1
|
Guo B, Liu W, Ji X, Xi B, Meng X, Xie W, Sun Y, Zhang M, Liu P, Zhang W, Yan X, Chen B. CSF3 aggravates acute exacerbation of pulmonary fibrosis by disrupting alveolar epithelial barrier integrity. Int Immunopharmacol 2024; 135:112322. [PMID: 38788452 DOI: 10.1016/j.intimp.2024.112322] [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] [Received: 03/14/2024] [Revised: 05/03/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive respiratory disorder characterized by poor prognosis, often presenting with acute exacerbation. The primary cause of death associated with IPF is acute exacerbation of IPF (AE-IPF). However, the pathophysiology of acute exacerbation has not been clearly elucidated yet. This study aims to investigate the underlying pathophysiological molecular mechanism in a mouse AE-PF model. C57BL/6J mice were intratracheally administered bleomycin (BLM, 5 mg/kg) to induce pulmonary fibrosis. After 14 days, lipopolysaccharide (LPS, 2 mg/kg) was injected via the trachea route. Histological assessments, including H&E and Masson staining, as well as inflammatory indicators, were included to evaluate the induction of AE-PF by BLM and LPS in mice. Transcriptomic profiling of pulmonary tissues identified CSF3 as one of the top 10 upregulated DEGs in AE-PF mice. Indeed, administration of exogenous CSF3 protein exacerbated AE-PF in mice. Mechanistically, CSF3 disrupted alveolar epithelial barrier integrity and permeability by regulating specialized cell adhesion complexes such as tight junctions (TJs) and adherens junctions (AJs) via PI3K/p-Akt/Snail pathway, contributing to the aggravation of AE-PF in mice. Moreover, the discovery of elevated sera CSF3 indicated a notable increase in IPF patients during the exacerbation of the disease. Pearson correlation analysis in IPF patients revealed significant positive associations between CSF3 levels and KL-6 levels, LDH levels, CRP levels, respectively. These results provide mechanistic insights into the role of CSF3 in exacerbating of lung fibrotic disease and indicate monitoring CSF3 levels may aid in early clinical decisions for alternative therapy in the management of rapidly progressing IPF.
Collapse
Affiliation(s)
- Bingnan Guo
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Department of Emergency Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Wenwen Liu
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xuan Ji
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China; Department of Respiratory Medicine, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, China
| | - Bin Xi
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xiao Meng
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Wanwan Xie
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Yitian Sun
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Maowei Zhang
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Pingli Liu
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Wenhui Zhang
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xianliang Yan
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Department of Emergency Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China; Department of Emergency Medicine, Suining People's Hospital, Xuzhou 221225, Jiangsu, China.
| | - Bi Chen
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Xuzhou Medical University, Department of Respiratory Medicine, School of First Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China.
| |
Collapse
|
2
|
Su JX, Li SJ, Zhou XF, Zhang ZJ, Yan Y, Liu SL, Qi Q. Chemotherapy-induced metastasis: molecular mechanisms and clinical therapies. Acta Pharmacol Sin 2023; 44:1725-1736. [PMID: 37169853 PMCID: PMC10462662 DOI: 10.1038/s41401-023-01093-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Chemotherapy, the most widely accepted treatment for malignant tumors, is dependent on cell death induced by various drugs including antimetabolites, alkylating agents, mitotic spindle inhibitors, antitumor antibiotics, and hormonal anticancer drugs. In addition to causing side effects due to non-selective cytotoxicity, chemotherapeutic drugs can initiate and promote metastasis, which greatly reduces their clinical efficacy. The knowledge of how they induce metastasis is essential for developing strategies that improve the outcomes of chemotherapy. Herein, we summarize the recent findings on chemotherapy-induced metastasis and discuss the underlying mechanisms including tumor-initiating cell expansion, the epithelial-mesenchymal transition, extracellular vesicle involvement, and tumor microenvironment alterations. In addition, the use of combination treatments to overcome chemotherapy-induced metastasis is also elaborated.
Collapse
Affiliation(s)
- Jin-Xuan Su
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Si-Jia Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiao-Feng Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhi-Jing Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yu Yan
- Functional Experimental Teaching Center, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Song-Lin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
3
|
Ólafsdóttir HS, Dalqvist E, Onjukka E, Klevebro F, Nilsson M, Gagliardi G, Alexandersson von Döbeln G. Postoperative complications after esophagectomy for cancer, neoadjuvant chemoradiotherapy compared to neoadjuvant chemotherapy: A single institutional cohort study. Clin Transl Radiat Oncol 2023; 40:100610. [PMID: 36936472 PMCID: PMC10018434 DOI: 10.1016/j.ctro.2023.100610] [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: 12/17/2022] [Revised: 02/22/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023] Open
Abstract
Background Complications after esophagectomy are common and the possible increase in postoperative complications associated with neoadjuvant chemoradiotherapy is of concern. The aim of our study was to analyze if the addition of radiotherapy to neoadjuvant chemotherapy increases the incidence and severity of postoperative complications, including evaluation of the relation between radiation doses to the heart and lungs and postoperative complications. Methods The study was based on an institutional surgical database for esophageal cancer. The study period was October 2008 to March 2020. Patients treated with neoadjuvant chemoradiotherapy were compared to patients treated with neoadjuvant chemotherapy and dose/volume parameters for the lungs and heart considered. The primary outcome was 30-day postoperative complications. Results During the study period, 274 patients underwent surgery for esophageal cancer, 93 patients after neoadjuvant chemotherapy and 181 patients after neoadjuvant chemoradiotherapy. The median prescribed radiation dose to the planning target volume was 41.4 Gy, the median of the mean lung dose was 6.2 Gy, and the median of the mean heart dose was 20.3 Gy. The addition of radiotherapy to neoadjuvant chemotherapy did not increase the incidence of postoperative complications. Neither were radiation doses to the lungs and heart associated with postoperative complications. Taxane-based chemotherapy regimens were however associated with an increased incidence of postoperative complications. Conclusions In our cohort, the addition of neoadjuvant radiotherapy to chemotherapy was not associated with postoperative complications. However, taxane-based chemotherapy regimens, with or without concomitant radiotherapy, were associated with postoperative complications.
Collapse
Affiliation(s)
- Halla Sif Ólafsdóttir
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-141 52 Huddinge, Sweden
- Department of Radiotherapy, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, SE-171 64 Solna, Sweden
- Corresponding author at: Department of Radiotherapy, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Anna Steckséns gata 41, SE-171 76 Stockholm, Sweden.
| | - Emmy Dalqvist
- Section of Radiotherapy Physics and Engineering, Department of Medical Radiation Physics and Nuclear Medicine, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, SE-171 64 Solna, Sweden
| | - Eva Onjukka
- Section of Radiotherapy Physics and Engineering, Department of Medical Radiation Physics and Nuclear Medicine, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, SE-171 64 Solna, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64 Solna, Sweden
| | - Fredrik Klevebro
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-141 52 Huddinge, Sweden
- Department of Upper Abdominal Diseases, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, SE-141 57 Huddinge, Sweden
| | - Magnus Nilsson
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-141 52 Huddinge, Sweden
- Department of Upper Abdominal Diseases, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, SE-141 57 Huddinge, Sweden
| | - Giovanna Gagliardi
- Section of Radiotherapy Physics and Engineering, Department of Medical Radiation Physics and Nuclear Medicine, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, SE-171 64 Solna, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 64 Solna, Sweden
| | - Gabriella Alexandersson von Döbeln
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-141 52 Huddinge, Sweden
- Medical Unit of Head, Neck, Lung and Skin Cancer, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, SE-171 64 Solna, Sweden
| |
Collapse
|
4
|
Natural Taxanes: From Plant Composition to Human Pharmacology and Toxicity. Int J Mol Sci 2022; 23:ijms232415619. [PMID: 36555256 PMCID: PMC9779243 DOI: 10.3390/ijms232415619] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Biologically active taxanes, present in small- to medium-sized evergreen conifers of various Taxus species, are widely used for their antioxidant, antimicrobial and anti-inflammatory effects, but mostly for their antitumour effects used in the treatment of solid tumours of the breast, ovary, lung, bladder, prostate, oesophagus and melanoma. More of the substances found in Taxus plant extracts have medical potential. Therefore, at the beginning of this review, we describe the methods of isolation, identification and determination of taxanes in different plant parts. One of the most important taxanes is paclitaxel, for which we summarize the pharmacokinetic parameters of its different formulations. We also describe toxicological risks during clinical therapy such as hypersensitivity, neurotoxicity, gastrointestinal, cardiovascular, haematological, skin and renal toxicity and toxicity to the respiratory system. Since the effect of the drug-form PTX is enhanced by various Taxus spp. extracts, we summarize published clinical intoxications and all fatal poisonings for the Taxus baccata plant. This showed that, despite their significant use in anticancer treatment, attention should also be focused on the risk of fatal intoxication due to ingestion of extracts from these plants, which are commonly found in our surroundings.
Collapse
|
5
|
Mohan N, Dalip D, Rampersad FS, Jaggernauth S. Paclitaxel-Induced Pneumonitis in Trinidad: A Case Report. Cureus 2022; 14:e26613. [PMID: 35949737 PMCID: PMC9357421 DOI: 10.7759/cureus.26613] [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] [Accepted: 07/06/2022] [Indexed: 11/22/2022] Open
Abstract
Paclitaxel-induced pneumonitis (PIP) is an immune-mediated disease resulting from a delayed hypersensitivity reaction (type IV) to paclitaxel, an anti-microtubule chemotherapeutic drug commonly used to treat breast cancer in both neoadjuvant and adjuvant settings. PIP is diagnosed by exclusion utilizing laboratory work-up, imaging, biopsy studies, and results of antibiotic therapy because there is no single diagnostic test. Ground-glass opacifications on CT, coupled with minimal restrictive disturbance with decreased diffusion on pulmonary function tests (PFTs), negative bronchoalveolar lavage (BAL), and bronchoscopy cultures, may assist physicians in diagnosing paclitaxel-induced pneumonitis. In this report, we describe a case of PIP present in Trinidad, West Indies, which has not been described previously in this region.
Collapse
|
6
|
Wu DM, He M, Zhao YY, Deng SH, Liu T, Zhang T, Zhang F, Wang YY, Xu Y. Increased susceptibility of irradiated mice to Aspergillus fumigatus infection via NLRP3/GSDMD pathway in pulmonary bronchial epithelia. Cell Commun Signal 2022; 20:98. [PMID: 35761358 PMCID: PMC9238178 DOI: 10.1186/s12964-022-00907-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022] Open
Abstract
Background Aspergillus fumigatus infection is difficult to diagnose clinically and can develop into invasive pulmonary aspergillosis, which has a high fatality rate. The incidence of Aspergillus fumigatus infection has increased die to widespread application of radiotherapy technology. However, knowledge regarding A. fumigatus infection following radiation exposure is limited, and the underlying mechanism remains unclear. In this study, we established a mouse model to explore the effect of radiation on A. fumigatus infection and the associated mechanisms. Methods In this study, a mouse model of A. fumigatus infection after radiation was established by irradiating with 5 Gy on the chest and instilling 5 × 107/ml Aspergillus fumigatus conidia into trachea after 24 h to explore the effect and study its function and mechanism. Mice were compared among the following groups: normal controls (CON), radiation only (RA), infection only (Af), and radiation + infection (RA + Af). Staining analyses were used to detect infection and damage in lung tissues. Changes in protein and mRNA levels of pyroptosis-related molecules were assessed by western blot analysis and quantitative reverse transcription polymerase chain reaction, respectively. Protein concentrations in the serum and alveolar lavage fluid were also measured. An immunofluorescence colocalization analysis was performed to confirm that NLRP3 inflammasomes activated pyroptosis. Results Radiation destroyed the pulmonary epithelial barrier and significantly increased the pulmonary fungal burden of A. fumigatus. The active end of caspase-1 and gasdermin D (GSDMD) were highly expressed even after infection. Release of interleukin-18 (IL-18) and interleukin-1β (IL-1β) provided further evidence of pyroptosis. NLRP3 knockout inhibited pyroptosis, which effectively attenuated damage to the pulmonary epithelial barrier and reduced the burden of A. fumigatus. Conclusions Our findings indicated that the activation of NLRP3 inflammasomes following radiation exposure increased susceptibility to A. fumigatus infection. Due to pyroptosis in lung epithelial cells, it resulted in the destruction of the lung epithelial barrier and further damage to lung tissue. Moreover, we found that NLRP3 knockout effectively inhibited the pyroptosis and reducing susceptibility to A. fumigatus infection and further lung damage. Overall, our results suggest that NLRP3/GSDMD pathway mediated-pyroptosis in the lungs may be a key event in this process and provide new insights into the underlying mechanism of infection. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00907-2.
Collapse
Affiliation(s)
- Dong-Ming Wu
- School of Clinical Medicine, Chengdu Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Miao He
- School of Clinical Medicine, Chengdu Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Yang-Yang Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Shi-Hua Deng
- School of Clinical Medicine, Chengdu Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Teng Liu
- School of Clinical Medicine, Chengdu Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Ting Zhang
- School of Clinical Medicine, Chengdu Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Feng Zhang
- School of Clinical Medicine, Chengdu Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Yuan-Yi Wang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Ying Xu
- School of Clinical Medicine, Chengdu Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
7
|
Combination of ruthenium (II) polypyridyl complex Δ-Ru1 and Taxol enhances the anti-cancer effect on Taxol-resistant cancer cells through Caspase-1/GSDMD-mediated pyroptosis. J Inorg Biochem 2022; 230:111749. [DOI: 10.1016/j.jinorgbio.2022.111749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 12/12/2022]
|
8
|
Nelin LD, Jin Y, Chen B, Liu Y, Rogers LK, Reese J. Cyclooxygenase-2 deficiency attenuates lipopolysaccharide-induced inflammation, apoptosis, and acute lung injury in adult mice. Am J Physiol Regul Integr Comp Physiol 2022; 322:R126-R135. [PMID: 34984926 PMCID: PMC9829472 DOI: 10.1152/ajpregu.00140.2021] [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] [Received: 06/01/2021] [Revised: 10/20/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023]
Abstract
Many lung diseases are caused by an excessive inflammatory response, and inflammatory lung diseases are often modeled using lipopolysaccharide (LPS) in mice. Cyclooxygenase-2 (COX-2) encoded by the Ptgs2 gene is induced in response to inflammatory stimuli including LPS. The objective of this study was to test the hypothesis that mice deficient in COX-2 (Ptgs2-/-) will be protected from LPS-induced lung injury. Wild-type (WT; CD1 mice) and Ptgs2-/- mice (on a CD1 background) were treated with LPS or vehicle for 24 h. LPS treatment resulted in histological evidence of lung injury, which was attenuated in the Ptgs2-/- mice. LPS treatment increased the mRNA levels for tumor necrosis factor-α, interleukin-10, and monocyte chemoattractant protein-1 in the lungs of WT mice, and the LPS-induced increases in these levels were attenuated in the Ptgs2-/- mice. The protein levels of active caspase-3 and caspase-9 were lower in the LPS-treated lungs of Ptgs2-/- mice than in LPS-treated WT mice, as were the number of terminal deoxynucleotide transferase dUTP nick end labeling-positive cells in lung sections. LPS exposure resulted in a greater lung wet-to-dry weight ratio (W/D) in WT mice, suggestive of pulmonary edema, while in LPS-treated Ptgs2-/- mice, the W/D was not different from controls and less than in LPS-treated WT mice. These results demonstrate that COX-2 is involved in the inflammatory response to LPS and suggest that COX-2 not only acts as a downstream participant in the inflammatory response, but also acts as a regulator of the inflammatory response likely through a feed-forward mechanism following LPS stimulation.
Collapse
Affiliation(s)
- Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Lynette K Rogers
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Jeff Reese
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
9
|
Karki P, Birukova AA. Microtubules as Major Regulators of Endothelial Function: Implication for Lung Injury. Front Physiol 2021; 12:758313. [PMID: 34777018 PMCID: PMC8582326 DOI: 10.3389/fphys.2021.758313] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/06/2021] [Indexed: 01/04/2023] Open
Abstract
Endothelial dysfunction has been attributed as one of the major complications in COVID-19 patients, a global pandemic that has already caused over 4 million deaths worldwide. The dysfunction of endothelial barrier is characterized by an increase in endothelial permeability and inflammatory responses, and has even broader implications in the pathogenesis of acute respiratory syndromes such as ARDS, sepsis and chronic illnesses represented by pulmonary arterial hypertension and interstitial lung disease. The structural integrity of endothelial barrier is maintained by cytoskeleton elements, cell-substrate focal adhesion and adhesive cell junctions. Agonist-mediated changes in endothelial permeability are directly associated with reorganization of actomyosin cytoskeleton leading to cell contraction and opening of intercellular gaps or enhancement of cortical actin cytoskeleton associated with strengthening of endothelial barrier. The role of actin cytoskeleton remodeling in endothelial barrier regulation has taken the central stage, but the impact of microtubules in this process remains less explored and under-appreciated. This review will summarize the current knowledge on the crosstalk between microtubules dynamics and actin cytoskeleton remodeling, describe the signaling mechanisms mediating this crosstalk, discuss epigenetic regulation of microtubules stability and its nexus with endothelial barrier maintenance, and overview a role of microtubules in targeted delivery of signaling molecules regulating endothelial permeability and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
10
|
Middleton JD, Sivakumar S, Hai T. Chemotherapy-Induced Changes in the Lung Microenvironment: The Role of MMP-2 in Facilitating Intravascular Arrest of Breast Cancer Cells. Int J Mol Sci 2021; 22:10280. [PMID: 34638621 PMCID: PMC8508901 DOI: 10.3390/ijms221910280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 11/18/2022] Open
Abstract
Previously, we showed that mice treated with cyclophosphamide (CTX) 4 days before intravenous injection of breast cancer cells had more cancer cells in the lung at 3 h after cancer injection than control counterparts without CTX. At 4 days after its injection, CTX is already excreted from the mice, allowing this pre-treatment design to reveal how CTX may modify the lung environment to indirectly affect cancer cells. In this study, we tested the hypothesis that the increase in cancer cell abundance at 3 h by CTX is due to an increase in the adhesiveness of vascular wall for cancer cells. Our data from protein array analysis and inhibition approach combined with in vitro and in vivo assays support the following two-prong mechanism. (1) CTX increases vascular permeability, resulting in the exposure of the basement membrane (BM). (2) CTX increases the level of matrix metalloproteinase-2 (MMP-2) in mouse serum, which remodels the BM and is functionally important for CTX to increase cancer abundance at this early stage. The combined effect of these two processes is the increased accessibility of critical protein domains in the BM, resulting in higher vascular adhesiveness for cancer cells to adhere. The critical protein domains in the vascular microenvironment are RGD and YISGR domains, whose known binding partners on cancer cells are integrin dimers and laminin receptor, respectively.
Collapse
Affiliation(s)
- Justin D. Middleton
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210, USA
| | - Subhakeertana Sivakumar
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
| | - Tsonwin Hai
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
11
|
Corso CR, Mulinari Turin de Oliveira N, Maria-Ferreira D. Susceptibility to SARS-CoV-2 infection in patients undergoing chemotherapy and radiation therapy. J Infect Public Health 2021; 14:766-771. [PMID: 34022735 PMCID: PMC7980522 DOI: 10.1016/j.jiph.2021.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/22/2021] [Accepted: 03/14/2021] [Indexed: 01/08/2023] Open
Abstract
The outbreak of the new coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly become a public health emergency of international concern, especially affecting the elderly people and patients with chronic disease, such as hypertension and respiratory syndromes. Patients undergoing chemotherapy treatment (e.g., bleomycin, cyclophosphamide, methotrexate, monoclonal antibodies, and paclitaxel therapy) are vulnerable to the development of respiratory syndromes induced by chemotherapeutic agents and are also more susceptible to viral infections as they are immunosuppressed. Neutropenia is an important risk factor for increased vulnerability to infections, as a respiratory syndrome involves an array of immune cells maintaining the balance between pathogen clearance and immunopathology. However, the differential diagnosis of pulmonary symptoms in cancer patients is broad, with complications being related to the malignancy itself, treatment toxicity, and infections. The risk factors depend on the specific type of cancer, chemotherapy, patient characteristics, and comorbidities. Thus, this review discusses the main events implicated in immunosuppression caused by chemotherapy and radiation therapy and the association of immunosuppression and other factors with SARS-CoV-2 infection susceptibility in cancer patients; and, importantly, how to deal with this situation in face of the current pandemic scenario.
Collapse
Affiliation(s)
- Claudia Rita Corso
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil.
| |
Collapse
|
12
|
Radiotherapy challenges in COVID era. BIOMEDICAL ENGINEERING TOOLS FOR MANAGEMENT FOR PATIENTS WITH COVID-19 2021. [PMCID: PMC8192315 DOI: 10.1016/b978-0-12-824473-9.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The pandemic caused by the new coronavirus (SARS-CoV-2) associated with a disease named COVID-19 by the World Health Organization that began in late 2019 in Wuhan city has become a global public health problem. Only 2 months later, the new virus affected most countries of the world, the consequence being an overload of health systems, especially Intensive Care Units. Considered a category of patients at high risk of developing severe forms of the disease, cancer patients can develop a severe form of the disease, complicated by acute respiratory distress syndrome requiring mechanical ventilation. Radiotherapy, as a treatment included in the multidisciplinary management of cancer for both curative and palliative purposes, is also affected by the COVID-19 pandemic. COVID-19-positive or -suspected patients are a special category for which the decision to postpone treatment should be made based on the particularities of tumor biology and the radiobiological effect of a gap in radiation fractions delivery. Emergencies including spinal cord compressions, tumor bleeding, and brain metastases not responsive to corticosteroid treatment, should be considered a priority but the palliative treatment should be limited from one single fraction to maximum five fractions for spinal cord compression and whole brain radiotherapy. Radiotherapy for brain metastases does not bring a benefit in terms of overall survival for patients with life expectancy of days or weeks and dexamethasone treatment is the correct choice in this situation. In all settings, the approach of radiotherapy treatment must be adapted for both scenarios of an outbreak pandemic, when general measures of social distancing and protection by specific equipment of patients and radiotherapy staff are a priority, but also for a long period of coexistence with the virus with possible new “pandemic waves.”
Collapse
|
13
|
Zhou F, Liu Z, Cai H, Miao Z, Wei F, Song C. Role of microRNA-15a-5p/TNFAIP3-interacting protein 2 axis in acute lung injury induced by traumatic hemorrhagic shock. Exp Ther Med 2020; 20:2. [PMID: 32934667 PMCID: PMC7471858 DOI: 10.3892/etm.2020.9130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to investigate the role of microRNA (miR)-15a-5p in the pathogenesis of acute lung injury induced by traumatic hemorrhagic shock (THS), and to explore the underlying molecular mechanism. The expression level of miR-15a-5p was detected using reverse transcription-quantitative (RT-qPCR) and the association between miR-15a-5p and TNFAIP3-interacting protein 2 (TNIP2) was revealed using TargetScan and dual luciferase reporter assays. To investigate the effect of miR-15a-5p on THS-induced acute lung injury, a THS rat model was established. Lung capillary permeability and lung edema were then determined. Moreover, proinflammatory factors in the bronchoalveolar lavage fluid (BALF) and serum of the THS rat model were detected using ELISA. In addition, protein levels in the current study were measured via western blotting. It was revealed that miR-15a-5p was significantly upregulated in both patients with THS and samples from the THS rat model. TNIP2 represents a direct target of miR-15a-5p, and it was downregulated in both patients with THS and the THS rat model. Further analyses indicated that downregulation of miR-15a-5p significantly relieved acute lung injury induced by THS, evidenced by a decreased ratio of Evan's blue dye (EBD) in the BALF to EBD in plasma of THS rats, decreased lung permeability index and reduced lung wet/dry ratio. Inhibition of miR-15a-5p also decreased THS-induced upregulation of pro-inflammatory factors. Furthermore, the data revealed that THS-induced NF-κB activation in the lung tissues of rats was inhibited by miR-15a-5p knockdown. Moreover, it was demonstrated that all the effects of miR-15a-5p on THS rats were ablated following TNIP2 silencing. Taken together, the data of the current study indicate that miR-15a-5p downregulation serves a protective role in THS-induced acute lung injury via directly targeting TNIP2.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Zhizhen Liu
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Zhenjun Miao
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Faxing Wei
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Chao Song
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
14
|
Zhao Z, He Z, Huang H, Chen J, He S, Yilihamu A, Nie Y. Drug-induced Interstitial Lung Disease in Breast Cancer Patients: A Lesson We Should Learn From Multi-Disciplinary Integration. BIO INTEGRATION 2020. [DOI: 10.15212/bioi-2020-0009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abstract Taxanes represented by paclitaxel and targeted therapy including trastuzumab are two common agents for human epidermal growth factor receptor-2 (HER-2)-positive breast cancer patients. Effectiveness, however, usually comes at the cost of many side effects, some of
which are even fatal. Drug-induced interstitial lung diseases (DILDs) comprise a group of drug-induced pulmonary injuries usually caused by using these medications. For DILDs, systemic therapy can be harmful to lung tissues and rapidly threaten the lives of some breast cancer patients. Through
the cases from our hospital and related studies in medical databases, we hope readers can learn a lesson from an angle of multi-disciplinary integration based on clinical practice and pharmacological mechanisms to make anti-cancer agents less harmful and reduce the incidence of DILD in breast
cancer patients during systemic therapy.
Collapse
Affiliation(s)
- Zijun Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, P.R. China
| | - Zhanghai He
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Hongyan Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, P.R. China
| | - Jiewen Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, P.R. China
| | - Shishi He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, P.R. China
| | - Ailifeire Yilihamu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, P.R. China
| | - Yan Nie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou 510120, P.R. China
| |
Collapse
|
15
|
Wang X, Liu M, Zhu MJ, Shi L, Liu L, Zhao YL, Cheng L, Gu YJ, Zhou MY, Chen L, Kumar A, Wang Y. Resveratrol protects the integrity of alveolar epithelial barrier via SIRT1/PTEN/p-Akt pathway in methamphetamine-induced chronic lung injury. Cell Prolif 2020; 53:e12773. [PMID: 32020692 PMCID: PMC7106965 DOI: 10.1111/cpr.12773] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/22/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
Objectives SIRT1 is an antioxidative factor, but its mechanism in methamphetamine (MA)‐induced lung injury remains unclear. The purpose of this study is to determine whether MA can disrupt the integrity of alveolar epithelial barrier, whether SIRT1 is involved in MA‐induced chronic lung injury and whether Resveratrol (Res) can protect the integrity of alveolar epithelial cells by regulating ROS to activate SIRT1/PTEN/p‐Akt pathway. Materials and methods The rats were randomly divided into control group and MA group. Extracted lungs were detected by Western blot, HE staining and immunohistochemistry. The alveolar epithelial cells were treated with MA or/and Res, following by Western blot, LDH leakage assay and flow cytometry. MOE is used for bio‐informatics prediction. Results Chronic exposure to MA can cause slower growth ratio of weight, increased RVI and induced lung injury including the reduced number of alveolar sacs and the thickened alveolar walls. MA‐induced apoptosis was associated with SIRT1‐related oxidative stress. Res suppressed ROS levels, activated SIRT1, negatively regulated PTEN, phosphorylated Akt, reduced LDH leakage, increased the expression of ZO‐1 and E‐cadherin and inhibited the apoptosis of alveolar epithelial cells to attenuate MA‐induced higher permeability of alveolar epithelium. Conclusions MA disrupted the integrity of alveolar epithelial barrier. Res inhibited oxidative stress and reversed MA‐induced higher permeability and apoptosis of alveolar epithelium by the activation of SIRT1/PTEN/p‐Akt pathway.
Collapse
Affiliation(s)
- Xin Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ming Liu
- Department of Drug Control, Criminal Investigation Police, University of China, Shenyang, China
| | - Mei-Jia Zhu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lin Shi
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lian Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yuan-Ling Zhao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lin Cheng
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ying-Jian Gu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ming-Yuan Zhou
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lei Chen
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ashok Kumar
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
16
|
Kawami M, Harada R, Ojima T, Yamagami Y, Yumoto R, Takano M. Association of cell cycle arrest with anticancer drug-induced epithelial-mesenchymal transition in alveolar epithelial cells. Toxicology 2019; 424:152231. [PMID: 31170432 DOI: 10.1016/j.tox.2019.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/28/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
Many drugs exert serious cytotoxic effects on pulmonary tissues. Although several reports have shown an association of epithelial-mesenchymal transition (EMT) with anticancer drug-induced lung injury, mechanisms of these effects are poorly understood. In the present study, we evaluated mechanisms of anticancer drug-induced EMT, with a focus on involvement of cell cycle arrest. We found that methotrexate (MTX) altered mRNA expression levels of many genes as determined by microarray analysis. Gene set enrichment analysis revealed that cell cycle arrest pathways may be associated with MTX-induced EMT. In addition, thymidine (THY) and nocodazole (NOC), which induce cell cycle arrest at S-phase and G2/M-phase, increased mRNA expression levels of α-smooth muscle actin (SMA), an EMT marker. Furthermore, α-SMA protein expression in cells arrested at S- and G2/M-phases by MTX and paclitaxel (PTX) was significantly higher than that in cells at G1. Notably, co-treatment of cells with THY or NOC and EMT-inducing anticancer drugs did not result in additional upregulation of α-SMA mRNA expression. These findings suggested that cell cycle arrest may be closely associated with anticancer drug-induced EMT in alveolar epithelial cells.
Collapse
Affiliation(s)
- Masashi Kawami
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Risako Harada
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Takamichi Ojima
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Yohei Yamagami
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Ryoko Yumoto
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Mikihisa Takano
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| |
Collapse
|
17
|
Teng Y, Feng C, Liu Y, Jin H, Gao Y, Li T. Anti-inflammatory effect of tranexamic acid against trauma-hemorrhagic shock-induced acute lung injury in rats. Exp Anim 2018; 67:313-320. [PMID: 29398669 PMCID: PMC6083028 DOI: 10.1538/expanim.17-0143] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
It has been demonstrated that tranexamic acid (TXA), a synthetic derivative of lysine,
alleviates lung damage in a trauma-hemorrhagic shock (T/HS) model. Nevertheless, the
mechanism of TXA against acute lung injury (ALI) has not deeply elaborated. In this study,
we generated a T/HS rat model based on previous research, and TXA (50 mg/kg and 100 mg/kg)
was intravenously injected into these rats prior to or post T/HS. The results revealed
that the decreased survival rate and impaired lung permeability of the rats caused by T/HS
were improved by TXA pretreatment or posttreatment. T/HS-triggered over-generation of
interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in bronchoalveolar fluid and
serum was inhibited by TXA, and the enzymatic activity of myeloperoxidase (MPO) in lung
tissues was suppressed by TXA as well. Furthermore, TXA treatment deactivated the poly
ADP-ribose polymerase-1 (PARP1)/nuclear factor κB (NF-κB) signaling pathway in the lungs
of T/HS rats, as evidenced by increased IκBα expression, and decreased cleaved PARP1,
p-p65 (Ser276), p-p65 (Ser529), p-IκBα (ser32/ser36), and intercellular adhesion
molecule-1. While the expression level of total p65 did not change after T/HS, its DNA
binding activity was strengthened. Both TXA pretreatment and posttreatment suppressed this
effect on the DNA binding activity of NF-κB. Taken together, our results reveal that
administration of TXA effectively relieves T/HS-induced ALI, at least in part, by
attenuating the abnormal pulmonary inflammation.
Collapse
Affiliation(s)
- Yue Teng
- Department of Emergency Medicine, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, P.R. China.,Department of Emergency Medicine, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang 110016, P.R. China
| | - Cong Feng
- Department of Emergency Medicine, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, P.R. China
| | - Yunen Liu
- Department of Emergency Medicine, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang 110016, P.R. China.,Laboratory of Rescue Center for Severe Wound and Trauma PLA, 83 Wenhua Road, Shenyang 110016, P.R. China.,Liaoning Key Laboratory of Severe Wound and Trauma and Organ Protection, 83 Wenhua Road, Shenyang 110016, P.R. China
| | - Hongxu Jin
- Department of Emergency Medicine, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang 110016, P.R. China
| | - Yan Gao
- Department of Emergency Medicine, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang 110016, P.R. China
| | - Tanshi Li
- Department of Emergency Medicine, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, P.R. China
| |
Collapse
|
18
|
Li F, Lu J, Liu J, Liang C, Wang M, Wang L, Li D, Yao H, Zhang Q, Wen J, Zhang ZK, Li J, Lv Q, He X, Guo B, Guan D, Yu Y, Dang L, Wu X, Li Y, Chen G, Jiang F, Sun S, Zhang BT, Lu A, Zhang G. A water-soluble nucleolin aptamer-paclitaxel conjugate for tumor-specific targeting in ovarian cancer. Nat Commun 2017; 8:1390. [PMID: 29123088 PMCID: PMC5680242 DOI: 10.1038/s41467-017-01565-6] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 09/27/2017] [Indexed: 12/05/2022] Open
Abstract
Paclitaxel (PTX) is among the most commonly used first-line drugs for cancer chemotherapy. However, its poor water solubility and indiscriminate distribution in normal tissues remain clinical challenges. Here we design and synthesize a highly water-soluble nucleolin aptamer-paclitaxel conjugate (NucA-PTX) that selectively delivers PTX to the tumor site. By connecting a tumor-targeting nucleolin aptamer (NucA) to the active hydroxyl group at 2' position of PTX via a cathepsin B sensitive dipeptide bond, NucA-PTX remains stable and inactive in the circulation. NucA facilitates the uptake of the conjugated PTX specifically in tumor cells. Once inside cells, the dipeptide bond linker of NucA-PTX is cleaved by cathepsin B and then the conjugated PTX is released for action. The NucA modification assists the selective accumulation of the conjugated PTX in ovarian tumor tissue rather than normal tissues, and subsequently resulting in notably improved antitumor activity and reduced toxicity.
Collapse
Affiliation(s)
- Fangfei Li
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Jun Lu
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Jin Liu
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Chao Liang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Maolin Wang
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Luyao Wang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Defang Li
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Houzong Yao
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Qiulong Zhang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Jia Wen
- College of Science, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, P.R. China
| | - Zong-Kang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Jie Li
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Quanxia Lv
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Xiaojuan He
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Baosheng Guo
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Daogang Guan
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Yuanyuan Yu
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Lei Dang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Xiaohao Wu
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Yongshu Li
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Guofen Chen
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Guangzhou, 510515, China
| | - Feng Jiang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Shiguo Sun
- College of Science, Northwest Agriculture and Forestry University, Yangling, 712100, Shaanxi, P.R. China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China.
| | - Aiping Lu
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
| | - Ge Zhang
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
| |
Collapse
|
19
|
Meng FY, Gao W, Ju YN. Parecoxib reduced ventilation induced lung injury in acute respiratory distress syndrome. BMC Pharmacol Toxicol 2017; 18:25. [PMID: 28356130 PMCID: PMC5372249 DOI: 10.1186/s40360-017-0131-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/24/2017] [Indexed: 11/25/2022] Open
Abstract
Background Cyclooxygenase-2 (COX-2) contributes to ventilation induced lung injury (VILI) and acute respiratory distress syndrome (ARDS). The objective of present study was to observe the therapeutic effect of parecoxib on VILI in ARDS. Methods In this parallel controlled study performed at Harbin Medical University, China between January 2016 and March 2016, 24 rats were randomly allocated into sham group (S), volume ventilation group/ARDS (VA), parecoxib/volume ventilation group/ARDS (PVA). Rats in the S group only received anesthesia; rats in the VA and PVA group received intravenous injection of endotoxin to induce ARDS, and then received ventilation. Rats in the VA and PVA groups were treated with intravenous injection of saline or parecoxib. The ratio of arterial oxygen pressure to fractional inspired oxygen (PaO2/FiO2), the wet to dry weight ratio of lung tissue, inflammatory factors in serum and bronchoalveolar lavage fluid (BALF), and histopathologic analyses of lung tissue were examined. In addition, survival was calculated at 24 h after VILI. Results Compared to the VA group, in the PVA group, PaO2/FiO2 was significantly increased; lung tissue wet to dry weight ratio; macrophage and neutrophil counts, total protein and neutrophil elastase levels in BALF; tumor necrosis factor-α, interleukin-1β, and prostaglandin E2 levels in BALF and serum; and myeloperoxidase (MPO) activity, malondialdehyde levels, and Bax and COX-2 protein levels in lung tissue were significantly decreased, while Bcl-2 protein levels were significantly increased. Lung histopathogical changes and apoptosis were reduced by parecpxib in the PVA group. Survival was increased in the PVA group. Conclusions Parecoxib improves gas exchange and epithelial permeability, decreases edema, reduces local and systemic inflammation, ameliorates lung injury and apoptosis, and increases survival in a rat model of VILI.
Collapse
Affiliation(s)
- Fan-You Meng
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Wei Gao
- Department of Anesthesiology, the Second Affiliated Hospital of the Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Ying-Nan Ju
- Department of Intensive Care Unit, the Third Affiliated Hospital of the Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
20
|
Peter A, Fatykhova D, Kershaw O, Gruber AD, Rueckert J, Neudecker J, Toennies M, Bauer TT, Schneider P, Schimek M, Eggeling S, Suttorp N, Hocke AC, Hippenstiel S. Localization and pneumococcal alteration of junction proteins in the human alveolar-capillary compartment. Histochem Cell Biol 2017; 147:707-719. [PMID: 28247028 DOI: 10.1007/s00418-017-1551-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2017] [Indexed: 02/03/2023]
Abstract
Loss of alveolar barrier function with subsequent respiratory failure is a hallmark of severe pneumonia. Although junctions between endo- and epithelial cells regulate paracellular fluid flux, little is known about their composition and regulation in the human alveolar compartment. High autofluorescence of human lung tissue in particular complicates the determination of subcellular protein localization. By comparing conventional channel mode confocal imaging with spectral imaging and linear unmixing, we demonstrate that background fluorescent spectra and fluorophore signals could be rigorously separated resulting in complete recovery of the specific signal at a high signal-to-noise ratio. Using this technique and Western blotting, we show the expression patterns of tight junction proteins occludin, ZO-1 as well as claudin-3, -4, -5 and -18 and adherence junction protein VE-cadherin in naive or Streptococcus pneumoniae-infected human lung tissue. In uninfected tissues, occludin and ZO-1 formed band-like structures in alveolar epithelial cells type I (AEC I), alveolar epithelial cells type II (AEC II) and lung capillaries, whereas claudin-3, -4 and -18 were visualised in AEC II. Claudin-5 was detected in the endothelium only. Claudin-3, -5, -18 displayed continuous band-like structures, while claudin-4 showed a dot-like expression. Pneumococcal infection reduced alveolar occludin, ZO-1, claudin-5 and VE-cadherin but did not change the presence of claudin-3, -4 and -18. Spectral confocal microscopy allows for the subcellular structural analysis of proteins in highly autofluorescent human lung tissue. The thereby observed deterioration of lung alveolar junctional organisation gives a structural explanation for alveolar barrier disruption in severe pneumococcal pneumonia.
Collapse
Affiliation(s)
- Andrea Peter
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Department for Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355, Berlin, Germany
| | - Diana Fatykhova
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Jens Rueckert
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Jens Neudecker
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Mario Toennies
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Torsten T Bauer
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Paul Schneider
- Department for General and Thoracic Surgery, DRK Clinics, Drontheimer Strasse 39-40, 13359, Berlin, Germany
| | - Maria Schimek
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Stephan Eggeling
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas C Hocke
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|