1
|
Syenina A, Tan YH, Tng DJH, Sim SXQ, Chew VSY, Yee JX, Ong EZ, Ooi EE, Low JGH, Ng DHL. Transcriptional and cytokine signatures of Mycobacterium abscessus complex pulmonary disease during disease progression and treatment. PLoS Negl Trop Dis 2025; 19:e0012943. [PMID: 40163531 PMCID: PMC11981118 DOI: 10.1371/journal.pntd.0012943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/09/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Mycobacterium abscessus complex pulmonary disease (MABC-PD) is a chronic and often relapsing disease with considerable morbidity, especially among individuals with other chronic pulmonary conditions. A major clinical challenge lies in distinguishing infection-related symptoms from underlying lung disease and identifying reliable prognosticators to guide treatment decisions and monitoring therapeutic response. METHODOLOGY/PRINCIPAL FINDINGS To address the gaps in clinically relevant indicators, we profiled whole blood transcriptome and 45 plasma proteins of MABC-PD patients across different disease and treatment phases. Whole blood bulk RNA-sequencing revealed that MABC-PD patients with progressive disease exhibited elevated expression of genes related to innate immune and inflammatory responses, with reduced abundance of genes associated with peripheral T and NK cells. Among the 45 plasma cytokines and chemokines profiled, plasma levels of TNFSF10 were significantly reduced, while IFNγ, interleukin-17F (IL17F) and IL17C were elevated in patients with disease progression, despite the reduced abundance of peripheral T and NK cell-associated genes, suggesting recruitment of activated T cells to infection sites in the lungs during disease progression. Receiver operating characteristic (ROC) curve analysis of IFNγ and IL17F demonstrated strong predictive performance for differentiating patients with disease progression from healthy controls, with AUCs of 0.946 (95% CI 0.829-1.000) and 0.875 (95% CI 0.6699-1), respectively. CONCLUSIONS These findings provide insights into the immune profiles of MABC-PD patients during disease progression and suggest that T cell-associated cytokines, such as IFNγ and IL17F, could serve as useful biomarkers for identifying those under watchful waiting or post-treatment who are at risk of disease progression, thereby aiding in more timely and targeted therapeutic interventions.
Collapse
Affiliation(s)
- Ayesa Syenina
- Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Yi Hern Tan
- Department of Respiratory Medicine and Critical Care, Singapore General Hospital, Singapore, Singapore
| | - Danny Jian Hang Tng
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Sandy Xue Qi Sim
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Valerie Shyn Yun Chew
- Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Jia Xin Yee
- Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Eugenia Ziying Ong
- Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Eng Eong Ooi
- Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Department of Clinical Translational Research, Singapore General Hospital, Singapore, Singapore
| | - Jenny Guek Hong Low
- Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Dorothy Hui Lin Ng
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
2
|
Ruffenach G, Medzikovic L, Aryan L, Sun W, Lertpanit L, O'Connor E, Dehghanitafti A, Hatamnejad MR, Li M, Reddy ST, Eghbali M. Intestinal IFNα4 promotes 15-HETE diet-induced pulmonary hypertension. Respir Res 2024; 25:419. [PMID: 39609844 PMCID: PMC11606228 DOI: 10.1186/s12931-024-03046-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVES Pulmonary arterial hypertension (PAH) is characterized by the remodeling of the pulmonary vascular bed leading to elevation of the pulmonary arterial pressure. Oxidized fatty acids, such as hydroxyeicosatetraenoic acids (HETEs), play a critical role in PAH. We have previously established that dietary supplementation of 15-HETE is sufficient to cause PH in mice, suggesting a role for the gut-lung axis. However, the mechanisms are not known. APPROACH Analysis of RNA-seq data obtained from the lungs and intestines of mice on 15-HETE diet together with transcriptomic data from PAH patient lungs identified IFN inducible protein 44 (IFI44) as the only gene significantly upregulated in mice and humans. We demonstrate that IFI44 is also significantly increased in PBMCs from PAH patients. In mice, 15-HETE diet enhances IFI44 and its inducer IFN⍺4 expression sequentially in the intestine first and then in the lungs. IFI44 expression in PAH is highly correlated with expression of Tumor Necrosis Factor Related Apoptosis Inducing Ligand (TRAIL), which is upregulated in CD8 cells in PH lungs of both mice and humans. We show that IFNα4 produced by intestinal epithelial cells facilitates IFI44 expression in CD8 cells. Finally, we demonstrate that IFN receptor 1-KO in mice do not develop PH on 15-HETE diet. In addition, silencing IFI44 expression in the lungs of mice on 15-HETE diet prevents the development of PH and is associated with significantly lower expression of IFI44 and TRAIL in CD8 cells in the lungs. CONCLUSION Our data reveal a novel gut-lung axis driven by 15-HETE in PH.
Collapse
Affiliation(s)
- Grégoire Ruffenach
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA.
| | - Lejla Medzikovic
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Laila Aryan
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Wasila Sun
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Long Lertpanit
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-7115, USA
| | - Ellen O'Connor
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-7115, USA
| | - Ateyeh Dehghanitafti
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Min Li
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-7115, USA
| | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA.
| |
Collapse
|
3
|
Ren Z, Zhou B, Wang L, Li J, Zhang R, Pan X. [Inhibitory effect of 5-hydroxy-6,7-dimethoxyflavone on H1N1 influenza virus-induced ferroptosis and inflammation in A549 cells and its possible mechanisms]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1070-1078. [PMID: 38977336 PMCID: PMC11237301 DOI: 10.12122/j.issn.1673-4254.2024.06.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
OBJECTIVE To investigate the protective effect of 5-hydroxy-6,7-dimethoxyflavone (5-HDF), a compound extracted from Elsholtzia blanda Benth., against lung injury induced by H1N1 influenza virus and explore its possible mechanism of action. METHODS 5-HDF was extracted from Elsholtzia blanda Benth. using ethanol reflux extraction and silica gel chromatography and characterized using NMR and MS analyses. In an A549 cell model of H1N1 influenza virus infection (MOI=0.1), the cytotoxicity of 5-HDF was assessed using MTT assay, and its effect on TRAIL and IL-8 expressions was examined using flow cytometry; Western blotting was used to detect the expression levels of inflammatory, apoptosis, and ferroptosis-related proteins. In a mouse model of H1N1 influenza virus infection established by nasal instillation of 50 μL H1N1 virus at the median lethal dose, the effects of 30 and 60 mg/kg 5-HDF by gavage on body weight, lung index, gross lung anatomy and lung histopathology were observed. RESULTS 5-HDF exhibited no significant cytotoxicity in A549 cells within the concentration range of 0-200 μg/mL. In H1N1-infected A549 cells, treatment with 5-HDF effectively inhibited the activation of phospho-p38 MAPK and phospho-NF-κB p65, lowered the expressions of IL-8, enhanced the expression of anti-ferroptosis proteins (SLC7A11 and GPX4), and inhibited the expressions of apoptosis markers PARP and caspase-3 and the apoptotic factor TRAIL. In H1N1-infected mice, treatment with 5-HDF for 7 days significantly suppressed body weight loss and increment of lung index and obviously alleviated lung tissue pathologies. CONCLUSION 5-HDF offers protection against H1N1 influenza virus infection in mice possibly by suppressing H1N1-induced ferroptosis, inflammatory responses, and apoptosis via upregulating SLC7A11 and GPX4, inhibiting the activation of phospho-NF-κB p65 and phospho-p38 MAPK, and decreasing the expression of cleaved caspase3 and cleaved PARP.
Collapse
Affiliation(s)
- Z Ren
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - B Zhou
- Gaozhou People's Hospital, Gaozhou 525200, China
| | - L Wang
- Guangzhou Laboratory, Guangzhou 510000, China
| | - J Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - R Zhang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - X Pan
- Guangzhou Laboratory, Guangzhou 510000, China
| |
Collapse
|
4
|
Taheri MM, Javan F, Poudineh M, Athari SS. CAR-NKT Cells in Asthma: Use of NKT as a Promising Cell for CAR Therapy. Clin Rev Allergy Immunol 2024; 66:328-362. [PMID: 38995478 DOI: 10.1007/s12016-024-08998-0] [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] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
NKT cells, unique lymphocytes bridging innate and adaptive immunity, offer significant potential for managing inflammatory disorders like asthma. Activating iNKT induces increasing IFN-γ, TGF-β, IL-2, and IL-10 potentially suppressing allergic asthma. However, their immunomodulatory effects, including granzyme-perforin-mediated cytotoxicity, and expression of TIM-3 and TRAIL warrant careful consideration and targeted approaches. Although CAR-T cell therapy has achieved remarkable success in treating certain cancers, its limitations necessitate exploring alternative approaches. In this context, CAR-NKT cells emerge as a promising approach for overcoming these challenges, potentially achieving safer and more effective immunotherapies. Strategies involve targeting distinct IgE-receptors and their interactions with CAR-NKT cells, potentially disrupting allergen-mast cell/basophil interactions and preventing inflammatory cytokine release. Additionally, targeting immune checkpoints like PDL-2, inducible ICOS, FASL, CTLA-4, and CD137 or dectin-1 for fungal asthma could further modulate immune responses. Furthermore, artificial intelligence and machine learning hold immense promise for revolutionizing NKT cell-based asthma therapy. AI can optimize CAR-NKT cell functionalities, design personalized treatment strategies, and unlock a future of precise and effective care. This review discusses various approaches to enhancing CAR-NKT cell efficacy and longevity, along with the challenges and opportunities they present in the treatment of allergic asthma.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyyed Shamsadin Athari
- Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
5
|
Liu Y, Zhu FM, Xu J, Deng YP, Sun J, He QY, Cheng ZY, Tang MM, Yang J, Fu L, Zhao H. Arsenic exposure and pulmonary function decline: Potential mediating role of TRAIL in chronic obstructive pulmonary disease patients. J Trace Elem Med Biol 2024; 83:127415. [PMID: 38377659 DOI: 10.1016/j.jtemb.2024.127415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Environmental arsenic (As) exposure is strongly related to the progression of chronic obstructive pulmonary disease (COPD). Pulmonary epithelial cells apoptosis is implicated in the pathophysiological mechanisms of COPD. However, the role of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), one biomarker of apoptosis, remains unclear in As-mediated pulmonary function alternations in COPD patients. METHODS This study included 239 COPD patients. The serum level of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was measured by enzyme-linked immunosorbent assay (ELISA). The blood As level was determined through inductively coupled plasma mass spectrometry (ICP-MS). RESULTS Blood As levels exhibited a negative and dose-dependent correlation with pulmonary function. Per unit elevation of blood arsenic concentrations was related to reductions of 0.339 L in FEV1, 0.311 L in FVC, 1.171% in FEV1/FVC%, and 7.999% in FEV1% in COPD subjects. Additionally, a positive dose-response correlation of blood As with serum TRAIL was found in COPD subjects. Additionally, the level of serum TRAIL was negatively linked to lung function. Elevated TRAIL significantly mediated As-induced decreases of 11.05%, 13.35%, and 31.78% in FVC, FEV1, and FEV1%, respectively among the COPD patients. CONCLUSION Blood As level is positively correlated with pulmonary function decline and serum TRAIL increase in individuals with COPD. Our findings suggest that elevated TRAIL levels may serve as a mediating mechanism through which As contributes to declining lung function in COPD patients.
Collapse
Affiliation(s)
- Ying Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Center for Big Data and Population Health of IHM, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Feng-Min Zhu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Juan Xu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - You-Peng Deng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Jing Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Qi-Yuan He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Zhen-Yu Cheng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Min-Min Tang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Jin Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Center for Big Data and Population Health of IHM, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China.
| |
Collapse
|
6
|
Garg A, Karhana S, Bano A, Khan IA, Reeta, Nidhi, Khan MA. Network pharmacology and molecular docking study-based approach to explore mechanism of benzimidazole-based anthelmintics for the treatment of lung cancer. J Biomol Struct Dyn 2023; 42:10739-10760. [PMID: 37740654 DOI: 10.1080/07391102.2023.2258419] [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: 11/07/2022] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
Emerging studies have reported the potential anticancer activity of benzimidazole-based anthelmintics (BBA) against lung cancer (LC). However, mechanism underlying the anticancer activity of BBA is unclear. Therefore, in the current study, network pharmacology and molecular docking-based approach were used to explore the potential molecular mechanism for the treatment of LC. The potential targets for BBA were obtained from multiple databases including SwissTargetPrediction, Drug Bank, Therapeutic Target Database, and Comparative Toxicogenomics Database while LC targets were collected from DisGeNet gene discovery platform, Integrated Genomic Database of NSCLC, Catalogue of Somatic Mutations in Cancer and Online Mendelian Inheritance in Man database. Protein-protein interaction (PPI) diagram of common targets was constructed using STRING online platform. Topological analysis was performed using Cytoscape and gene enrichment analysis was conducted using FunRich software. Highest degree targets were then confirmed using molecular docking and molecular dynamics simulations. The BBA were prioritized according to their S scores, with ricobendazole ranking highest followed by flubendazole, fenbendazole, mebendazole, triclabendazole, albendazole, oxibendazole, parbendazole, thiabendazole and oxfendazole. The potential targets of BBA identified using topological analysis and molecular docking were found to be CCND1 (cyclin D1), EGFR (Epidermal Growth Factor Receptor), ERBB2 (Erb-B2 Receptor Tyrosine Kinase 2/CD340), PTGS2 (Prostaglandin-endoperoxide synthase 2), and SRC (Proto-oncogene tyrosine-protein kinase). Furthermore, molecular dynamics confirmed that CCND1 and EGFR are the potential targets of ricobendazole for the treatment of LC. BBA can be further explored as a therapeutic strategy for the treatment of lung cancer under in vitro and in vivo studies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aakriti Garg
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sonali Karhana
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Aysha Bano
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Imran A Khan
- Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Reeta
- Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, New Delhi, India
| | - Nidhi
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Ashif Khan
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
7
|
Riera-Domingo C, Leite-Gomes E, Charatsidou I, Zhao P, Carrá G, Cappellesso F, Mourao L, De Schepper M, Liu D, Serneels J, Alameh MG, Shuvaev VV, Geukens T, Isnaldi E, Prenen H, Weissman D, Muzykantov VR, Soenen S, Desmedt C, Scheele CL, Sablina A, Di Matteo M, Martín-Pérez R, Mazzone M. Breast tumors interfere with endothelial TRAIL at the premetastatic niche to promote cancer cell seeding. SCIENCE ADVANCES 2023; 9:eadd5028. [PMID: 36947620 PMCID: PMC10032608 DOI: 10.1126/sciadv.add5028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
Endothelial cells (ECs) grant access of disseminated cancer cells to distant organs. However, the molecular players regulating the activation of quiescent ECs at the premetastatic niche (PMN) remain elusive. Here, we find that ECs at the PMN coexpress tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its cognate death receptor 5 (DR5). Unexpectedly, endothelial TRAIL interacts intracellularly with DR5 to prevent its signaling and preserve a quiescent vascular phenotype. In absence of endothelial TRAIL, DR5 activation induces EC death and nuclear factor κB/p38-dependent EC stickiness, compromising vascular integrity and promoting myeloid cell infiltration, breast cancer cell adhesion, and metastasis. Consistently, both down-regulation of endothelial TRAIL at the PMN by proangiogenic tumor-secreted factors and the presence of the endogenous TRAIL inhibitors decoy receptor 1 (DcR1) and DcR2 favor metastasis. This study discloses an intracrine mechanism whereby TRAIL blocks DR5 signaling in quiescent endothelia, acting as gatekeeper of the vascular barrier that is corrupted by the tumor during cancer cell dissemination.
Collapse
Affiliation(s)
- Carla Riera-Domingo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Eduarda Leite-Gomes
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Iris Charatsidou
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Peihua Zhao
- Laboratory for Mechanisms of Cell Transformation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Mechanisms of Cell Transformation, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Giovanna Carrá
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
- Molecular Biotechnology Center, Torino, Italy
| | - Federica Cappellesso
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Larissa Mourao
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Maxim De Schepper
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Dana Liu
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jens Serneels
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Vladimir V. Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tatjana Geukens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Edoardo Isnaldi
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Hans Prenen
- Department of Oncology, University Hospital Antwerp, Edegem, Belgium
| | - Drew Weissman
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefaan Soenen
- Leuven Cancer Institute, KU Leuven, Belgium
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Colinda L. G. J. Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Anna Sablina
- Laboratory for Mechanisms of Cell Transformation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Mechanisms of Cell Transformation, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mario Di Matteo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Rosa Martín-Pérez
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Boucly A, Tu L, Guignabert C, Rhodes C, De Groote P, Prévot G, Bergot E, Bourdin A, Beurnier A, Roche A, Jevnikar M, Jaïs X, Montani D, Wilkins MR, Humbert M, Sitbon O, Savale L. Cytokines as prognostic biomarkers in pulmonary arterial hypertension. Eur Respir J 2023; 61:2201232. [PMID: 36549710 DOI: 10.1183/13993003.01232-2022] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/12/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Risk stratification and assessment of disease progression in patients with pulmonary arterial hypertension (PAH) are challenged by the lack of accurate disease-specific and prognostic biomarkers. To date, brain natriuretic peptide (BNP) and/or its N-terminal fragment (NT-proBNP) are the only markers for right ventricular dysfunction used in clinical practice, in association with echocardiographic and invasive haemodynamic variables to predict outcome in patients with PAH. METHODS This study was designed to identify an easily measurable biomarker panel in the serum of 80 well-phenotyped PAH patients with idiopathic, heritable or drug-induced PAH at baseline and at first follow-up. The prognostic value of identified cytokines of interest was secondly analysed in an external validation cohort of 125 PAH patients. RESULTS Among the 20 biomarkers studied with the multiplex Ella platform, we identified a three-biomarker panel composed of β-NGF, CXCL9 and TRAIL that were independently associated with prognosis both at the time of PAH diagnosis and at the first follow-up after initiation of PAH therapy. β-NGF and CXCL9 were predictors of death or transplantation, whereas high levels of TRAIL were associated with a better prognosis. Furthermore, the prognostic value of the three cytokines was more powerful for predicting survival than usual non-invasive variables (New York Heart Association Functional Class, 6-min walk distance and BNP/NT-proBNP). The results were validated in a fully independent external validation cohort. CONCLUSION The monitoring of β-NGF, CXCL9 and TRAIL levels in serum should be considered in the management and treatment of patients with PAH to objectively guide therapeutic options.
Collapse
Affiliation(s)
- Athénaïs Boucly
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | | | - Pascal De Groote
- Université de Lille, Service de Cardiologie, CHU Lille, Institut Pasteur de Lille, Inserm U1167, Lille, France
| | - Grégoire Prévot
- CHU de Toulouse, Hôpital Larrey, Service de Pneumologie, Toulouse, France
| | - Emmanuel Bergot
- Unicaen, UFR Santé, Service de Pneumologie & Oncologie Thoracique, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Arnaud Bourdin
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR_9214, Montpellier, France
| | - Antoine Beurnier
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Anne Roche
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Mitja Jevnikar
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Xavier Jaïs
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - David Montani
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Marc Humbert
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Olivier Sitbon
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- These authors contributed equally to this work
| | - Laurent Savale
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- These authors contributed equally to this work
| |
Collapse
|
9
|
Unterhuber M, Kresoja KP, Lurz P, Thiele H. Artificial intelligence in proteomics: new frontiers from risk prediction to treatment? Eur Heart J 2022; 43:4525-4527. [PMID: 35869894 DOI: 10.1093/eurheartj/ehac391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Matthias Unterhuber
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Strümpellstraße 39, Leipzig 04289, Germany
| | - Karl-Patrik Kresoja
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Strümpellstraße 39, Leipzig 04289, Germany
| | - Philipp Lurz
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Strümpellstraße 39, Leipzig 04289, Germany
| | - Holger Thiele
- Department of Cardiology, Heart Center Leipzig at University of Leipzig, Strümpellstraße 39, Leipzig 04289, Germany.,Leipzig Heart Institute at Heart Center Leipzig, Russenstraße 69A, Leipzig 04289, Germany
| |
Collapse
|
10
|
TRAIL protects the immature lung from hyperoxic injury. Cell Death Dis 2022; 13:614. [PMID: 35840556 PMCID: PMC9287454 DOI: 10.1038/s41419-022-05072-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
The hyperoxia-induced pro-inflammatory response and tissue damage constitute pivotal steps leading to bronchopulmonary dysplasia (BPD) in the immature lung. The pro-inflammatory cytokines are considered attractive candidates for a directed intervention but the complex interplay between inflammatory and developmental signaling pathways requires a comprehensive evaluation before introduction into clinical trials as studied here for the death inducing ligand TRAIL. At birth and during prolonged exposure to oxygen and mechanical ventilation, levels of TRAIL were lower in tracheal aspirates of preterm infants <29 weeks of gestation which developed moderate/severe BPD. These findings were reproduced in the newborn mouse model of hyperoxic injury. The loss of TRAIL was associated with increased inflammation, apoptosis induction and more pronounced lung structural simplification after hyperoxia exposure for 7 days while activation of NFκB signaling during exposure to hyperoxia was abrogated. Pretreatment with recombinant TRAIL rescued the developmental distortions in precision cut lung slices of both wildtype and TRAIL-/- mice exposed to hyperoxia. Of importance, TRAIL preserved alveolar type II cells, mesenchymal progenitor cells and vascular endothelial cells. In the situation of TRAIL depletion, our data ascribe oxygen toxicity a more injurious impact on structural lung development. These data are not surprising taking into account the diverse functions of TRAIL and its stimulatory effects on NFκB signaling as central driver of survival and development. TRAIL exerts a protective role in the immature lung as observed for the death inducing ligand TNF-α before.
Collapse
|
11
|
Xia Y, Wang Y, Xiao Y, Shan M, Hao Y, Zhang L. Identification of a Diagnostic Signature and Immune Cell Infiltration Characteristics in Keloids. Front Mol Biosci 2022; 9:879461. [PMID: 35669563 PMCID: PMC9163372 DOI: 10.3389/fmolb.2022.879461] [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: 02/19/2022] [Accepted: 04/13/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Keloid disorder is a recurrent fibroproliferative cutaneous tumor. Due to the lack of early identification of keloid patients before the formation of keloids, it is impossible to carry out pre-traumatic intervention and prevention for these patients. This led us to identify and determine signatures with diagnostic significance for keloids. Methods: Public series of matrix files were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were calculated from expression profiling data, and the diagnostic signature was identified by constructing a protein-protein interaction (PPI) network. The diagnostic efficacy of the screened signature was assessed by employing receiver operating characteristic (ROC) curves. Furthermore, we calculated the proportion of different immune cells in the gene expression matrix microenvironment by the “ssGSEA” algorithm, and assessed the difference in immune cell abundance between keloids and control groups and the relationship between the signature and immune cell infiltration. Clinical keloid and normal skin tissues were collected, and the expression of the screened diagnostic signature was validated by RT-qPCR and immunohistochemical assay. Results: By screening the key genes in PPI, TGM2 was recognized and validated as a diagnostic signature and the infiltrating abundance of 10 immune cells was significantly correlated with TGM2 expression. Gene ontology enrichment analysis demonstrated that TGM2 and molecules interacting with it were mainly enriched in processes involving wound healing and collagen fiber organization. TGM2 correlated positively with HIF-1A (R = 0.82, p-value = 1.4e-05), IL6 (R = 0.62, p-value = 0.0053), and FN1 (R = 0.66, p-value = 0.0019). Besides, TGM2 was significantly upregulated in clinical keloid samples compared to normal skin tissues. Conclusion: TGM2 may serve as an auxiliary diagnostic indicator for keloids. However, the role of TGM2 in keloids has not been adequately reported in the current literature, which may provide a new direction for molecular studies of keloids.
Collapse
Affiliation(s)
- Yijun Xia
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Youbin Wang,
| | - Yingjie Xiao
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mengjie Shan
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Hao
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lingyun Zhang
- Department of Plastic Surgery, Heze Municipal Hospital, Heze, China
| |
Collapse
|
12
|
Abstract
Pulmonary hypertension (PH) describes heterogeneous population of patients with a mean pulmonary arterial pressure >20 mm Hg. Rarely, PH presents as a primary disorder but is more commonly part of a complex phenotype associated with comorbidities. Regardless of the cause, PH reduces life expectancy and impacts quality of life. The current clinical classification divides PH into 1 of 5 diagnostic groups to assign treatment. There are currently no pharmacological cures for any form of PH. Animal models are essential to help decipher the molecular mechanisms underlying the disease, to assign genotype-phenotype relationships to help identify new therapeutic targets, and for clinical translation to assess the mechanism of action and putative efficacy of new therapies. However, limitations inherent of all animal models of disease limit the ability of any single model to fully recapitulate complex human disease. Within the PH community, we are often critical of animal models due to the perceived low success upon clinical translation of new drugs. In this review, we describe the characteristics, advantages, and disadvantages of existing animal models developed to gain insight into the molecular and pathological mechanisms and test new therapeutics, focusing on adult forms of PH from groups 1 to 3. We also discuss areas of improvement for animal models with approaches combining several hits to better reflect the clinical situation and elevate their translational value.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allan Lawrie
- Dept of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK & Insigneo institute for in silico medicine, Sheffield, UK
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
13
|
Latt KZ, Heymann J, Jessee JH, Rosenberg AZ, Berthier CC, Arazi A, Eddy S, Yoshida T, Zhao Y, Chen V, Nelson GW, Cam M, Kumar P, Mehta M, Kelly MC, Kretzler M, Ray PE, Moxey-Mims M, Gorman GH, Lechner BL, Regunathan-Shenk R, Raj DS, Susztak K, Winkler CA, Kopp JB. Urine Single-Cell RNA Sequencing in Focal Segmental Glomerulosclerosis Reveals Inflammatory Signatures. Kidney Int Rep 2022; 7:289-304. [PMID: 35155868 PMCID: PMC8821042 DOI: 10.1016/j.ekir.2021.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Individuals with focal segmental glomerular sclerosis (FSGS) typically undergo kidney biopsy only once, which limits the ability to characterize kidney cell gene expression over time. METHODS We used single-cell RNA sequencing (scRNA-seq) to explore disease-related molecular signatures in urine cells from subjects with FSGS. We collected 17 urine samples from 12 FSGS subjects and captured these as 23 urine cell samples. The inflammatory signatures from renal epithelial and immune cells were evaluated in bulk gene expression data sets of FSGS and minimal change disease (MCD) (The Nephrotic Syndrome Study Network [NEPTUNE] study) and an immune single-cell data set from lupus nephritis (Accelerating Medicines Partnership). RESULTS We identified immune cells, predominantly monocytes, and renal epithelial cells in the urine. Further analysis revealed 2 monocyte subtypes consistent with M1 and M2 monocytes. Shed podocytes in the urine had high expression of marker genes for epithelial-to-mesenchymal transition (EMT). We selected the 16 most highly expressed genes from urine immune cells and 10 most highly expressed EMT genes from urine podocytes as immune signatures and EMT signatures, respectively. Using kidney biopsy transcriptomic data from NEPTUNE, we found that urine cell immune signature and EMT signature genes were more highly expressed in FSGS biopsies compared with MCD biopsies. CONCLUSION The identification of monocyte subsets and podocyte expression signatures in the urine samples of subjects with FSGS suggests that urine cell profiling might serve as a diagnostic and prognostic tool in nephrotic syndrome. Furthermore, this approach may aid in the development of novel biomarkers and identifying personalized therapies targeting particular molecular pathways in immune cells and podocytes.
Collapse
Affiliation(s)
- Khun Zaw Latt
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jurgen Heymann
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph H. Jessee
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Celine C. Berthier
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Arnon Arazi
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Sean Eddy
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Teruhiko Yoshida
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Yongmei Zhao
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., National Cancer Institute, Frederick, Maryland, USA
| | - Vicky Chen
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., National Cancer Institute, Frederick, Maryland, USA
| | - George W. Nelson
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., National Cancer Institute, Frederick, Maryland, USA
| | - Margaret Cam
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., National Cancer Institute, Frederick, Maryland, USA
| | - Parimal Kumar
- Center for Cancer Research Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Monika Mehta
- Center for Cancer Research Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Michael C. Kelly
- Cancer Research Technology Program, Single-Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - The Nephrotic Syndrome Study Network (NEPTUNE)
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, USA
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., National Cancer Institute, Frederick, Maryland, USA
- Center for Cancer Research Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
- Cancer Research Technology Program, Single-Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia, USA
- Division of Nephrology, Children’s National Hospital, Washington, District of Columbia, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
- Section on Pediatric Nephrology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, Maryland, USA
- Division of Kidney Disease and Hypertension, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - The Accelerating Medicines Partnership in Rheumatoid Arthritis and Systemic Lupus Erythematosus (AMP RA/SLE) Consortium
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, USA
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., National Cancer Institute, Frederick, Maryland, USA
- Center for Cancer Research Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
- Cancer Research Technology Program, Single-Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia, USA
- Division of Nephrology, Children’s National Hospital, Washington, District of Columbia, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
- Section on Pediatric Nephrology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, Maryland, USA
- Division of Kidney Disease and Hypertension, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Patricio E. Ray
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Marva Moxey-Mims
- Division of Nephrology, Children’s National Hospital, Washington, District of Columbia, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Gregory H. Gorman
- Section on Pediatric Nephrology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, Maryland, USA
| | - Brent L. Lechner
- Section on Pediatric Nephrology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, Maryland, USA
| | - Renu Regunathan-Shenk
- Division of Kidney Disease and Hypertension, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Dominic S. Raj
- Division of Kidney Disease and Hypertension, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cheryl A. Winkler
- Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Jeffrey B. Kopp
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Peterson‐Reynolds C, Mantis NJ. Differential ER stress as a driver of cell fate following ricin toxin exposure. FASEB Bioadv 2022; 4:60-75. [PMID: 35024573 PMCID: PMC8728110 DOI: 10.1096/fba.2021-00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/16/2021] [Accepted: 08/30/2021] [Indexed: 11/11/2022] Open
Abstract
Inhalation of trace amounts of ricin toxin, a plant-derived ribosome-inactivating protein, results in ablation of alveolar macrophages, widespread epithelial damage, and the onset of acute respiratory distress syndrome (ARDS). While ricin's receptors are ubiquitous, certain cell types are more sensitive to ricin-induced cell death than others for reasons that remain unclear. For example, we demonstrate in side-by-side studies that macrophage-like differentiated THP-1 (dTHP-1) cells are hyper-sensitive to ricin, while lung epithelium-derived A549 cells are relatively insensitive, even though both cell types experience similar degrees of translational inhibition and p38 MAPK activation in response to ricin. Using a variety of small molecule inhibitors, we provide evidence that ER stress contributes to ricin-mediated cytotoxicity of dTHP-1 cells, but not A549 cells. On the other hand, the insensitivity of A549 cells to ricin was overcome by the addition of (TNF)-related apoptosis-inducing ligand (TRAIL; CD253), a known stimulator of extrinsic programmed cell death. These results have implications for understanding the complex pathophysiology of ricin-induced ARDS in that they demonstrate that intrinsic (e.g., ER stress) and extrinsic (e.g., TRAIL) factors may ultimately determine the fate of specific cell types following ricin intoxication.
Collapse
Affiliation(s)
- Claire Peterson‐Reynolds
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| | - Nicholas J. Mantis
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| |
Collapse
|
15
|
Latt KZ, Heymann J, Yoshida T, Kopp JB. Glomerular Kidney Diseases in the Single-Cell Era. Front Med (Lausanne) 2021; 8:761996. [PMID: 34778322 PMCID: PMC8585743 DOI: 10.3389/fmed.2021.761996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
Abstract
Recent advances in single-cell technology have enabled investigation of genomic profiles and molecular crosstalk among individual cells obtained from tissues and biofluids at unprecedented resolution. Glomerular diseases, either primary or secondary to systemic diseases, often manifest elements of inflammation and of innate and adaptive immune responses. Application of single-cell methods have revealed cellular signatures of inflammation, cellular injury, and fibrosis. From these signatures, potential therapeutic targets can be inferred and in theory, this approach might facilitate identification of precision therapeutics for these diseases. Single-cell analyses of urine samples and skin lesions from patients with lupus nephritis and of urine samples from patients with diabetic nephropathy and focal segmental glomerulosclerosis have presented potential novel approaches for the diagnosis and monitoring of disease activity. These single-cell approaches, in contrast to kidney biopsy, are non-invasive and could be repeated multiple times as needed.
Collapse
Affiliation(s)
- Khun Zaw Latt
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | | | | |
Collapse
|
16
|
Alfhili MA, Basudan AM, Aljaser FS, Dera A, Alsughayyir J. Bioymifi, a novel mimetic of TNF-related apoptosis-induced ligand (TRAIL), stimulates eryptosis. Med Oncol 2021; 38:138. [PMID: 34633592 DOI: 10.1007/s12032-021-01589-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/21/2021] [Indexed: 11/26/2022]
Abstract
Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) is a cytokine that initiates apoptosis upon binding to death receptor 5 (DR5) on cancer cells. Small molecule TRAIL mimetics have therefore been investigated as promising chemotherapeutic agents. Since anemia of chemotherapy is common, our goal is to investigate the hemolytic and eryptotic properties of novel DR5 agonist bioymifi (BMF) and identify the underlying molecular mechanisms. Whole blood (WB) was stimulated with 100 μM of BMF, whereas red blood cells (RBCs) were treated with 10-100 μM of BMF for 24 h at 37 °C. WB was analyzed for RBC, leukocyte, and platelet indices, while RBCs were examined for hemolysis by light absorbance of free hemoglobin, membrane scrambling by Annexin V-FITC, calcium by Fluo4/AM, cellular morphology by light scatter, and oxidative stress by 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) using flow cytometry. Caspase inhibitor Z-VAD-FMK, p38 inhibitor SB203580, casein kinase 1α inhibitor D4476, receptor-interacting protein 1 inhibitor necrostatin-2, reduced glutathione, or cyclooxygenase (COX) inhibitor aspirin were added accordingly. BMF exerted dose-responsive, calcium-independent hemolysis, reduced RBC hemoglobin, significantly increased Annexin V-, Fluo4-, and DCF-positive cells, along with a dual effect on forward and side light scatter. Notably, the cytotoxic potential of BMF was significantly mitigated upon pharmacological inhibition of p38. Furthermore, BMF exhibited selective toxicity to eosinophils and significantly diminished reticulocyte hemoglobin content. Altogether, these novel findings highlight the adverse outcomes of BMF exposure on RBC physiology and provide the first toxicological assessment of BMF as an antitumor agent.
Collapse
Affiliation(s)
- Mohammad A Alfhili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Ahmed M Basudan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Feda S Aljaser
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ayed Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia
| | - Jawaher Alsughayyir
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
17
|
Zhang R, Ren Z, Celedón JC, Chen W. Inference of large modified Poisson-type graphical models: Application to RNA-seq data in childhood atopic asthma studies. Ann Appl Stat 2021. [DOI: 10.1214/20-aoas1413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Rong Zhang
- Department of Statistics, University of Pittsburgh
| | - Zhao Ren
- Department of Statistics, University of Pittsburgh
| | - Juan C. Celedón
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh
| | - Wei Chen
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh
| |
Collapse
|
18
|
Song Q, Chen P, Liu XM. The role of cigarette smoke-induced pulmonary vascular endothelial cell apoptosis in COPD. Respir Res 2021; 22:39. [PMID: 33546691 PMCID: PMC7866753 DOI: 10.1186/s12931-021-01630-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common chronic respiratory diseases with high morbidity and mortality. It has become the fifth most burdened and the third most deadly disease in the global economy and increases year by year. The prevention and treatment of COPD are urgent. Smoking is the main and most common risk factor for COPD. Cigarette smoke (CS) contains a large number of toxic substances, can cause a series of changes in the trachea, lung tissue, pulmonary blood vessels, and promotes the occurrence and development of COPD. In recent years, the development of epigenetics and molecular biology have provided new guidance for revealing the pathogenesis, diagnosis, and treatment of diseases. The latest research indicates that pulmonary vascular endothelial cell apoptosis initiates and participates in the pathogenesis of COPD. In this review, we summarize the current research on the epigenetic mechanisms and molecular biology of CS-induced pulmonary vascular endothelial cell apoptosis in COPD, providing a new research direction for pathogenesis of COPD and a new target for the diagnosis, treatment, and prevention of COPD.
Collapse
Affiliation(s)
- Qing Song
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Research Unit of Respiratory Disease, Diagnosis and Treatment Center of Respiratory Disease, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Research Unit of Respiratory Disease, Diagnosis and Treatment Center of Respiratory Disease, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China.
| | - Xiang-Ming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Research Unit of Respiratory Disease, Diagnosis and Treatment Center of Respiratory Disease, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, China
| |
Collapse
|
19
|
Metz JK, Wiegand B, Schnur S, Knoth K, Schneider-Daum N, Groß H, Croston G, Reinheimer TM, Lehr CM, Hittinger M. Modulating the Barrier Function of Human Alveolar Epithelial (hAELVi) Cell Monolayers as a Model of Inflammation. Altern Lab Anim 2021; 48:252-267. [DOI: 10.1177/0261192920983015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The incidence of inflammatory lung diseases such as acute respiratory distress syndrome (ARDS) remains an important problem, particularly in the present time with the Covid-19 pandemic. However, an adequate in vitro test system to monitor the barrier function of the alveolar epithelium during inflammation and for assessing anti-inflammatory drugs is urgently needed. Therefore, we treated human Alveolar Epithelial Lentivirus-immortalised cells (hAELVi cells) with the pro-inflammatory cytokines TNF-α (25 ng/ml) and IFN-γ (30 ng/ml), in the presence or absence of hydrocortisone (HC). While TNF-α and IFN-γ are known to reduce epithelial barrier properties, HC could be expected to protect the barrier function and result in an anti-inflammatory effect. We investigated the impact of anti-inflammatory/inflammatory treatment on transepithelial electrical resistance (TEER) and the apparent permeability coefficient (P app) of the low permeability marker sodium fluorescein (NaFlu). After incubating hAELVi cells for 48 hours with a combination of TNF-α and IFN-γ, there was a significant decrease in TEER and a significant increase in the P app. The presence of HC maintained the TEER values and barrier properties, so that no significant P app change was observed. By using hAELVi cells to study anti-inflammatory drugs in vitro, the need for animal experiments could be reduced and pulmonary drug development accelerated.
Collapse
Affiliation(s)
- Julia Katharina Metz
- PharmBioTec GmbH, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Sabrina Schnur
- PharmBioTec GmbH, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Nicole Schneider-Daum
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | | | | | | | - Claus-Michael Lehr
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Marius Hittinger
- PharmBioTec GmbH, Saarbrücken, Germany
- 3RProducts Marius Hittinger, Blieskastel, Germany
| |
Collapse
|
20
|
Cools F, Delputte P, Cos P. The search for novel treatment strategies for Streptococcus pneumoniae infections. FEMS Microbiol Rev 2021; 45:6064299. [PMID: 33399826 PMCID: PMC8371276 DOI: 10.1093/femsre/fuaa072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/01/2021] [Indexed: 12/13/2022] Open
Abstract
This review provides an overview of the most important novel treatment strategies against Streptococcus pneumoniae infections published over the past 10 years. The pneumococcus causes the majority of community-acquired bacterial pneumonia cases, and it is one of the prime pathogens in bacterial meningitis. Over the last 10 years, extensive research has been conducted to prevent severe pneumococcal infections, with a major focus on (i) boosting the host immune system and (ii) discovering novel antibacterials. Boosting the immune system can be done in two ways, either by actively modulating host immunity, mostly through administration of selective antibodies, or by interfering with pneumococcal virulence factors, thereby supporting the host immune system to effectively overcome an infection. While several of such experimental therapies are promising, few have evolved to clinical trials. The discovery of novel antibacterials is hampered by the high research and development costs versus the relatively low revenues for the pharmaceutical industry. Nevertheless, novel enzymatic assays and target-based drug design, allow the identification of targets and the development of novel molecules to effectively treat this life-threatening pathogen.
Collapse
Affiliation(s)
- F Cools
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Delputte
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
21
|
Reynolds-Peterson C, Ehrbar DJ, McHale SM, LaRocca TJ, Mantis NJ. Sensitization of Airway Epithelial Cells to Toxin-Induced Death by TNF Superfamily Cytokines. Methods Mol Biol 2021; 2248:19-42. [PMID: 33185865 DOI: 10.1007/978-1-0716-1130-2_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The TNF superfamily of proinflammatory and proapoptotic cytokines influence tissue-wide responses to molecular insults such as small molecules, toxins, and viral infections that perturb cellular homeostasis at the level of DNA replication, transcription, and translation. In the context of acute lung injury, for example, TNF superfamily members like TNF-α and TRAIL can severely exacerbate disease pathophysiology. This chapter describes a systematic approach to optimization of mammalian cell viability assays and transcriptional profiling through nCounter® Technology to permit a detailed examination of how TNF-α and TRAIL modulate programmed cell death pathways in concert with ricin toxin, a ribosome-inactivating protein (RIP) and a potent inducer of acute respiratory distress. We compare two widely used luciferase- and colorimetric-based cell viability assays and provide optimization protocols for adherent and non-adherent cell lines. We provide a computational workflow to facilitate downstream analysis of datasets generated from nCounter® gene expression panels. While combined treatment with ricin toxin and TRAIL serves as the exemplar, the methodologies are applicable to any TNF superfamily member in combination with any biological agent of interest.
Collapse
Affiliation(s)
- Claire Reynolds-Peterson
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Dylan J Ehrbar
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Susanne M McHale
- Advanced Genomic Technologies Cluster, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Timothy J LaRocca
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA.
| |
Collapse
|
22
|
Zaigham S, Dencker M, Karlsson MK, Thorsson O, Wollmer P. Lung function is associated with tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) levels in school-aged children. Respir Med 2020; 176:106235. [PMID: 33249302 DOI: 10.1016/j.rmed.2020.106235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with inflammatory and apoptotic properties. A complex relationship exists between TRAIL and the lung where both elevated TRAIL and TRAIL deficiency are associated with lung impairment. In neonatal mice, TRAIL is thought to translate respiratory infections into chronic lung disease but the association between TRAIL and lung function in childhood has not been assessed. AIM To assess the cross-sectional relationship between TRAIL levels and lung function in school-aged children. METHODS The study cohort consisted of 170 school-aged children attending four schools in Malmö, Sweden. Lung volumes, impulse oscillometry (IOS) and serum TRAIL were measured for all children. Linear regression was used to assess changes in lung function per 1-SD increase in TRAIL. General linear models were used to assess mean lung function by tertiles (T) of TRAIL. RESULTS Mean age was 9.9 years (±0.6). A 1-SD increase in TRAIL was associated with lower values of FEV1 and FEV1/VC (change in FEV1 (L) and FEV1/VC ratio: -0.047, p-value 0.002, and -0.011, p-value 0.020, respectively) and higher values of lung resistance (change in R5 and R20 (kPa/(L/s)): 0.035, p-value <0.001 and 0.027, p-value 0.004, respectively). These associations remained significant after excluding children with pre-existing lung disease. Higher TRAIL levels were associated with more negative values for X5 in general linear models (Mean X5 (kPa/(L/s)) in T1 (low TRAIL): -0.193 vs T3 (high TRAIL): -0.216, p-value 0.026). CONCLUSIONS High TRAIL levels are significantly associated with markers of pulmonary airflow obstruction in school-aged children.
Collapse
Affiliation(s)
- Suneela Zaigham
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
| | - Magnus Dencker
- Department of Translational Medicine, Clinical Physiology and Nuclear Medicine, Skåne University Hospital (SUS), Malmö, Sweden.
| | - Magnus K Karlsson
- Department of Orthopedics and Clinical Sciences Malmö, Lund University, Skåne University Hospital (SUS), Malmö, Sweden.
| | - Ola Thorsson
- Department of Translational Medicine, Clinical Physiology and Nuclear Medicine, Skåne University Hospital (SUS), Malmö, Sweden.
| | - Per Wollmer
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Department of Translational Medicine, Clinical Physiology and Nuclear Medicine, Skåne University Hospital (SUS), Malmö, Sweden.
| |
Collapse
|
23
|
Marks M, Steele C, Moore WC, Meyers DA, Rector B, Ampleford E, Bleecker ER, Hastie AT. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and receptors in type 1, type 2 and type 17 inflammation in cross-sectional asthma study. Thorax 2020; 75:808-811. [PMID: 32482836 PMCID: PMC7476262 DOI: 10.1136/thoraxjnl-2019-214496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/02/2020] [Accepted: 04/25/2020] [Indexed: 12/03/2022]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) reportedly promotes, or conversely, resolves inflammation in asthma. In this study of TRAIL and cell receptors in sputum, bronchoalveolar lavage and biopsy from subjects in the Severe Asthma Research Program at Wake Forest, the high TRAIL group had significant increases in all leucocytes, and was associated with increased type 1, type 2 and type 17 cytokines, but not type 9 interleukin 9. Two variants at loci in the TRAIL gene were associated with higher sputum levels of TRAIL. Increased TRAIL decoy receptor R3/DcR1 was observed on sputum leucocytes compared with death receptor R1/DR4, suggesting reduced apoptosis and prolonged cellular inflammation.
Collapse
Affiliation(s)
- Michelle Marks
- Department of Internal Medicine, Pulmonary Section, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Chad Steele
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Wendy C Moore
- Department of Internal Medicine, Pulmonary Section, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Deborah A Meyers
- Department of Medicine, Division of Genetics, Genomics and Precision Medicine, University of Arizona Health Sciences, Tuscon, Arizona, USA
| | - Brian Rector
- Department of Internal Medicine, Pulmonary Section, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Elizabeth Ampleford
- Department of Internal Medicine, Pulmonary Section, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Eugene R Bleecker
- Department of Medicine, Division of Genetics, Genomics and Precision Medicine, University of Arizona Health Sciences, Tuscon, Arizona, USA
| | - Annette T Hastie
- Department of Internal Medicine, Pulmonary Section, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
24
|
Gao S, Fang Y, Tu S, Chen H, Shao A. Insight into the divergent role of TRAIL in non-neoplastic neurological diseases. J Cell Mol Med 2020; 24:11070-11083. [PMID: 32827246 PMCID: PMC7576257 DOI: 10.1111/jcmm.15757] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/04/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Tumour necrosis factor–related apoptosis‐inducing ligand (TRAIL) is a member of the tumour necrosis factor (TNF) superfamily which mainly induces apoptosis of tumour cells and transformed cell lines with no systemic toxicity, whereas they share high sequence homology with TNF and CD95L. These unique effects of TRAIL have made it an important molecule in oncology research. However, the research on TRAIL‐related antineoplastic agents has lagged behind and has been limited by the extensive drug resistance in cancer cells. Given the several findings showing that TRAIL is involved in immune regulation and other pleiotropic biological effects in non‐malignant cells, TRAIL and its receptors have attracted widespread attention from researchers. In the central nervous system (CNS), TRAIL is highly correlated with malignant tumours such as glioma and other non‐neoplastic disorders such as acute brain injury, CNS infection and neurodegenerative disease. Many clinical and animal studies have revealed the dual roles of TRAIL in which it causes damage by inducing cell apoptosis, and confers protection by enhancing both pro‐ and non‐apoptosis effects in different neurological disorders and at different sites or stages. Its pro‐apoptotic effect produces a pro‐survival effect that cannot be underestimated. This review extensively covers in vitro and in vivo experiments and clinical studies investigating TRAIL. It also provides a summary of the current knowledge on the TRAIL signalling pathway and its involvement in pathogenesis, diagnosis and therapeutics of CNS disorders as a basis for future research.
Collapse
Affiliation(s)
- Shiqi Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Huaijun Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Hypermethylation of tumor necrosis factor decoy receptor gene in non-small cell lung cancer. Oncol Lett 2020; 20:155-164. [PMID: 32565943 PMCID: PMC7286129 DOI: 10.3892/ol.2020.11565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 03/06/2020] [Indexed: 01/16/2023] Open
Abstract
Abnormal methylation of the TNFRSF10C and TNFRSF10D genes has been observed in numerous types of cancer; however, no studies have investigated the methylation of these genes in non-small cell lung cancer (NSCLC). The aim of the present study was to investigate the association between TNFRSF10C and TNFRSF10D methylation and NSCLC. Methylation levels of 44 pairs of NSCLC tumor tissues and distant non-tumor tissues were analyzed using quantitative methylation specific PCR and methylation reference percentage values (PMR). The methylation levels of the TNFRSF10C gene in NSCLC tumor tissue samples were significantly higher compared with those in the distant non-tumor tissues (median PMR, 2.73% vs. 0.75%; P=0.013). Subgroup analysis demonstrated that the methylation levels of TNFRSF10C in tumor tissues from male patients were significantly higher compared with those in distant non-tumor tissues (median PMR, 2.73% vs. 0.75%; P=0.041). The levels of TNFRSF10C methylation were also higher in the tumor tissues of patients who were non-smokers compared with their distant non-tumor tissues (median PMR, 2.50% vs. 0.63%; P=0.013). TNFRSF10C methylation levels were higher in the tumor tissues from male patients compared with those from female patients (median PMR, 2.50% vs. 0.63%; P=0.031). However, no significant differences in the methylation levels of the TNFRSF10D gene were observed between the sexes. Using the cBioPortal and The Cancer Genome Atlas lung cancer data, it was demonstrated that TNFRSF10C methylation levels were inversely correlated with TNFRSF10C mRNA expression levels (r=-0.379; P=0.008). In addition, demethylation of lung cancer cell lines A549 and NCI-H1299 using 5'-aza-deoxycytidine further confirmed that TNFRSF10C hypomethylation was associated with significant upregulation of TNFRSF10C mRNA expression levels [A549 fold-change (FC)=8; P=1.0×10-4; NCI-H1299 FC=3.163; P=1.143×10-5]. A dual luciferase reporter gene assay was also performed with the insert of TNFRSF10C promoter region, and the results revealed that the TNFRSF10C gene fragment significantly enhanced the transcriptional activity of the reporter gene compared with that in the control group (FC=1.570; P=0.032). Overall, the results of the present study demonstrated that hypermethylation of TNFRSF10C was associated with NSCLC.
Collapse
|
26
|
Wong SHM, Kong WY, Fang CM, Loh HS, Chuah LH, Abdullah S, Ngai SC. The TRAIL to cancer therapy: Hindrances and potential solutions. Crit Rev Oncol Hematol 2019; 143:81-94. [PMID: 31561055 DOI: 10.1016/j.critrevonc.2019.08.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
Apoptosis is an ordered and orchestrated cellular process that occurs in physiological and pathological conditions. Resistance to apoptosis is a hallmark of virtually all malignancies. Despite being a cause of pathological conditions, apoptosis could be a promising target in cancer treatment. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of TNF cytokine superfamily. It is a potent anti-cancer agent owing to its specific targeting towards cancerous cells, while sparing normal cells, to induce apoptosis. However, resistance occurs either intrinsically or after multiple treatments which may explain why cancer therapy fails. This review summarizes the apoptotic mechanisms via extrinsic and intrinsic apoptotic pathways, as well as the apoptotic resistance mechanisms. It also reviews the current clinically tested recombinant human TRAIL (rhTRAIL) and TRAIL receptor agonists (TRAs) against TRAIL-Receptors, TRAIL-R1 and TRAIL-R2, in which the outcomes of the clinical trials have not been satisfactory. Finally, this review discusses the current strategies in overcoming resistance to TRAIL-induced apoptosis in pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Sonia How Ming Wong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Wei Yang Kong
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Hwei-San Loh
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Syahril Abdullah
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia; UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, 43400 UPM, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|