-derived IL-10 is critical for the resolution of LPS-induced ALI and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Zhun Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Anning Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Hongwei Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Donglin Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Meiying Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Erdeng Cheng
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Mengyuan Luo
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaoren Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China.
| | - Guojun Qian
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Fesenmeier DJ, Suresh MV, Kim S, Raghavendran K, Won YY. Polymer Lung Surfactants Attenuate Direct Lung Injury in Mice. ACS Biomater Sci Eng 2023; 9:2716-2730. [PMID: 37079432 PMCID: PMC11973738 DOI: 10.1021/acsbiomaterials.3c00061] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
If not properly managed, acute lung injuries, either through direct or indirect causes, have the potential to present serious risk for many patients worldwide. One of the mechanisms for the transition from acute lung injury (ALI) to the more serious acute respiratory distress syndrome (ARDS) is the deactivation of the native lung surfactant by injury-induced infiltrates to the alveolar space. Currently, there are no surfactant replacement therapies that are used to treat ALI and subsequent ARDS. In this paper, we present an indepth efficacy study of using a novel polymer lung surfactant (PLS, composed of poly(styrene-block-ethylene glycol) (PS-PEG) block copolymer micelles), which has unique properties compared to other tested surfactant replacements, in two different mouse models of lung injury. The results demonstrate that pharyngeal administration of PLS after the instillation of either acid (HCl) or lipopolysaccharide (LPS) can decrease the severity of lung injury as measured by multiple injury markers.
Collapse
Affiliation(s)
- Daniel J. Fesenmeier
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN 47907, United States
| | | | - Seyoung Kim
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN 47907, United States
| | - Krishnan Raghavendran
- Department of Surgery, University of Michigan Medical
School, Ann Arbor, MI 48109, USA
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN 47907, United States
- Purdue University Center for Cancer Research, Purdue
University, West Lafayette, IN 47907, United States
| |
Collapse
|
7
|
Shimizu M, Hojo M, Ikushima K, Yamamoto Y, Maeno A, Sakamoto Y, Ishimaru N, Taquahashi Y, Kanno J, Hirose A, Suzuki J, Inomata A, Nakae D. Continuous infiltration of small peritoneal macrophages in the mouse peritoneum through CCR2-dependent and -independent routes during fibrosis and mesothelioma development induced by a multiwalled carbon nanotube, MWNT-7. J Toxicol Sci 2023; 48:617-639. [PMID: 38044124 DOI: 10.2131/jts.48.617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Although toxicities of multiwalled carbon nanotube (MWCNT) have been found to be related with activities of macrophages phagocytosing the fibers, the exact relationship between macrophage population and pathogenesis of fibrosis and mesotheliomas induced by MWCNTs is largely unknown. CCL2-CCR2 axis, a major monocyte/macrophage infiltration route, is thought to be involved in not only acute inflammation but also the formation of tumor microenvironment. We therefore described a time-course of alteration of macrophage population in an attempt to clarify the contribution of the Ccr2 gene to mesotheliomagenesis. Wild-type (WT) C57BL/6 mice and Ccr2-knockout (KO) mice were intraperitoneally administered with MWNT-7 and were sequentially necropsied at 1, 7, 28, 90, and 245 day(s) after the injection. Peritoneal fibrosis was prominent in all MWCNT-treated mice, with a lower severity in the KO mice. No differences were observed in the incidences of neoplastic lesions of mesothelia between WT and KO mice. A flow cytometric analysis revealed that after gross disappearance of macrophages after MWCNT exposure, small peritoneal macrophages (SPMs) were exclusively refurbished by the CCR2-dependent route at day 1 (as Ly-6C+MHC class II- cells), followed by additional CCR2-independent routes (as Ly-6C-MHC class II- cells); i.e., the only route in KO mice; with a delay of 1-7 days. The SPMs derived from both routes appeared to differentiate into maturated cells as Ly-6C-MHC class II+, whose ratio increased in a time-dependent manner among the total SPM population. Additionally, most macrophages expressed M1-like features, but a small fraction of macrophages exhibited an M1/M2 mixed status in MWCNT-treated animals. Our findings demonstrate a long-persistent activation of the CCL2-CCR2 axis after MWCNT exposure and enable a better understanding of the participation and potential roles of SPMs in fibrous material-induced chronic toxicities.
Collapse
Affiliation(s)
- Motomu Shimizu
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Motoki Hojo
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Kiyomi Ikushima
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Yukio Yamamoto
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Ai Maeno
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Yoshimitsu Sakamoto
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences
| | - Yuhji Taquahashi
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences
| | - Jun Kanno
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences
| | - Akihiko Hirose
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute, Japan
| | - Jin Suzuki
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Akiko Inomata
- Department of Pharmaceutical and Environmental Sciences, Tokyo Metropolitan Institute of Public Health
| | - Dai Nakae
- Department of Medical Sports, Faculty of Health Care and Medical Sports, Teikyo Heisei University
| |
Collapse
|
8
|
Suresh MV, Yalamanchili G, Rao TC, Aktay S, Kralovich A, Shah YM, Raghavendran K. Hypoxia‐inducible factor (HIF)‐1α‐induced regulation of lung injury in pulmonary aspiration is mediated through NF‐kB. FASEB Bioadv 2022; 4:309-328. [PMID: 35520392 PMCID: PMC9065579 DOI: 10.1096/fba.2021-00132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/11/2022] Open
Abstract
Aspiration‐induced lung injury is a common grievance encountered in the intensive care unit (ICU). It is a significant risk factor for improving ventilator‐associated pneumonia (VAP) and acute respiratory distress syndrome (ARDS). Hypoxia‐inducible factor (HIF)‐1α is one of the primary transcription factors responsible for regulating the cellular response to changes in oxygen tension. Here, we sought to determine the role of HIF‐1α and specifically the role of type 2 alveolar epithelial cells in generating the acute inflammatory response following acid and particles (CASP) aspiration. Previous studies show HIF‐1 α is involved in regulating the hypoxia‐stimulated expression of MCP‐1 in mice and humans. The CASP was induced in C57BL/6, ODD‐Luc, HIF‐1α (+/+) control, and HIF‐1α conditional knockout (HIF‐1α (−/−) mice). Following an injury in ODD mice, explanted organs were subjected to IVIS imaging to measure the degree of hypoxia. HIF‐1α expression, BAL albumin, cytokines, and histology were measured following CASP. In C57BL/6 mice, the level of HIF‐1α was increased at 1 h after CASP. There were significantly increased levels of albumin and cytokines in C57BL/6 and ODD‐Luc mice lungs following CASP. HIF‐1α (+/+) mice given CASP demonstrated a synergistic increase in albumin leakage, increased pro‐inflammatory cytokines, and worse injury. MCP‐1 antibody neutralized HIF‐1α (+/+) mice showed reduced granuloma formation. The NF‐κB expression was increased substantially in the HIF‐1α (+/+) mice following CASP compared to HIF‐1α (−/−) mice. Our data collectively identify that HIF‐1α upregulation of the acute inflammatory response depends on NF‐κB following CASP.
Collapse
Affiliation(s)
| | | | - Tejeshwar C. Rao
- Department of Cell, Developmental, and Integrative Biology The University of Alabama at Birmingham Birmingham UK
| | - Sinan Aktay
- Department of Surgery University of Michigan Ann Arbor Michigan USA
| | - Alex Kralovich
- Department of Surgery University of Michigan Ann Arbor Michigan USA
| | - Yatrik M. Shah
- Molecular & Integrative Physiology University of Michigan Ann Arbor Michigan USA
| | | |
Collapse
|
9
|
Shiraishi W, Yamasaki R, Hashimoto Y, Ko S, Kobayakawa Y, Isobe N, Matsushita T, Kira JI. Clearance of peripheral nerve misfolded mutant protein by infiltrated macrophages correlates with motor neuron disease progression. Sci Rep 2021; 11:16438. [PMID: 34385589 PMCID: PMC8360983 DOI: 10.1038/s41598-021-96064-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Macrophages expressing C-C chemokine receptor type 2 (CCR2) infiltrate the central and peripheral neural tissues of amyotrophic lateral sclerosis (ALS) patients. To identify the functional role of CCR2+ macrophages in the pathomechanisms of ALS, we used an ALS animal model, mutant Cu/Zn superoxide dismutase 1G93A (mSOD1)-transgenic (Tg) mice. To clarify the CCR2 function in the model, we generated SOD1G93A/CCR2Red fluorescence protein (RFP)/Wild type (WT)/CX3CR1Green fluorescence protein (GFP)/WT-Tg mice, which heterozygously express CCR2-RFP and CX3CR1-GFP, and SOD1G93A/CCR2RFP/RFP-Tg mice, which lack CCR2 protein expression and present with a CCR2-deficient phenotype. In mSOD1-Tg mice, mSOD1 accumulated in the sciatic nerve earlier than in the spinal cord. Furthermore, spinal cords of SOD1G93A/CCR2RFP/WT/CX3CR1GFP/WT mice showed peripheral macrophage infiltration that emerged at the end-stage, whereas in peripheral nerves, macrophage infiltration started from the pre-symptomatic stage. Before disease onset, CCR2+ macrophages harboring mSOD1 infiltrated sciatic nerves earlier than the lumbar cord. CCR2-deficient mSOD1-Tg mice showed an earlier onset and axonal derangement in the sciatic nerve than CCR2-positive mSOD1-Tg mice. CCR2-deficient mSOD1-Tg mice showed an increase in deposited mSOD1 in the sciatic nerve compared with CCR2-positive mice. These findings suggest that CCR2+ and CX3CR1+ macrophages exert neuroprotective functions in mSOD1 ALS via mSOD1 clearance from the peripheral nerves.
Collapse
Affiliation(s)
- Wataru Shiraishi
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan ,grid.415432.50000 0004 0377 9814Department of Neurology, Kokura Memorial Hospital, Fukuoka, 802-8555 Japan
| | - Ryo Yamasaki
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Yu Hashimoto
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Senri Ko
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Yuko Kobayakawa
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Noriko Isobe
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Takuya Matsushita
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Jun-ichi Kira
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan ,grid.411731.10000 0004 0531 3030Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy At Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Ookawa, Fukuoka 831-8501 Japan ,grid.411731.10000 0004 0531 3030Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, 2-6-11 Yakuin, Chuo-ku, Fukuoka, 810-0022 Japan
| |
Collapse
|
10
|
Hachemi Y, Rapp AE, Lee S, Dorn AK, Krüger BT, Kaiser K, Ignatius A, Tuckermann J. Intact Glucocorticoid Receptor Dimerization Is Deleterious in Trauma-Induced Impaired Fracture Healing. Front Immunol 2021; 11:628287. [PMID: 33679723 PMCID: PMC7927427 DOI: 10.3389/fimmu.2020.628287] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/29/2020] [Indexed: 11/13/2022] Open
Abstract
Following severe trauma, fracture healing is impaired because of overwhelming systemic and local inflammation. Glucocorticoids (GCs), acting via the glucocorticoid receptor (GR), influence fracture healing by modulating the trauma-induced immune response. GR dimerization-dependent gene regulation is essential for the anti-inflammatory effects of GCs. Therefore, we investigated in a murine trauma model of combined femur fracture and thoracic trauma, whether effective GR dimerization influences the pathomechanisms of trauma-induced compromised fracture healing. To this end, we used mice with decreased GR dimerization ability (GRdim). The healing process was analyzed by cytokine/chemokine multiplex analysis, flow cytometry, gene-expression analysis, histomorphometry, micro-computed tomography, and biomechanical testing. GRdim mice did not display a systemic or local hyper-inflammation upon combined fracture and thorax trauma. Strikingly, we discovered that GRdim mice were protected from fracture healing impairment induced by the additional thorax trauma. Collectively and in contrast to previous studies describing the beneficial effects of intact GR dimerization in inflammatory models, we report here an adverse role of intact GR dimerization in trauma-induced compromised fracture healing.
Collapse
Affiliation(s)
- Yasmine Hachemi
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Anna E Rapp
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Sooyeon Lee
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Ann-Kristin Dorn
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Benjamin T Krüger
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Kathrin Kaiser
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| |
Collapse
|
11
|
Forsythoside A inhibits adhesion and migration of monocytes to type II alveolar epithelial cells in lipopolysaccharide-induced acute lung injury through upregulating miR-124. Toxicol Appl Pharmacol 2020; 407:115252. [PMID: 32987027 DOI: 10.1016/j.taap.2020.115252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
Abstract
Acute lung injury (ALI) is a severe disease for which effective drugs are still lacking at present. Forsythia suspensa is a traditional Chinese medicine commonly used to relieve respiratory symptoms in China, but its functional mechanisms remain unclear. Therefore, forsythoside A (FA), the active constituent of F. suspensa, was studied in the present study. Inflammation models of type II alveolar epithelial MLE-12 cells and BALB/c mice stimulated by lipopolysaccharide (LPS) were established to explore the effects of FA on ALI and the underlying mechanisms. We found that FA inhibited the production of monocyte chemoattractant protein-1 (MCP-1/CCL2) in LPS-stimulated MLE-12 cells in a dose-dependent manner. Moreover, FA decreased the adhesion and migration of monocytes to MLE-12 cells. Furthermore, miR-124 expression was upregulated after FA treatment. The luciferase report assay showed that miR-124 mimic reduced the activity of CCL2 in MLE-12 cells. However, the inhibitory effects of FA on CCL2 expression and monocyte adhesion and migration to MLE-12 cells were counteracted by treatment with a miR-124 inhibitor. Critically, FA ameliorated LPS-induced pathological damage, decreased the serum levels of tumor necrosis factor-α and interleukin-6, and inhibited CCL2 secretion and macrophage infiltration in lungs in ALI mice. Meanwhile, administration of miR-124 inhibitor attenuated the protective effects of FA. The present study suggests that FA attenuates LPS-induced adhesion and migration of monocytes to type II alveolar epithelial cells though upregulating miR-124, thereby inhibiting the expression of CCL2. These findings indicate that the potential application of FA is promising and that miR-124 mimics could also be used in the treatment of ALI.
Collapse
|
12
|
Suresh MV, Dolgachev VA, Zhang B, Balijepalli S, Swamy S, Mooliyil J, Kralovich G, Thomas B, Machado-Aranda D, Karmakar M, Lalwani S, Subramanian A, Anantharam A, Moore BB, Raghavendran K. TLR3 absence confers increased survival with improved macrophage activity against pneumonia. JCI Insight 2019; 4:131195. [PMID: 31801911 DOI: 10.1172/jci.insight.131195] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 10/17/2019] [Indexed: 12/14/2022] Open
Abstract
Toll-like receptor 3 (TLR3) is a pathogen recognition molecule associated with viral infection with double-stranded RNA (dsRNA) as its ligand. We evaluated the role of TLR3 in bacterial pneumonia using Klebsiella pneumoniae (KP). WT and TLR3-/- mice were subjected to a lethal model of KP. Alveolar macrophage polarization, bactericidal activity, and phagocytic capacity were compared. RNA-sequencing was performed on alveolar macrophages from the WT and TLR3-/- mice. Adoptive transfers of alveolar macrophages from TLR3-/- mice to WT mice with KP were evaluated for survival. Expression of TLR3 in postmortem human lung samples from patients who died from gram-negative pneumonia and pathological grading of pneumonitis was determined. Mortality was significantly lower in TLR3-/-, and survival improved in WT mice following antibody neutralization of TLR3 and with TLR3/dsRNA complex inhibitor. Alveolar macrophages from TLR3-/- mice demonstrated increased bactericidal and phagocytic capacity. RNA-sequencing showed an increased production of chemokines in TLR3-/- mice. Adoptive transfer of alveolar macrophages from the TLR3-/- mice restored the survival in WT mice. Human lung samples demonstrated a good correlation between the grade of pneumonitis and TLR3 expression. These data represent a paradigm shift in understanding the mechanistic role of TLR3 in bacterial pneumonia.
Collapse
Affiliation(s)
| | | | - Boya Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Samantha Swamy
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Jashitha Mooliyil
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Georgia Kralovich
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Bivin Thomas
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Monita Karmakar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Sanjeev Lalwani
- Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Arulselvi Subramanian
- Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, India
| | | | - Bethany B Moore
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
13
|
Suresh MV, Balijepalli S, Zhang B, Singh VV, Swamy S, Panicker S, Dolgachev VA, Subramanian C, Ramakrishnan SK, Thomas B, Rao TC, Delano MJ, Machado-Aranda D, Shah YM, Raghavendran K. Hypoxia-Inducible Factor (HIF)-1α Promotes Inflammation and Injury Following Aspiration-Induced Lung Injury in Mice. Shock 2019; 52:612-621. [PMID: 30601332 PMCID: PMC6591116 DOI: 10.1097/shk.0000000000001312] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acid aspiration-induced lung injury is a common disease in the intensive care unit (ICU) and acute respiratory distress syndrome (ARDS). Hypoxia-inducible factor (HIF)-1α is a major transcription factor responsible for regulating the cellular response to changes in oxygen tension. A clear understanding of the function of HIF-1α in lung inflammatory response is currently lacking. Here, we sought to determine the role of HIF-1α in type 2 alveolar epithelial cells (AEC) in the generation of the acute inflammatory response following gastric aspiration (GA). GA led to profound hypoxia at very early time points following GA. This correlated to a robust increase in HIF-1α, tissue albumin and pro-inflammatory mediators following GA in AECs. The extent of lung injury and the release of pro/anti-inflammatory cytokines were significantly reduced in HIF-1α (-/-) mice. Finally, we report that HIF-1α upregulation of the acute inflammatory response is dependent on NF-κB following GA.
Collapse
Affiliation(s)
| | | | - Boya Zhang
- Department of Surgery, University of Michigan, Ann Arbor
| | | | - Samantha Swamy
- Department of Surgery, University of Michigan, Ann Arbor
| | | | | | | | | | - Bivin Thomas
- Department of Surgery, University of Michigan, Ann Arbor
| | - Tejeshwar C. Rao
- Department of Cell, Development and Integrative Biology, University of Alabama at Birmingham, Birmingham
| | | | | | - Yatrik M. Shah
- Molecular & Integrative Physiology, University of Michigan, Ann Arbor
| | | |
Collapse
|
14
|
Wang S, Liu F, Tan KS, Ser HL, Tan LTH, Lee LH, Tan W. Effect of (R)-salbutamol on the switch of phenotype and metabolic pattern in LPS-induced macrophage cells. J Cell Mol Med 2019; 24:722-736. [PMID: 31680470 PMCID: PMC6933346 DOI: 10.1111/jcmm.14780] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/05/2019] [Accepted: 08/04/2019] [Indexed: 12/21/2022] Open
Abstract
Evidence demonstrates that M1 macrophage polarization promotes inflammatory disease. Here, we discovered that (R)‐salbutamol, a β2 receptor agonist, inhibits and reprograms the cellular metabolism of RAW264.7 macrophages. (R)‐salbutamol significantly inhibited LPS‐induced M1 macrophage polarization and downregulated expressions of typical M1 macrophage cytokines, including monocyte chemotactic protein‐1 (MCP‐1), interleukin‐1β (IL‐1β) and tumour necrosis factor α (TNF‐α). Also, (R)‐salbutamol significantly decreased the production of inducible nitric oxide synthase (iNOS), nitric oxide (NO) and reactive oxygen species (ROS), while increasing the reduced glutathione (GSH)/oxidized glutathione (GSSG) ratio. In contrast, (S)‐salbutamol increased the production of NO and ROS. Bioenergetic profiles showed that (R)‐salbutamol significantly reduced aerobic glycolysis and enhanced mitochondrial respiration. Untargeted metabolomics analysis demonstrated that (R)‐salbutamol modulated metabolic pathways, of which three metabolic pathways, namely, (a) phenylalanine metabolism, (b) the pentose phosphate pathway and (c) glycerophospholipid metabolism were the most noticeably impacted pathways. The effects of (R)‐salbutamol on M1 polarization were inhibited by a specific β2 receptor antagonist, ICI‐118551. These findings demonstrated that (R)‐salbutamol inhibits the M1 phenotype by downregulating aerobic glycolysis and glycerophospholipid metabolism, which may propose (R)‐salbutamol as the major pharmacologically active component of racemic salbutamol for the treatment of inflammatory diseases and highlight the medicinal value of (R)‐salbutamol.
Collapse
Affiliation(s)
- Shanping Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Fei Liu
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Keai Sinn Tan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Hooi-Leng Ser
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.,Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Loh Teng-Hern Tan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.,Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Wen Tan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.,Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
15
|
Zhang B, Swamy S, Balijepalli S, Panicker S, Mooliyil J, Sherman MA, Parkkinen J, Raghavendran K, Suresh MV. Direct pulmonary delivery of solubilized curcumin reduces severity of lethal pneumonia. FASEB J 2019; 33:13294-13309. [PMID: 31530014 DOI: 10.1096/fj.201901047rr] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute respiratory distress syndrome (ARDS), the most severe form of acute lung injury, is associated with reduced lung compliance and hypoxemia. Curcumin exhibits potent anti-inflammatory properties but has poor solubility and rapid plasma clearance. To overcome these physiochemical limitations and uncover the full therapeutic potential of curcumin in lung inflammation, in this study we utilized a novel water-soluble curcumin formulation (CDC) and delivered it directly into the lungs of C57BL/6 mice inoculated with a lethal dose of Klebsiella pneumoniae (KP). Administration of CDC led to a significant reduction in mortality, in bacterial presence within blood and lungs, as well as in lung injury, inflammation, and oxidative stress. The expression of Klebsiella hemolysin gene; TNF-α; IFN-β; nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3; hypoxia-inducible factor 1/2α; and NF-κB were also decreased following CDC treatment, suggesting modulation of the inflammasome complex and hypoxia signaling pathways as an underlying mechanism by which CDC reduces the severity of pneumonia. On a cellular level, CDC led to diminished cell death, improved viability, and protection of human lung epithelial cells in vitro. Overall, our studies demonstrate that CDC administration improves cell survival and reduces injury, inflammation, and mortality in a murine model of lethal gram-negative pneumonia. CDC, therefore, has promising anti-inflammatory potential in pneumonia and likely other inflammatory lung diseases, demonstrating the importance of optimizing the physicochemical properties of active natural products to optimize their clinical application.-Zhang, B., Swamy, S., Balijepalli, S., Panicker, S., Mooliyil, J., Sherman, M. A., Parkkinen, J., Raghavendran, K., Suresh, M. V. Direct pulmonary delivery of solubilized curcumin reduces severity of lethal pneumonia.
Collapse
Affiliation(s)
- Boya Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Samantha Swamy
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Sreehari Panicker
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Jashitha Mooliyil
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew A Sherman
- Department of Pediatrics, Children's National Medical Center, Washington, DC, USA
| | - Jaakko Parkkinen
- Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | | | | |
Collapse
|
16
|
Molecular Characterization of Hypoxic Alveolar Epithelial Cells After Lung Contusion Indicates an Important Role for HIF-1α. Ann Surg 2019; 267:382-391. [PMID: 27811509 DOI: 10.1097/sla.0000000000002070] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To understand the fate and regulation of hypoxic type II alveolar epithelial cells (AECs) after lung contusion (LC). BACKGROUND LC due to thoracic trauma is a major risk factor for the development of acute respiratory distress syndrome. AECs have recently been implicated as a primary driver of inflammation in LC. The main pathological consequence of LC is hypoxia, and a key mediator of adaptation to hypoxia is hypoxia-inducible factor (HIF)-1. We have recently published that HIF-1α is a major driver of acute inflammation after LC through type II AEC. METHODS LC was induced in wild-type mice (C57BL/6), luciferase-based hypoxia reporter mice (ODD-Luc), and HIF-1α conditional knockout mice. The degree of hypoxia was assessed using hypoxyprobe and in vivo imaging system. The fate of hypoxic AEC was evaluated by luciferase dual staining with caspases-3 and Ki-67, terminal deoxynucleotidyl transferase dUTP nick end labeling, and flow cytometry with ApoStat. NLRP-3 expression was determined by western blot. Laser capture microdissection was used to isolate AECs in vivo, and collected RNA was analyzed by Q-PCR for HIF-related pathways. RESULTS Global hypoxia was present after LC, but hypoxic foci were not uniform. Hypoxic AECs preferentially undergo apoptosis. There were significant reductions in NLRP-3 in HIF-1α conditional knockout mice. The expression of proteins involved in HIF-related pathways and inflammasome activation were significantly increased in hypoxic AECs. CONCLUSIONS These are the first in vivo data to identify, isolate, and characterize hypoxic AECs. HIF-1α regulation through hypoxic AECs is critical to the initiation of acute inflammation after LC.
Collapse
|
17
|
|
18
|
Dolgachev V, Panicker S, Balijepalli S, McCandless LK, Yin Y, Swamy S, Suresh MV, Delano MJ, Hemmila MR, Raghavendran K, Machado-Aranda D. Electroporation-mediated delivery of FER gene enhances innate immune response and improves survival in a murine model of pneumonia. Gene Ther 2018; 25:359-375. [PMID: 29907877 PMCID: PMC6195832 DOI: 10.1038/s41434-018-0022-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/02/2018] [Accepted: 05/11/2018] [Indexed: 12/23/2022]
Abstract
Previously, we reported that electroporation-mediated (EP) delivery of the FER gene improved survival in a combined trauma-pneumonia model. The mechanism of this protective effect is unknown. In this paper, we performed a pneumonia model in C57/BL6 mice with 500 CFU of Klebsiella pneumoniae. After inoculation, a plasmid encoding human FER was delivered by EP into the lung (PNA/pFER-EP). Survival of FER-treated vs. controls (PNA; PNA/EP-pcDNA) was recorded. In parallel cohorts, bronchial alveolar lavage (BAL) and lung were harvested at 24 and 72 h with markers of infection measured. FER-EP-treated animals reduced bacterial counts and had better 5-day survival compared to controls (80 vs. 20 vs. 25%; p < 0.05). Pre-treatment resulted in 100% survival. With FER, inflammatory monocytes were quickly recruited into BAL. These cells had increased surface expression for Toll-receptor 2 and 4, and increased phagocytic and myeloperoxidase activity at 24 h. Samples from FER electroporated animals had increased phosphorylation of STAT transcription factors, varied gene expression of IL1β, TNFα, Nrf2, Nlrp3, Cxcl2, HSP90 and increased cytokine production of TNF-α, CCL-2, KC, IFN-γ, and IL-1RA. In a follow-up experiment, using Methicillin-resistant Staphylococcus aureus (MRSA) similar bacterial reduction effects were obtained with FER gene delivery. We conclude that FER overexpression improves survival through STAT activation enhancing innate immunity and accelerating bacterial clearance in the lung. This constitutes a novel mechanism of inflammatory regulation with therapeutic potential in the setting of hospital-acquired pneumonia.
Collapse
Affiliation(s)
- Vladislav Dolgachev
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Sreehari Panicker
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Sanjay Balijepalli
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Lane Kelly McCandless
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Yue Yin
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Samantha Swamy
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - M V Suresh
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Matthew J Delano
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Mark R Hemmila
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - Krishnan Raghavendran
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA
| | - David Machado-Aranda
- Division of Acute Care Surgery, University of Michigan, 1500 E Medical Center Dr, UH-1C421, SPC 5033, Ann Arbor, MI, 48109-5033, USA.
| |
Collapse
|
19
|
Machado-Aranda D. [The Use Of Pulmonary Gene Therapy In The Treatment Of Experimental Models Of Pneumonia And Septicemia]. GACETA MEDICA DE CARACAS 2018; 126:5-14. [PMID: 30100668 PMCID: PMC6086359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Affiliation(s)
- David Machado-Aranda
- Laboratorio del Estudio de la Biología y Terapia Molecular para el Manejo del Trauma Pulmonar
- División de Cirugía de Trauma, Quemados y Urgencias - Terapia Intensiva Quirúrgica, Universidad de Michigan, Ann Arbor, Michigan, Estados Unidos de América
| |
Collapse
|
20
|
Toll-Like Receptor-9 (TLR9) is Requisite for Acute Inflammatory Response and Injury Following Lung Contusion. Shock 2018; 46:412-9. [PMID: 26939039 DOI: 10.1097/shk.0000000000000601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lung contusion (LC) is a significant risk factor for the development of acute respiratory distress syndrome. Toll-like receptor 9 (TLR9) recognizes specific unmethylated CpG motifs, which are prevalent in microbial but not vertebrate genomic DNA, leading to innate and acquired immune responses. TLR9 signaling has recently been implicated as a critical component of the inflammatory response following lung injury. The aim of the present study was to evaluate the contribution of TLR9 signaling to the acute physiologic changes following LC. Nonlethal unilateral closed-chest LC was induced in TLR9 (-/-) and wild-type (WT) mice. The mice were sacrificed at 5, 24, 48, and 72-h time points. The extent of injury was assessed by measuring bronchoalveolar lavage, cells (cytospin), albumin (permeability injury), and cytokines (inflammation). Following LC, only the TLR9 (-/-) mice showed significant reductions in the levels of albumin; release of pro-inflammatory cytokines IL-1β, IL-6, and Keratinocyte chemoattractant; production of macrophage chemoattractant protein 5; and recruitment of alveolar macrophages and neutrophil infiltration. Histological evaluation demonstrated significantly worse injury at all-time points for WT mice. Macrophages, isolated from TLR9 (-/-) mice, exhibited increased phagocytic activity at 24 h after LC compared with those isolated from WT mice. TLR9, therefore, appears to be functionally important in the development of progressive lung injury and inflammation following LC. Our findings provide a new framework for understanding the pathogenesis of lung injury and suggest blockade of TLR9 as a new therapeutic strategy for the treatment of LC-induced lung injury.
Collapse
|
21
|
Hypercapnic Conditions After Experimental Blunt Chest Trauma Increase Efferocytosis of Alveolar Macrophages and Reduce Local Inflammation. Shock 2017; 48:104-111. [DOI: 10.1097/shk.0000000000000813] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
McGrath-Morrow SA, Ndeh R, Collaco JM, Poupore AK, Dikeman D, Zhong Q, Singer BD, D'Alessio F, Scott A. The innate immune response to lower respiratory tract E. Coli infection and the role of the CCL2-CCR2 axis in neonatal mice. Cytokine 2017. [PMID: 28628889 DOI: 10.1016/j.cyto.2017.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Neonates have greater morbidity/mortality from lower respiratory tract infections (LRTI) compared to older children. Lack of conditioning of the pulmonary immune system due to limited environmental exposures and/or infectious challenges likely contributes to the increase susceptibility in the neonate. In this study, we sought to gain insights into the nature and dynamics of the neonatal pulmonary immune response to LRTI using a murine model. METHODS Wildtype (WT) and Ccr2-/- C57BL/6 neonatal and juvenile mice received E. coli or PBS by direct pharyngeal aspiration. Flow cytometry was used to measure immune cell dynamics and identify cytokine-producing cells. Real-time PCR and ELISA were used to measure cytokine/chemokine expression. RESULTS Innate immune cell recruitment in response to E. coli-induced LRTI was delayed in the neonatal lung compared to juvenile lung. Lung clearance of bacteria was also significantly delayed in the neonate. Ccr2-/- neonates, which lack an intact CCL2-CCR2 axis, had higher mortality after E. coli challenged than Ccr2+/+ neonates. A greater percentage of CD8+ T cells and monocytes from WT neonates challenged with E. coli produced TNF compared to controls. CONCLUSION The pulmonary immune response to E. coli-induced LRTI differed significantly between neonatal and juvenile mice. Neonates were more susceptible to increasing doses of E. coli and exhibited greater mortality than juveniles. In the absence of an intact CCL2-CCR2 axis, susceptibility to LRTI-induced mortality was further increased in neonatal mice. Taken together these findings underscore the importance of age-related differences in the innate immune response to LRTI during early stages of postnatal life.
Collapse
Affiliation(s)
- Sharon A McGrath-Morrow
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| | - Roland Ndeh
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Joseph M Collaco
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Amy K Poupore
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dustin Dikeman
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Qiong Zhong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, United States
| | - Benjamin D Singer
- Northwestern University Feinberg School of Medicine, Medicine, Chicago, IL, United States
| | - Franco D'Alessio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, United States
| | - Alan Scott
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
23
|
Sharpe JP, Khan NR, Chatterjee AR, Huang J, Magnotti LJ, Croce MA, Fabian TC. Investigating Cyclooxygenase Inhibition in a Rat Pulmonary Contusion Model: A Laboratory Study Finding No Improvement with Ibuprofen. Am Surg 2017. [DOI: 10.1177/000313481708300635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Minimal advances have been made in the management of pulmonary contusions (PCs). The purpose of this study was to evaluate the impact of cyclooxygenase inhibition on outcomes following PC in a rat model. PC was induced in anesthetized adult rats. Ibuprofen was given to the treatment group (TG) and water was given to the control group (CG). Lung injury was assessed with pulse oximetry, arterial blood gases, CT, and histopathologic examination. Inflammation was measured with both serum and bronchoalveolar lavage (BAL) levels of tumor necrosis factor a and interleukin-6. Rats in the TG did not differ from rats in the CG with respect to oxygenation. Pathologic examination demonstrated a trend toward more inflammatory infiltrate in the CG, yet the sizes of the contusions were larger in the TG. The CG trended toward decreased levels of interleukin-6 in the serum and BAL at both three and seven days. While BAL levels of tumor necrosis factor a were increased in the TG at three days compared to the CG, they trended toward a reduced amount at seven days. Our data do not support cyclooxygenase inhibition for treatment to decrease the respiratory compromise associated with PC in this model of rat PCs.
Collapse
Affiliation(s)
- John P. Sharpe
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee and Department of Radiology
| | - Nick R. Khan
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee and Department of Radiology
| | | | - Jinsong Huang
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee and Department of Radiology
| | - Louis J. Magnotti
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee and Department of Radiology
| | - Martin A. Croce
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee and Department of Radiology
| | - Timothy C. Fabian
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee and Department of Radiology
| |
Collapse
|
24
|
Double-Stranded RNA Interacts With Toll-Like Receptor 3 in Driving the Acute Inflammatory Response Following Lung Contusion. Crit Care Med 2017; 44:e1054-e1066. [PMID: 27509390 DOI: 10.1097/ccm.0000000000001879] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVES Lung contusion is a major risk factor for the development of acute respiratory distress syndrome. We set to determine the role of toll-like receptor 3 and the binding of double-stranded RNA in the pathogenesis of sterile injury following lung contusion. DESIGN Toll-like receptor 3 expression was analyzed in postmortem lung samples from patients with lung contusion. Unilateral lung contusion was induced in toll-like receptor 3 (-/-), TIR-domain-containing adapter-inducing interferon-β (-/-), and wild-type mice. Subsequently, lung injury and inflammation were evaluated. Apoptotic indices, phagocytic activity, and phenotypic characterization of the macrophages were determined. Double-stranded RNA in bronchoalveolar lavage and serum samples following lung contusion was measured. A toll-like receptor 3/double-stranded RNA ligand inhibitor was injected into wild-type mice prior to lung contusion. MEASUREMENTS AND MAIN RESULTS Toll-like receptor 3 expression was higher in patients and wild-type mice with lung contusion. The degree of lung injury, inflammation, and macrophage apoptosis was reduced in toll-like receptor 3 (-/-), TIR-domain-containing adapter-inducing interferon-β (-/-), and wild-type mice with toll-like receptor 3 antibody neutralization. Alveolar macrophages from toll-like receptor 3 (-/-) mice had a lower early apoptotic index, a predominant M2 phenotype and increased surface translocation of toll-like receptor 3 from the endosome to the surface. When compared with viral activation pathways, lung injury in lung contusion demonstrated increased p38 mitogen-activated protein kinases, extracellular signal-regulated kinase 1/2 phosphorylation with inflammasome activation without a corresponding increase in nuclear factor-κB or type-1 interferon production. Additionally, pretreatment with toll-like receptor 3/double-stranded RNA ligand inhibitor led to a reduction in injury, inflammation, and macrophage apoptosis. CONCLUSIONS We conclude that the interaction of double-stranded RNA from injured cells with toll-like receptor 3 drives the acute inflammatory response following lung contusion.
Collapse
|
25
|
Obi AT, Andraska E, Kanthi Y, Luke CE, Elfline M, Madathilparambil S, Siahaan TJ, Jaffer FA, Wakefield TW, Raghavendran K, Henke PK. Gram-Negative Pneumonia Alters Large-Vein Cell-Adhesion Molecule Profile and Potentiates Experimental Stasis Venous Thrombosis. J Vasc Res 2016; 53:186-195. [PMID: 27771726 DOI: 10.1159/000447299] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/28/2016] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND/AIMS Pneumonia is a significant risk factor for the development of venous thrombosis (VT). Cell-adhesion molecules (CAMs) are linked to the pathogenesis of both pneumonia and VT. We hypothesized that remote infection would confer a prothrombogenic milieu via systemic elevation of CAMs. METHODS Lung injury was induced in wild-type (C57BL/6) mice by lung contusion or intratracheal inoculation with Klebsiella pneumoniae or saline controls. K. pneumoniae-treated mice and controls additionally underwent inferior vena cava (IVC) ligation to generate VT. RESULTS Lung-contusion mice demonstrated no increase in E-selectin or P-selectin whereas mice infected with K. pneumoniae demonstrated increased circulating P-selectin, ICAM-1, VCAM-1 and thrombin-antithrombin (TAT) complexes. Mice with pneumonia formed VT 3 times larger than controls, demonstrated significantly more upregulation of vein-wall and systemic CAMs, and formed erythrocyte-rich thrombi. CONCLUSION Elevated CAM expression was identified in mice with pneumonia, but not lung contusion, indicating that the type of inflammatory stimulus and the presence of infection drive the vein-wall response. Elevation of CAMs was associated with amplified VT and may represent an alternate mechanism by which to target the prevention of VT.
Collapse
Affiliation(s)
- Andrea T Obi
- Conrad Jobst Vascular Research Laboratory, University of Michigan Medical School, Ann Arbor, Mich., USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Electroporation-mediated delivery of the FER gene in the resolution of trauma-related fatal pneumonia. Gene Ther 2016; 23:785-796. [PMID: 27454317 PMCID: PMC5096957 DOI: 10.1038/gt.2016.58] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/19/2016] [Accepted: 07/11/2016] [Indexed: 12/18/2022]
Abstract
Injured patients with lung contusion (LC) are at risk of developing bacterial pneumonia (PNA) followed by sepsis and death. A recent genome-wide association study (GWAS) showed FER gene expression positively correlating with survival rates among individuals with above conditions. We sought to determine whether electroporation (EP)-mediated delivery of FER gene could indeed improve survival, in a lethal model of combined LC and PNA. C57BL/6 mice sustained unilateral LC, which preceded a 500 Klebsiella colony forming unit (CFU) inoculation by 6 h. In-between these insults, human FER plasmid (pFER) was introduced into the lungs followed by eight EP pulses applied externally (10 ms at 200 V cm-1). Control groups included EP of empty vector (pcDNA3) or Na+/K+-ATPase genes (pPump) and no treatment (LC+PNA). We recorded survival, histology, lung mechanics, bronchial alveolar lavage (BAL) fluid, FER and inflammatory gene expression and bacteriology. The data show that 7-day survival was significantly improved by pFER compared with control groups. pFER increased BAL monocytes and activated antibacterial response genes (nitric oxide synthase (NOS), Fizz). pFER treatment showed decreased lung and blood Klebsiella counts reaching, in some cases, complete sterilization. In conclusion, FER gene delivery promoted survival in LC+PNA mice via recruitment of activated immune cells, improving efficiency of bacterial clearance within contused lung.
Collapse
|
27
|
Chan JK, Glass GE, Ersek A, Freidin A, Williams GA, Gowers K, Espirito Santo AI, Jeffery R, Otto WR, Poulsom R, Feldmann M, Rankin SM, Horwood NJ, Nanchahal J. Low-dose TNF augments fracture healing in normal and osteoporotic bone by up-regulating the innate immune response. EMBO Mol Med 2016; 7:547-61. [PMID: 25770819 PMCID: PMC4492816 DOI: 10.15252/emmm.201404487] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which trauma initiates healing remains unclear. Precise understanding of these events may define interventions for accelerating healing that could be translated to the clinical arena. We previously reported that addition of low-dose recombinant human TNF (rhTNF) at the fracture site augmented fracture repair in a murine tibial fracture model. Here, we show that local rhTNF treatment is only effective when administered within 24 h of injury, when neutrophils are the major inflammatory cell infiltrate. Systemic administration of anti-TNF impaired fracture healing. Addition of rhTNF enhanced neutrophil recruitment and promoted recruitment of monocytes through CCL2 production. Conversely, depletion of neutrophils or inhibition of the chemokine receptor CCR2 resulted in significantly impaired fracture healing. Fragility, or osteoporotic, fractures represent a major medical problem as they are associated with permanent disability and premature death. Using a murine model of fragility fractures, we found that local rhTNF treatment improved fracture healing during the early phase of repair. If translated clinically, this promotion of fracture healing would reduce the morbidity and mortality associated with delayed patient mobilization.
Collapse
Affiliation(s)
- James K Chan
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Graeme E Glass
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Adel Ersek
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Freidin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Garry A Williams
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Kate Gowers
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Rosemary Jeffery
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - William R Otto
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Richard Poulsom
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sara M Rankin
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | |
Collapse
|
28
|
Nogo-B protects mice against lipopolysaccharide-induced acute lung injury. Sci Rep 2015; 5:12061. [PMID: 26174362 PMCID: PMC4502524 DOI: 10.1038/srep12061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/09/2015] [Indexed: 12/11/2022] Open
Abstract
Nogo-B, a member of the reticulon 4 protein family, plays a critical role in tissue repair and acute inflammation. Its role in acute lung injury (ALI) remains unclear. Here, we assessed the function of Nogo-B during tissue injury in a lipopolysaccharide (LPS)-induced ALI mouse model. We found that pulmonary Nogo-B was significantly repressed after LPS instillation in C57BL/6 mice. Over-expression of pulmonary Nogo-B using an adenovirus vector carrying the Nogo-B-RFP-3flag gene (Ad-Nogo-B) significantly prolonged the survival of mice challenged with a lethal dose of LPS. The Ad-Nogo-B-treated mice also had less severe lung injury, less alveolar protein exudation, and a higher number of macrophages but less neutrophil infiltration compared with Ad-RFP-treated mice. Interestingly, microarray analysis showed that the Ad-Nogo-B-treated mice had different gene expression profiles compared with the controls and the prominent expression of genes related to wound healing and the humoral immune response after LPS induction. Of the 49 differently expressed genes, we found that the expression of PTX3 was significantly up-regulated following Nogo-B over-expression as observed in lung tissues and RAW264.7 cells. In conclusion, Nogo-B plays a protective role against LPS-induced ALI, and this effect might be exerted through the modulation of alveolar macrophage recruitment and PTX3 production.
Collapse
|
29
|
Interleukin 10 overexpression alters survival in the setting of gram-negative pneumonia following lung contusion. Shock 2014; 41:301-10. [PMID: 24430542 DOI: 10.1097/shk.0000000000000123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Lung contusion injury produces a vulnerable window within the inflammatory defenses of the lung that predisposes the patient to pneumonia. Interleukin 10 (IL-10) is a known anti-inflammatory mediator produced by macrophages and capable of downregulating acute lung inflammation. We investigated the impact of increased levels of IL-10 within the lung on survival and the host response to trauma in the setting of lung contusion (LC) and gram-negative pneumonia. DESIGN A bitransgenic, tetracycline-inducible, lung-specific human IL-10 overexpression (IL-10 OE) mouse model and single transgenic (TG-) control mice were used. Mice underwent LC injury or sham injury (sham) at time -6 h. At time 0, animals were inoculated intratracheally with 500 colony-forming units of Klebsiella pneumoniae (pneu). Bronchoalveolar lavage fluid, lung tissue specimens, or purified macrophages were collected. Lung tissue and blood bacteria levels were quantified. Cytokine levels were assayed by enzyme-linked immunosorbent assay, and gene expression levels were evaluated by real-time polymerase chain reaction. Cell-type identification and quantification were done using real-time polymerase chain reaction and flow cytometry. MAIN RESULTS Interleukin 10 OE mice demonstrated decreased 5-day survival compared with TG- mice following LC + pneu (0 vs. 30%, P < 0.0001). Interleukin 10 OE mice had significantly higher lung bacteria counts (P = 0.02) and levels of bacteremia (P = 0.001) at 24 h. The IL-10 OE mice recruited more neutrophils into the alveoli as measured in bronchoalveolar lavage fluid compared with TG- mice. Alveolar macrophages from IL-10 OE mice displayed increased alternative activation (M2 macrophages, P = 0.046), whereas macrophages from TG- mice exhibited classic activation (M1 macrophages) and much higher intracellular bacterial killing potential (P = 0.03). Interleukin 6, keratinocyte-derived chemokine, and macrophage inflammatory protein 2 levels were significantly elevated in IL-10 OE LC + pneu animals (P < 0.05). CONCLUSIONS Lung-specific IL-10 overexpression induces alternative activation of alveolar macrophages. This shift in macrophage phenotype decreases intracellular bacterial killing, resulting in a more pronounced bacteremia and accelerated mortality in a model of LC and pneumonia.
Collapse
|
30
|
Suresh MV, Ramakrishnan SK, Thomas B, Machado-Aranda D, Bi Y, Talarico N, Anderson E, Yatrik SM, Raghavendran K. Activation of hypoxia-inducible factor-1α in type 2 alveolar epithelial cell is a major driver of acute inflammation following lung contusion. Crit Care Med 2014; 42:e642-53. [PMID: 25014067 DOI: 10.1097/ccm.0000000000000488] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Lung contusion is a major risk factor for the development of acute respiratory distress syndrome. Hypoxia-inducible factor-1α is the primary transcription factor that is responsible for regulating the cellular response to changes in oxygen tension. We set to determine if hypoxia-inducible factor-1α plays a role in the pathogenesis of acute inflammatory response and injury in lung contusion. DESIGN Nonlethal closed-chest unilateral lung contusion was induced in a hypoxia reporter mouse model and type 2 cell-specific hypoxia-inducible factor-1α conditional knockout mice. The mice were killed at 5-, 24-, 48-, and 72-hour time points, and the extent of systemic and tissue hypoxia was assessed. In addition, injury and inflammation were assessed by measuring bronchoalveolar lavage cells (flow cytometry and cytospin), albumin (permeability injury), and cytokines (inflammation). Isolated type 2 cells from the hypoxia-inducible factor-1α conditional knockout mice were isolated and evaluated for proinflammatory cytokines following lung contusion. Finally, the role of nuclear factor-κB and interleukin-1β as intermediates in this interaction was studied. RESULTS Lung contusion induced profound global hypoxia rapidly. Increased expression of hypoxia-inducible factor-1α from lung samples was observed as early as 60 minutes, following the insult. The extent of lung injury following lung contusion was significantly reduced in conditional knockout mice at all the time points, when compared with the wild-type littermate mice. Release of proinflammatory cytokines, such as interleukin-1β, interleukin-6, macrophage inflammatory protein-2, and keratinocyte chemoattractant, was significantly lower in conditional knockout mice. These actions are in part mediated through nuclear factor-κB. Hypoxia-inducible factor-1α in lung epithelial cells was shown to regulate interleukin-1β promoter activity. CONCLUSION Activation of hypoxia-inducible factor-1α in type 2 cell is a major driver of acute inflammation following lung contusion.
Collapse
Affiliation(s)
- Madathilparambil V Suresh
- 1Department of Surgery, University of Michigan Medical School, Ann Arbor, MI. 2Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI. 3Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Alveolar macrophage depletion increases the severity of acute inflammation following nonlethal unilateral lung contusion in mice. J Trauma Acute Care Surg 2014; 76:982-90. [PMID: 24662861 DOI: 10.1097/ta.0000000000000163] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Lung contusion (LC) is a common injury resulting from blunt thoracic trauma. LC is an important risk factor for the development acute lung injury, adult respiratory distress syndrome, and ventilator-associated pneumonia, all of which increase mortality from trauma. LC produces a nonspecific immune cellular response. Neutrophil recruitment is known to increase the severity of inflammation during LC. However, the exact role of macrophages in modulating the response to LC has not been well described. METHODS We used a cortical contusion impactor to induce unilateral LC in mice. Thoracic micro computed tomographic scans of these animals were obtained to document radiologic changes over time following LC. To understand the role of macrophages during LC, liposomal clodronate was used to deplete macrophage levels before traumatic insult. Acute inflammatory attributes after LC were assessed, by measuring pressure-volume mechanics; quantifying bronchial alveolar lavage levels of leukocytes, albumin, and cytokines; and finally examining lung specimen histopathology at 5, 24, 48, and 72 hours after injury. RESULTS After LC, alveolar macrophage numbers were significantly reduced and exhibited slowed recovery. Simultaneously, there was a significant increase in bronchial alveolar lavage neutrophil counts. The loss of macrophages could be attributed to both cellular apoptosis and necrosis. Pretreatment with clodronate increased the severity of lung inflammation as measured by worsened pulmonary compliance, increased lung permeability, amplification of neutrophil recruitment, and increases in early proinflammatory cytokine levels. CONCLUSION The presence of regulatory alveolar macrophages plays an important role in the pathogenesis of acute inflammation following LC.
Collapse
|
32
|
Ozbek C, Kestelli M, Bozok S, Ilhan G, Yurekli I, Ozpak B, Akyuz M, Bademci M. Surgical stimulation of angiogenesis. Asian Cardiovasc Thorac Ann 2014; 22:36-9. [PMID: 24585641 DOI: 10.1177/0218492312468285] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The results of surgical approaches are unsatisfactory in patients with a distal arterial bed that is ineligible for revascularization. In this retrospective study, we investigated outcomes in patients who underwent surgical interventions that are reported to induce angiogenesis. METHOD 6 patients diagnosed with thromboangiitis obliterans were included in this study. Of these 6 patients, 2 underwent femoropopliteal bypass surgery using reversed great saphenous vein as a conduit. The other 4 underwent ascending venous arterialization (bypass from the popliteal artery to the great saphenous vein at the level of medial malleolus, using reversed great saphenous vein). RESULTS During the early postoperative period, all of the reversed saphenous vein grafts were occluded. The mean postoperative ankle-brachial index increased from 0.33 to 0.83. During the postoperative period, intermittent claudication disappeared in all patients. Angiograms taken during the postoperative period showed evidence of neovascularization when compared to those taken during preoperative evaluation. CONCLUSION Wound healing is an inflammatory process that simultaneously activates angiogenesis. We propose that the improved ankle-brachial index values and neovascularization shown in our patients were associated with this inflammatory process.
Collapse
Affiliation(s)
- Cengiz Ozbek
- Department of Cardiovascular Surgery, Izmir Ataturk Training and Research Hospital, Izmir, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Machado-Aranda D, Raghavendran K. Electroporation-mediated delivery of genes in rodent models of lung contusion. Methods Mol Biol 2014; 1121:205-21. [PMID: 24510825 DOI: 10.1007/978-1-4614-9632-8_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Several of the biological processes involved in the pathogenesis of acute lung injury and acute respiratory distress syndrome after lung contusion are regulated at a genetic and epigenetic level. Thus, strategies to manipulate gene expression in this context are highly desirable not only to elucidate the mechanisms involved but also to look for potential therapies. In the present chapter, we describe mouse and rat models of inducing blunt thoracic injury followed by electroporation-mediated gene delivery to the lung. Electroporation is a highly efficient and easily reproducible technique that allows circumvention of several of lung gene delivery challenges and safety issues present with other forms of lung gene therapy.
Collapse
Affiliation(s)
- David Machado-Aranda
- Division of Acute Care Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
34
|
The protective role of MnTBAP in oxidant-mediated injury and inflammation in a rat model of lung contusion. Surgery 2013; 154:980-90. [PMID: 24139490 DOI: 10.1016/j.surg.2013.05.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/10/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Lung contusion (LC) is a unique direct and focal insult that is considered a major risk factor for the initiation of acute lung injury and acute respiratory distress syndrome. We have shown recently that consumption of nitric oxide (due to excess superoxide) resulting in peroxynitrite formation leads to decreased vascular reactivity after LC. In this study, we set out to determine whether the superoxide scavenger Mn (III) tetrakis (4-benzoic acid) porphyrin chloride (MnTBAP) plays a protective role in alleviating acute inflammatory response and injury in LC. METHODS Nonlethal, closed-chest, bilateral LC was induced in a rodent model. Administration of the superoxide dismutase mimetic MnTBAP concurrently in LC in rats was performed, and bronchoalveolar lavage (BAL) and lung samples were analyzed for degree of injury and inflammation at 5 and 24 h after the insult. The extent of injury was assessed by the measurement of cells and albumin with cytokine levels in the BAL and lungs. Lung samples were subjected to H&E and superoxide staining with dihydro-ethidium. Protein-bound dityrosine and nitrotyrosine levels were quantified in lung tissue by tandem mass spectrometry. RESULTS The degrees of lung injury after LC as determined by BAL albumin levels were significantly decreased in the MnTBAP-administered rats at all the time points when compared to the corresponding controls. The release of proinflammatory cytokines and BAL neutrophils was significantly less in the rats administered MnTBAP after LC. Administration of MnTBAP decreased tissue damage and decreased necrosis and neutrophil-rich exudate at the 24-h time point. Staining for superoxide anions showed significantly greater intensity in the lung samples from the LC group compared to the LC+ MnTBAP group. High-performance liquid chromatography/tandem mass spectrometry revealed that MnTBAP treatment significantly attenuated dityrosine and nitrotyrosine levels, consistent with decreased oxidant injury. CONCLUSION Superoxide dismutase mimetic-MnTBAP reduced permeability and oxidative injury in LC and may have a therapeutic role in diminishing inflammation in LC.
Collapse
|
35
|
Foskett AM, Bazhanov N, Ti X, Tiblow A, Bartosh TJ, Prockop DJ. Phase-directed therapy: TSG-6 targeted to early inflammation improves bleomycin-injured lungs. Am J Physiol Lung Cell Mol Physiol 2013; 306:L120-31. [PMID: 24242012 DOI: 10.1152/ajplung.00240.2013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Previous reports demonstrated that bleomycin-induced injury of lungs in mice can be improved by the administration of murine multipotent adult stem/progenitor cells (MSCs) from the bone marrow. Recently some of the beneficial effects of MSCs have been explained by the cells being activated by signals from injured tissues to express the inflammation modulating protein TNF-α-stimulated gene/protein 6 (TSG-6). In this study, we elected to test the hypothesis that targeting the early phase of bleomycin-induced lung injury with systemic TSG-6 administration may produce therapeutic effects such as preventing the deterioration of lung function and increasing survival by modulation of the inflammatory cascade. Lung injury in C57Bl/6J mice was induced by intratracheal administration of bleomycin. Mice then received intravenous injections of TSG-6 or sham controls. Pulse oximetry was used to monitor changes in lung function. Cell infiltration was evaluated by flow cytometry, cytokine expression was measured by ELISA assays, and lungs were assessed for histological attributes. The results demonstrated that intravenous infusion of TSG-6 during the early inflammatory phase decreased cellular infiltration into alveolar spaces. Most importantly, it improved both the subsequent decrease in arterial oxygen saturation levels and the survival of the mice. These findings demonstrated that the beneficial effects of TSG-6 in a model of bleomycin-induced lung injury are largely explained by the protein modulating the early inflammatory phase. Similar phase-directed strategy with TSG-6 or other therapeutic factors that MSCs produce may be useful for other lung diseases and diseases of other organs.
Collapse
Affiliation(s)
- Andrea M Foskett
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott and White, 5701 Airport Rd., Module C, Temple, TX 76502.
| | | | | | | | | | | |
Collapse
|
36
|
Nemzek-Hamlin JA, Hwang H, Hampel JA, Yu B, Raghavendran K. Development of a murine model of blunt hepatic trauma. Comp Med 2013; 63:398-408. [PMID: 24210016 PMCID: PMC3796750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/25/2013] [Accepted: 04/15/2013] [Indexed: 06/02/2023]
Abstract
Despite the prevalence of blunt hepatic trauma in humans, there are few rodent models of blunt trauma that can be used to study the associated inflammatory responses. We present a mouse model of blunt hepatic trauma that was created by using a cortical contusion device. Male mice were anesthetized with ketamine-xylazine-buprenorphine and placed in left lateral recumbency. A position of 2 mm ventral to the posterior axillary line and 5 mm caudal to the costal margin on the right side was targeted for impact. An impact velocity of 6 m/s and a piston depth of 12 mm produced a consistent pattern of hepatic injury with low mortality. All mice that recovered from anesthesia survived without complication for the length of the study. Mice were euthanized at various time points (n = 5 per group) until 7 d after injury for gross examination and collection of blood and peritoneal lavage fluids. Some mice were reanesthetized for serial monitoring of hepatic lesions via MRI. At 2 h after trauma, mice consistently displayed laceration, hematoma, and discoloration of the right lateral and caudate liver lobes, with intraabdominal hemorrhage but no other gross injuries. Blood and peritoneal lavage fluid were collected from all mice for cytokine analysis. At 2 h after trauma, there were significant increases in plasma IL10 as well as peritoneal lavage fluid IL6 and CXCL1/KC; however, these levels decreased within 24 h. At 7 d after trauma, the mice had regained body weight, and the hepatic lesions, which initially had increased in size during the first 48 h, had returned to their original size. In summary, this technique produced a reliable, low mortality, murine model that recreates features of blunt abdominal liver injury in human subjects with similar acute inflammatory response.
Collapse
Affiliation(s)
- Jean A Nemzek-Hamlin
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | | | | | |
Collapse
|
37
|
Ndisang JF, Mishra M. The heme oxygenase system selectively suppresses the proinflammatory macrophage m1 phenotype and potentiates insulin signaling in spontaneously hypertensive rats. Am J Hypertens 2013; 26:1123-31. [PMID: 23757400 DOI: 10.1093/ajh/hpt082] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The mechanisms by which heme oxygenase (HO) improves glucose metabolism in essential hypertension are not completely understood. Because dysfunctional insulin signaling is associated with elevated inflammation and high cholesterol and triglycerides, we investigated the effects of HO on the proinflammatory macrophage M1 phenotype and the anti-inflammatory macrophage M2 phenotype in spontaneously hypertensive rats (SHRs). SHRs are a model of human essential hypertension with features of metabolic syndrome, including impaired glucose metabolism. METHODS Spectrophotometric analysis, enzyme immunoassay, enzyme-linked immunosorbent assay, and Western immunoblotting were used. HO was enhanced with hemin or inhibited with chromium-mesoporphyrin (CrMP). RESULTS Hemin suppressed inflammation by abating proinflammatory macro phage M1 phenotype (ED1) and chemokines such as macrophage chemoattractant protein 1 (MCP-1) and macrophage inflammatory protein 1 alpha (MIP-1α) while enhancing anti-inflammatory macrophage M2 phenotype by potentiating ED2, CD206, and CD14. Similarly, hemin improved insulin signaling by enhancing insulin receptor substrate 1 (IRS-1), IRS-2, phosphatidylinositol 3 kinase (PI3K), and glucose transporter 4 (GLUT4) but reduced total cholesterol and triglycerides. These effects were accompanied by increased HO-1, HO activity, and cyclic guanosine monophosphate (cGMP), whereas the HO inhibitor CrMP nullified the hemin effects. Importantly, the effects of the HO system on ED1, ED2, CD206, and CD14 in SHRs are novel. CONCLUSIONS Hemin abated inflammation in SHRs by selectively enhancing the anti-inflammatory macrophage M2 phenotype that dampens inflammation while suppressing the pronflammatory macrophage M1 phenotype and related chemokines such as MCP-1 and MIP-1α. Importantly, the reduction of inflammation, total cholesterol, and triglycerides was accompanied by the enhancement of important proteins implicated in insulin signaling, including IRS-1, IRS-2, PI3K, and GLUT4. Thus, the concomitant reduction of inflammation, total cholesterol and triglycerides and the corresponding potentiation of insulin signaling are among the multifaceted mechanisms by which the HO system improves glucose metabolism in essential hypertension.
Collapse
Affiliation(s)
- Joseph Fomusi Ndisang
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | |
Collapse
|
38
|
Role of pulmonary artery reactivity and nitric oxide in injury and inflammation following lung contusion. Shock 2013; 39:278-85. [PMID: 23364426 DOI: 10.1097/shk.0b013e318281d6ed] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mechanisms contributing to hypoxia in lung contusion (LC) remain unclear and not temporally associated with the peak onset of acute inflammation. We investigated the role of oxidative stress in alteration of pulmonary arterial (PA) reactivity following LC. In addition, the role of antioxidants in reversing this process was examined. PaO2 and PA reactivity were measured in rats subjected to bilateral LC. Rings were pretreated with a nitric oxide synthase (NOS) inhibitor, L-nitro arginine (10(-3) M), or PEG-superoxide dismutase (SOD) and PEG-catalase (CAT), or both (L-nitro arginine + SOD/CAT). Rings were constricted with norepinephrine and relaxed with an NOS agonist (A23187) or NO donor (SNAP [S-nitrosyl amino penicillamine]). Immunochemical and mass spectrometric quantification for nitrotyrosine was performed. Rats were hypoxemic at 4 h after contusion compared with controls, but recovered by 24 h (PaO(2)/FIO(2) ratio: baseline, 443 ± 28; 4 h, 288 ± 46; and 24 h, 417 ± 23). Pulmonary arterial constriction to NOS inhibition and relaxation to A23187 were impaired 4 h after LC. Pulmonary arterial relaxation to SNAP was decreased at 4 and 24 h after LC. These alterations in PA reactivity were reversed by SOD/CAT pretreatment. SOD1 and 2 mRNA were upregulated, and soluble guanylyl cyclase mRNA was downregulated 24 h after LC. Immunohistochemistry and mass spectrometry revealed that levels of 3-nitrotyrosine were increased markedly at 4 h following LC consistent with superoxide generation and formation of peroxynitrite. Collectively, these data suggest that consumption of NO due to excess superoxide resulting in peroxynitrite formation leads to diminished vascular reactivity following LC.
Collapse
|