1
|
Wang E, Jiang Y, Zhao C. Hydroxytyrosol isolation, comparison of synthetic routes and potential biological activities. Food Sci Nutr 2024; 12:6899-6912. [PMID: 39479663 PMCID: PMC11521723 DOI: 10.1002/fsn3.4349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 11/02/2024] Open
Abstract
Hydroxytyrosol (HT) is a polyphenol found in the olive plant (Olea europaea) that has garnered attention from the food, feed, supplement, and pharmaceutical industries. HT has evolved from basic separation and extraction to chemical and biocatalytic synthesis. The yield of HT can reach 1.93 g/L/h through chemical synthesis and 7.7 g/L/h through biocatalysis; however, both methods are subject to inherent limitations. Furthermore, the potential health benefits associated with HT have been highlighted, including its ability to act as an antioxidant, reduce inflammation, combat cancer and obesity, and exert antibacterial and antiviral effects. Its neuroprotective effects, skin protection, and wound healing capabilities are also discussed. Given these remarkable biological properties, HT stands out as one of the most extensively investigated natural phenols. This review highlights future methods and pathways for the synthesis of HT, providing insights based on its bioactivity characteristics, health benefits, and potential future applications.
Collapse
Affiliation(s)
- Enhui Wang
- Beijing Qingyan Boshi Health Management Co., LtdBeijingChina
| | - Yanfei Jiang
- Beijing Qingyan Boshi Health Management Co., LtdBeijingChina
| | - Chunyue Zhao
- Beijing Qingyan Boshi Health Management Co., LtdBeijingChina
| |
Collapse
|
2
|
Vinhas A, Rodrigues MT, Gonçalves AI, Gomes ME. Immunomodulatory Behavior of Tendon Magnetic Cell Sheets can be Modulated in Hypoxic Environments under Magnetic Stimulus. ACS APPLIED MATERIALS & INTERFACES 2024; 16:44440-44450. [PMID: 39143034 DOI: 10.1021/acsami.4c08154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Tissue environments play a crucial role in orchestrating cell behavior, guided by a complex interplay of various factors. Long lasting inflammatory signals compromise tendon homeostasis and promote tissue degeneration, while tissue oxygen levels affect local cells' responses with hypoxic environments influencing apoptosis, inflammatory mediators, and matrix production. Recent works have unveiled the therapeutic potential of pulsed electromagnetic field (PEMF) in modulating inflammatory signals expressed by human tendon cells (hTDCs), and in mitigating the hypoxia-induced effects on the regulation of inflammatory cytokines. Thus, we sought to investigate the role of hypoxic environments, namely, 1 and 2% oxygen tension, in the inflammatory profiles of magnetic cell sheets (magCSs) formed by magnetic nanoparticles internalized in contiguous hTDCs with intact cell-cell junctions and deposited matrix. We also aimed to explore the impact of PEMF over hypoxia-treated magCSs, including IL-1β-primed-magCSs, with the objective of harnessing magnetic stimulation to guide abnormal inflammatory cell responses toward efficient treatments supporting tendon regenerative potential. Our findings revealed that low oxygen tensions amplified the expression of hypoxia-associated genes and of inflammatory markers in IL-1β-primed-magCSs with an involvement of the NF-κB signaling pathway. Encouragingly, when PEMF was applied to IL-1β-primed-magCSs under hypoxic conditions, it successfully modulated inflammatory cues by favoring IL-10 and IL-4, via the NF-κB pathway. These results signify the remarkable potential of PEMF in driving proregenerative strategies and opens up new approaches in tendon therapies, highlighting the transformative impact of immunomodulatory magnetic cell sheets.
Collapse
Affiliation(s)
- Adriana Vinhas
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark─Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
| | - Márcia T Rodrigues
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark─Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark─Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark─Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's─PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
| |
Collapse
|
3
|
Cordonnier C, Mandalasi M, Gigley J, Wohlfert EA, West CM, Blader IJ. The Toxoplasma oxygen-sensing protein, TgPhyA, is required for resistance to interferon gamma-mediated nutritional immunity in mice. PLoS Biol 2024; 22:e3002690. [PMID: 38857298 PMCID: PMC11192375 DOI: 10.1371/journal.pbio.3002690] [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: 05/16/2023] [Revised: 06/21/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
As Toxoplasma gondii disseminates through its host, the parasite must sense and adapt to its environment and scavenge nutrients. Oxygen (O2) is one such environmental factor and cytoplasmic prolyl 4-hydroxylases (PHDs) are evolutionarily conserved O2 cellular sensing proteins that regulate responses to changes in O2 availability. Toxoplasma expresses 2 PHDs. One of them, TgPHYa hydroxylates SKP1, a subunit of the SCF-E3 ubiquitin ligase complex. In vitro, TgPHYa is important for growth at low O2 levels. However, studies have yet to examine the role that TgPHYa or any other pathogen-encoded PHD plays in virulence and disease. Using a type II ME49 Toxoplasma TgPHYa knockout, we report that TgPHYa is important for Toxoplasma virulence and brain cyst formation in mice. We further find that while TgPHYa mutant parasites can establish an infection in the gut, they are unable to efficiently disseminate to peripheral tissues because the mutant parasites are unable to survive within recruited immune cells. Since this phenotype was abrogated in IFNγ knockout mice, we studied how TgPHYa mediates survival in IFNγ-treated cells. We find that TgPHYa is not required for release of parasite-encoded effectors into host cells that neutralize anti-parasitic processes induced by IFNγ. In contrast, we find that TgPHYa is required for the parasite to scavenge tryptophan, which is an amino acid whose levels are decreased after IFNγ up-regulates the tryptophan-catabolizing enzyme, indoleamine dioxygenase (IDO). We further find, relative to wild-type mice, that IDO knockout mice display increased morbidity when infected with TgPHYa knockout parasites. Together, these data identify the first parasite mechanism for evading IFNγ-induced nutritional immunity and highlight a novel role that oxygen-sensing proteins play in pathogen growth and virulence.
Collapse
Affiliation(s)
- Charlotte Cordonnier
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Msano Mandalasi
- Department of Biochemistry & Molecular Biology, Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Jason Gigley
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming, United States of America
| | - Elizabeth A. Wohlfert
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Christopher M. West
- Department of Biochemistry & Molecular Biology, Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| |
Collapse
|
4
|
Yuan X, Ruan W, Bobrow B, Carmeliet P, Eltzschig HK. Targeting hypoxia-inducible factors: therapeutic opportunities and challenges. Nat Rev Drug Discov 2024; 23:175-200. [PMID: 38123660 DOI: 10.1038/s41573-023-00848-6] [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] [Accepted: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Hypoxia-inducible factors (HIFs) are highly conserved transcription factors that are crucial for adaptation of metazoans to limited oxygen availability. Recently, HIF activation and inhibition have emerged as therapeutic targets in various human diseases. Pharmacologically desirable effects of HIF activation include erythropoiesis stimulation, cellular metabolism optimization during hypoxia and adaptive responses during ischaemia and inflammation. By contrast, HIF inhibition has been explored as a therapy for various cancers, retinal neovascularization and pulmonary hypertension. This Review discusses the biochemical mechanisms that control HIF stabilization and the molecular strategies that can be exploited pharmacologically to activate or inhibit HIFs. In addition, we examine medical conditions that benefit from targeting HIFs, the potential side effects of HIF activation or inhibition and future challenges in this field.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Wei Ruan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Anaesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bentley Bobrow
- Department of Emergency Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Holger K Eltzschig
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Outcomes Research Consortium, Cleveland, OH, USA.
| |
Collapse
|
5
|
Noto JM, Piazuelo MB, Romero-Gallo J, Delgado AG, Suarez G, Akritidou K, Girod Hoffman M, Roa JC, Taylor CT, Peek RM. Targeting hypoxia-inducible factor-1 alpha suppresses Helicobacter pylori-induced gastric injury via attenuation of both cag-mediated microbial virulence and proinflammatory host responses. Gut Microbes 2023; 15:2263936. [PMID: 37828903 PMCID: PMC10578190 DOI: 10.1080/19490976.2023.2263936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/24/2023] [Indexed: 10/14/2023] Open
Abstract
Helicobacter pylori-induced inflammation is the strongest known risk factor for gastric adenocarcinoma. Hypoxia-inducible factor-1 (HIF-1α) is a key transcriptional regulator of immunity and carcinogenesis. To examine the role of this mediator within the context of H. pylori-induced injury, we first demonstrated that HIF-1α levels were significantly increased in parallel with the severity of gastric lesions in humans. In interventional studies targeting HIF-1α, H. pylori-infected mice were treated ± dimethyloxalylglycine (DMOG), a prolyl hydroxylase inhibitor that stabilizes HIF-1α. H. pylori significantly increased proinflammatory chemokines/cytokines and inflammation in vehicle-treated mice; however, this was significantly attenuated in DMOG-treated mice. DMOG treatment also significantly decreased function of the H. pylori type IV secretion system (T4SS) in vivo and significantly reduced T4SS-mediated NF-κB activation and IL-8 induction in vitro. These results suggest that prolyl hydroxylase inhibition protects against H. pylori-mediated pathologic responses, and is mediated, in part, via attenuation of H. pylori cag-mediated virulence and suppression of host proinflammatory responses.
Collapse
Affiliation(s)
- Jennifer M. Noto
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Blanca Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Judith Romero-Gallo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G. Delgado
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Giovanni Suarez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Center for Cancer Prevention and Control (CECAN), Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Cormac T. Taylor
- School of Medicine, Systems Biology Ireland and The Conway Institute, University College Dublin, Dublin, Ireland
| | - Richard M. Peek
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
6
|
Wong SL, Kardia E, Vijayan A, Umashankar B, Pandzic E, Zhong L, Jaffe A, Waters SA. Molecular and Functional Characteristics of Airway Epithelium under Chronic Hypoxia. Int J Mol Sci 2023; 24:ijms24076475. [PMID: 37047450 PMCID: PMC10095024 DOI: 10.3390/ijms24076475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
Localized and chronic hypoxia of airway mucosa is a common feature of progressive respiratory diseases, including cystic fibrosis (CF). However, the impact of prolonged hypoxia on airway stem cell function and differentiated epithelium is not well elucidated. Acute hypoxia alters the transcription and translation of many genes, including the CF transmembrane conductance regulator (CFTR). CFTR-targeted therapies (modulators) have not been investigated in vitro under chronic hypoxic conditions found in CF airways in vivo. Nasal epithelial cells (hNECs) derived from eight CF and three non-CF participants were expanded and differentiated at the air-liquid interface (26-30 days) at ambient and 2% oxygen tension (hypoxia). Morphology, global proteomics (LC-MS/MS) and function (barrier integrity, cilia motility and ion transport) of basal stem cells and differentiated cultures were assessed. hNECs expanded at chronic hypoxia, demonstrating epithelial cobblestone morphology and a similar proliferation rate to hNECs expanded at normoxia. Hypoxia-inducible proteins and pathways in stem cells and differentiated cultures were identified. Despite the stem cells' plasticity and adaptation to chronic hypoxia, the differentiated epithelium was significantly thinner with reduced barrier integrity. Stem cell lineage commitment shifted to a more secretory epithelial phenotype. Motile cilia abundance, length, beat frequency and coordination were significantly negatively modulated. Chronic hypoxia reduces the activity of epithelial sodium and CFTR ion channels. CFTR modulator drug response was diminished. Our findings shed light on the molecular pathophysiology of hypoxia and its implications in CF. Targeting hypoxia can be a strategy to augment mucosal function and may provide a means to enhance the efficacy of CFTR modulators.
Collapse
Affiliation(s)
- Sharon L Wong
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Egi Kardia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Abhishek Vijayan
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Bala Umashankar
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Elvis Pandzic
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW 2052, Australia
| | - Adam Jaffe
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW 2052, Australia
| | - Shafagh A Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW 2052, Australia
| |
Collapse
|
7
|
Golub IJ, Ng MK, Conway CA, Vakharia RM, Cannada LK, Kang KK. How does sleep apnea impact outcomes following primary total hip arthroplasty for femoral neck fractures: a matched-control analysis. Arch Orthop Trauma Surg 2023; 143:295-300. [PMID: 34287701 DOI: 10.1007/s00402-021-04070-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/11/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Cross-sectional studies have demonstrated that the prevalence of sleep apnea (SA) to be increasing within the United States. While studies have shown the association of SA and its association on complications following elective orthopedic procedures, well-powered studies investigating its impact in a traumatic setting are limited. The purpose of this study was to determine whether SA patients undergoing primary total hip arthroplasty (THA) for femoral neck fractures have higher rates of: (1) hospital lengths of stay (LOS); (2) readmissions; (3) complications; and (4) healthcare expenditures. METHODS The 100% Medicare Standard Analytical Files was queried from 2005 to 2014 for patients who sustained femoral neck fractures and were treated with primary THA. The study group consisted of patients with concomitant diagnoses of SA, whereas patients without SA served as controls. Study group patients were matched to controls in a 1:5 ratio by age, sex, and various comorbid conditions. Demographics of the cohorts were compared using Pearson's chi-squared analyses, and multivariate logistic regression analyses were used to calculate the odds (OR) of the effects of SA on postoperative outcomes. A p value less than 0.006 was considered to be statistically significant. RESULTS The final query yielded 24,936 patients within the study (n = 4166) and control (n = 20,770) cohorts. SA patients had significantly longer in-hospital LOS (6 vs. 5 days, p < 0.0001) but similar readmission rates (24.12 vs. 20.50%; OR: 1.03, p = 0.476). SA patients had significantly higher frequency and odds of developing medical complications (72.66 vs. 43.85%; OR: 1.57, p < 0.0001), and higher healthcare costs ($22,743.79 vs. $21,572.89, p < 0.0001). CONCLUSION SA is associated with longer in-hospital LOS, higher rates of complications and healthcare expenditures. This study is vital as it can allow orthopaedists to educate patients with SA on the potential complications which may occur following their procedure. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
- Ivan J Golub
- Department of Orthopaedic Surgery, Maimonides Medical Center, 4802 10th Avenue, Brooklyn, NY, 11219, USA
| | - Mitchell K Ng
- Department of Orthopaedic Surgery, Maimonides Medical Center, 4802 10th Avenue, Brooklyn, NY, 11219, USA
| | - Charles A Conway
- Department of Orthopaedic Surgery, Maimonides Medical Center, 4802 10th Avenue, Brooklyn, NY, 11219, USA
| | - Rushabh M Vakharia
- Department of Orthopaedic Surgery, Maimonides Medical Center, 4802 10th Avenue, Brooklyn, NY, 11219, USA.
| | | | - Kevin K Kang
- Department of Orthopaedic Surgery, Maimonides Medical Center, 4802 10th Avenue, Brooklyn, NY, 11219, USA
| |
Collapse
|
8
|
Evaluating Bacterial Pathogenesis Using a Model of Human Airway Organoids Infected with Pseudomonas aeruginosa Biofilms. Microbiol Spectr 2022; 10:e0240822. [PMID: 36301094 PMCID: PMC9769610 DOI: 10.1128/spectrum.02408-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Pseudomonas aeruginosa is one of the leading invasive agents of human pulmonary infection, especially in patients with compromised immunity. Prior studies have used various in vitro models to establish P. aeruginosa infection and to analyze transcriptomic profiles of either the host or pathogen, and yet how much those works are relevant to the genuine human airway still raises doubts. In this study, we cultured and differentiated human airway organoids (HAOs) that recapitulate, to a large extent, the histological and physiological features of the native human mucociliary epithelium. HAOs were then employed as a host model to monitor P. aeruginosa biofilm development. Through dual-species transcriptome sequencing (RNA-seq) analyses, we found that quorum sensing (QS) and several associated protein secretion systems were significantly upregulated in HAO-associated bacteria. Cocultures of HAOs and QS-defective mutants further validated the role of QS in the maintenance of a robust biofilm and disruption of host tissue. Simultaneously, the expression magnitude of multiple inflammation-associated signaling pathways was higher in the QS mutant-infected HAOs, suggesting that QS promotes immune evasion at the transcriptional level. Altogether, modeling infection of HAOs by P. aeruginosa captured several crucial facets in host responses and bacterial pathogenesis, with QS being the most dominant virulence pathway showing profound effects on both bacterial biofilm and host immune responses. Our results revealed that HAOs are an optimal model for studying the interaction between the airway epithelium and bacterial pathogens. IMPORTANCE Human airway organoids (HAOs) are an organotypic model of human airway mucociliary epithelium. The HAOs can closely resemble their origin organ in terms of epithelium architecture and physiological function. Accumulating studies have revealed the great values of the HAO cultures in host-pathogen interaction research. In this study, HAOs were used as a host model to grow Pseudomonas aeruginosa biofilm, which is one of the most common pathogens found in pulmonary infection cases. Dual transcriptome sequencing (RNA-seq) analyses showed that the cocultures have changed the gene expression pattern of both sides significantly and simultaneously. Bacterial quorum sensing (QS), the most upregulated pathway, contributed greatly to biofilm formation, disruption of barrier function, and subversion of host immune responses. Our study therefore provides a global insight into the transcriptomic responses of both P. aeruginosa and human airway epithelium.
Collapse
|
9
|
Kurata Y, Tanaka T, Nangaku M. An evaluation of roxadustat for the treatment of anemia associated with chronic kidney disease. Expert Opin Pharmacother 2021; 23:19-28. [PMID: 34686069 DOI: 10.1080/14656566.2021.1993821] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Anemia is one of the major complications of chronic kidney disease (CKD). Erythropoiesis-stimulating agents (ESAs) have been the mainstay of renal anemia treatment. However, there are several safety drawbacks, and a safer and more effective alternative treatment has been sought. AREAS COVERED Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PHIs) have been developed as a novel orally active therapeutic agent for renal anemia. HIF-PHIs stimulate endogenous EPO and optimize iron utilization. Roxadustat is a first-in-class HIF-PHI for the treatment of anemia in CKD patients approved in China, Japan, South Korea, and Chile. The authors herein evaluate the pharmacology of roxadustat and give their expert perspectives on its use. EXPERT OPINION Phase 3 clinical trials have demonstrated that roxadustat effectively increases and maintains hemoglobin (Hb) levels in both nondialysis-dependent and dialysis-dependent CKD patients. Roxadustat also improved iron metabolism and reduced intravenous (IV) iron requirements. However, pooled analyses of phase 3 studies have revealed frequent thromboembolic events in the roxadustat group, which might be attributed to rapid changes in Hb and inadequate iron supplementation. Roxadustat is an attractive alternative treatment especially for patients with ESA hyporesponsive due to impaired iron utilization, and so appropriate selection of target patients and its proper use are crucially important.
Collapse
Affiliation(s)
- Yu Kurata
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Malkov MI, Lee CT, Taylor CT. Regulation of the Hypoxia-Inducible Factor (HIF) by Pro-Inflammatory Cytokines. Cells 2021; 10:cells10092340. [PMID: 34571989 PMCID: PMC8466990 DOI: 10.3390/cells10092340] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 12/28/2022] Open
Abstract
Hypoxia and inflammation are frequently co-incidental features of the tissue microenvironment in a wide range of inflammatory diseases. While the impact of hypoxia on inflammatory pathways in immune cells has been well characterized, less is known about how inflammatory stimuli such as cytokines impact upon the canonical hypoxia-inducible factor (HIF) pathway, the master regulator of the cellular response to hypoxia. In this review, we discuss what is known about the impact of two major pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), on the regulation of HIF-dependent signaling at sites of inflammation. We report extensive evidence for these cytokines directly impacting upon HIF signaling through the regulation of HIF at transcriptional and post-translational levels. We conclude that multi-level crosstalk between inflammatory and hypoxic signaling pathways plays an important role in shaping the nature and degree of inflammation occurring at hypoxic sites.
Collapse
Affiliation(s)
- Mykyta I. Malkov
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Chee Teik Lee
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Cormac T. Taylor
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Correspondence:
| |
Collapse
|
11
|
Del Mar Cendra M, Torrents E. Differential adaptability between reference strains and clinical isolates of Pseudomonas aeruginosa into the lung epithelium intracellular lifestyle. Virulence 2021; 11:862-876. [PMID: 32697923 PMCID: PMC7549915 DOI: 10.1080/21505594.2020.1787034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Intracellular invasion is an advantageous mechanism used by pathogens to evade host defense and antimicrobial therapy. In patients, the intracellular microbial lifestyle can lead to infection persistence and recurrence, thus worsening outcomes. Lung infections caused by Pseudomonas aeruginosa, especially in cystic fibrosis (CF) patients, are often aggravated by intracellular invasion and persistence of the pathogen. Proliferation of the infectious species relies on a continuous deoxyribonucleotide (dNTP) supply, for which the ribonucleotide reductase enzyme (RNR) is the unique provider. The large genome plasticity of P. aeruginosa and its ability to rapidly adapt to different environments are challenges for studying the pathophysiology associated with this type of infection. Using different reference strains and clinical isolates of P. aeruginosa independently combined with alveolar (A549) and bronchial (16HBE14o- and CF-CFBE41o-) epithelial cells, we analyzed host-pathogen interactions and intracellular bacterial persistence with the aim of determining a cell type-directed infection promoted by the P. aeruginosa strains. The oscillations in cellular toxicity and oxygen consumption promoted by the intracellular persistence of the strains were also analyzed among the different infectious lung models. Significantly, we identified class II RNR as the enzyme that supplies dNTPs to intracellular P. aeruginosa. This discovery could contribute to the development of RNR-targeted strategies against the chronicity occurring in this type of lung infection. Overall our study demonstrates that the choice of bacterial strain is critical to properly study the type of infectious process with relevant translational outcomes.
Collapse
Affiliation(s)
- Maria Del Mar Cendra
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Science and Technology , Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Science and Technology , Barcelona, Spain.,Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona , Barcelona, Spain
| |
Collapse
|
12
|
Lee JY, Stevens RP, Kash M, Alexeyev MF, Balczon R, Zhou C, Renema P, Koloteva A, Kozhukhar N, Pastukh V, Gwin MS, Voth S, deWeever A, Wagener BM, Pittet JF, Eslaamizaad Y, Siddiqui W, Nawaz T, Clarke C, Fouty BW, Audia JP, Alvarez DF, Stevens T. Carbonic Anhydrase IX and Hypoxia Promote Rat Pulmonary Endothelial Cell Survival During Infection. Am J Respir Cell Mol Biol 2021; 65:630-645. [PMID: 34251286 DOI: 10.1165/rcmb.2020-0537oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Low tidal volume ventilation protects the lung in mechanically ventilated patients. The impact of the accompanying permissive hypoxemia and hypercapnia on endothelial cell recovery from injury is poorly understood. Carbonic anhydrase IX (CA IX) is expressed in pulmonary microvascular endothelial cells (PMVECs), where it contributes to CO2 and pH homeostasis, bioenergetics and angiogenesis. We hypothesized that CA IX is important for PMVEC survival, and CA IX expression and release from PMVECs are increased during infection. While plasma CA IX was unchanged in human and rat pneumonia, there was a trend towards increasing CA IX in bronchoalveolar fluid of mechanically ventilated critically ill pneumonia patients and a significant increase in CA IX in lung tissue lysate of rat pneumonia. To investigate functional implications of the lung CA IX increase, we generated PMVEC cell lines harboring domain-specific CA IX mutations. Using these cells, we found that infection promotes intracellular expression, release and metalloproteinase-mediated extracellular cleavage of CA IX in PMVECs. Intracellular domain deletion uniquely impaired CA IX membrane localization. Loss of the CA IX intracellular domain promoted cell death following infection, suggesting the important role of intracellular domain in PMVEC survival. We also found that hypoxia improves survival, whereas hypercapnia reverses the protective effect of hypoxia, during infection. Thus, we report that: (1) CA IX increases in rat pneumonia lung; and, (2) the CA IX intracellular domain and hypoxia promote PMVEC survival during infection.
Collapse
Affiliation(s)
- Ji Young Lee
- University of South Alabama, 5557, Mobile, Alabama, United States;
| | - Reece P Stevens
- University of South Alabama, 5557, Mobile, Alabama, United States
| | - Mary Kash
- University of South Alabama, 5557, Mobile, Alabama, United States
| | | | - Ronald Balczon
- University of South Alabama, 5557, Biochemistry and Molecular Biology, Mobile, Alabama, United States
| | - Chun Zhou
- University of South Alabama, 5557, Mobile, Alabama, United States
| | - Phoibe Renema
- University of South Alabama, 5557, Mobile, Alabama, United States
| | - Anna Koloteva
- University of South Alabama, 5557, Mobile, Alabama, United States
| | | | | | - Meredith S Gwin
- University of South Alabama, 5557, Physiology and Cell Biology, Mobile, Alabama, United States
| | - Sarah Voth
- University of South Alabama, 5557, Physiology and Cell Biology, Mobile, Alabama, United States
| | - Althea deWeever
- University of South Alabama College of Medicine, 12214, Physiology and Cell Biology, Mobile, Alabama, United States
| | - Brant M Wagener
- The University of Alabama at Birmingham, 9968, Department of Anesthesiology and Perioperative Medicine, Birmingham, Alabama, United States
| | - Jean-François Pittet
- The University of Alabama at Birmingham, 9968, Department of Anesthesiology and Perioperative Medicine, Birmingham, Alabama, United States
| | | | - Waqar Siddiqui
- University of South Alabama, 5557, Mobile, Alabama, United States
| | - Talha Nawaz
- University of South Alabama, 5557, Mobile, Alabama, United States
| | | | - Brian W Fouty
- University of South Alabama, 5557, Mobile, Alabama, United States
| | - Jonathon P Audia
- University of South Alabama, 5557, Mobile, Alabama, United States
| | - Diego F Alvarez
- Sam Houston State University, 4038, Huntsville, Texas, United States
| | - Troy Stevens
- University of South Alabama, 5557, Physiology and Cell Biology, Mobile, Alabama, United States
| |
Collapse
|
13
|
Karan S, Cho MY, Lee H, Lee H, Park HS, Sundararajan M, Sessler JL, Hong KS. Near-Infrared Fluorescent Probe Activated by Nitroreductase for In Vitro and In Vivo Hypoxic Tumor Detection. J Med Chem 2021; 64:2971-2981. [PMID: 33711229 DOI: 10.1021/acs.jmedchem.0c02162] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tumor hypoxia is correlated with increased resistance to chemotherapy and poor overall prognoses across a number of cancer types. We present here a cancer cell-selective and hypoxia-responsive probe (fol-BODIPY) designed on the basis of density functional theory (DFT)-optimized quantum chemical calculations. The fol-BODIPY probe was found to provide a rapid fluorescence "off-on" response to hypoxia relative to controls, which lack the folate or nitro-benzyl moieties. In vitro confocal microscopy and flow cytometry analyses, as well as in vivo near-infrared optical imaging of CT26 solid tumor-bearing mice, provided support for the contention that fol-BODIPY is more readily accepted by folate receptor-positive CT26 cancer cells and provides a superior fluorescence "off-on" signal under hypoxic conditions than the controls. Based on the findings of this study, we propose that fol-BODIPY may serve as a tumor-targeting, hypoxia-activatable probe that allows for direct cancer monitoring both in vitro and in vivo.
Collapse
Affiliation(s)
- Sanu Karan
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Mi Young Cho
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Hyunseung Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Hwunjae Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Hye Sun Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Mahesh Sundararajan
- Theoretical Chemistry Section, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Kwan Soo Hong
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
14
|
Page LK, Staples KJ, Spalluto CM, Watson A, Wilkinson TMA. Influence of Hypoxia on the Epithelial-Pathogen Interactions in the Lung: Implications for Respiratory Disease. Front Immunol 2021; 12:653969. [PMID: 33868294 PMCID: PMC8044850 DOI: 10.3389/fimmu.2021.653969] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Under normal physiological conditions, the lung remains an oxygen rich environment. However, prominent regions of hypoxia are a common feature of infected and inflamed tissues and many chronic inflammatory respiratory diseases are associated with mucosal and systemic hypoxia. The airway epithelium represents a key interface with the external environment and is the first line of defense against potentially harmful agents including respiratory pathogens. The protective arsenal of the airway epithelium is provided in the form of physical barriers, and the production of an array of antimicrobial host defense molecules, proinflammatory cytokines and chemokines, in response to activation by receptors. Dysregulation of the airway epithelial innate immune response is associated with a compromised immunity and chronic inflammation of the lung. An increasing body of evidence indicates a distinct role for hypoxia in the dysfunction of the airway epithelium and in the responses of both innate immunity and of respiratory pathogens. Here we review the current evidence around the role of tissue hypoxia in modulating the host-pathogen interaction at the airway epithelium. Furthermore, we highlight the work needed to delineate the role of tissue hypoxia in the pathophysiology of chronic inflammatory lung diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease in addition to novel respiratory diseases such as COVID-19. Elucidating the molecular mechanisms underlying the epithelial-pathogen interactions in the setting of hypoxia will enable better understanding of persistent infections and complex disease processes in chronic inflammatory lung diseases and may aid the identification of novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Lee K. Page
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Karl J. Staples
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - C. Mirella Spalluto
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
- Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Tom M. A. Wilkinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
15
|
Lauer AN, Scholtysik R, Beineke A, Baums CG, Klose K, Valentin-Weigand P, Ishikawa H, Schroten H, Klein-Hitpass L, Schwerk C. A Comparative Transcriptome Analysis of Human and Porcine Choroid Plexus Cells in Response to Streptococcus suis Serotype 2 Infection Points to a Role of Hypoxia. Front Cell Infect Microbiol 2021; 11:639620. [PMID: 33763387 PMCID: PMC7982935 DOI: 10.3389/fcimb.2021.639620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/01/2021] [Indexed: 11/14/2022] Open
Abstract
Streptococcus suis (S. suis) is an important opportunistic pathogen, which can cause septicemia and meningitis in pigs and humans. Previous in vivo observations in S. suis-infected pigs revealed lesions at the choroid plexus (CP). In vitro experiments with primary porcine CP epithelial cells (PCPEC) and human CP epithelial papilloma (HIBCPP) cells demonstrated that S. suis can invade and traverse the CP epithelium, and that the CP contributes to the inflammatory response via cytokine expression. Here, next generation sequencing (RNA-seq) was used to compare global transcriptome profiles of PCPEC and HIBCPP cells challenged with S. suis serotype (ST) 2 infected in vitro, and of pigs infected in vivo. Identified differentially expressed genes (DEGs) were, amongst others, involved in inflammatory responses and hypoxia. The RNA-seq data were validated via quantitative PCR of selected DEGs. Employing Gene Set Enrichment Analysis (GSEA), 18, 28, and 21 enriched hallmark gene sets (GSs) were identified for infected HIBCPP cells, PCPEC, and in the CP of pigs suffering from S. suis ST2 meningitis, respectively, of which eight GSs overlapped between the three different sample sets. The majority of these GSs are involved in cellular signaling and pathways, immune response, and development, including inflammatory response and hypoxia. In contrast, suppressed GSs observed during in vitro and in vivo S. suis ST2 infections included those, which were involved in cellular proliferation and metabolic processes. This study suggests that similar cellular processes occur in infected human and porcine CP epithelial cells, especially in terms of inflammatory response.
Collapse
Affiliation(s)
- Alexa N Lauer
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rene Scholtysik
- Institute for Cell Biology, University Hospital Essen, Essen, Germany
| | - Andreas Beineke
- Institute for Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Christoph Georg Baums
- Faculty of Veterinary Medicine, Institute of Bacteriology and Mycology, Leipzig University, Leipzig, Germany
| | - Kristin Klose
- Faculty of Veterinary Medicine, Institute of Veterinary Pathology, Leipzig University, Leipzig, Germany
| | | | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
16
|
Chen Y, Gaber T. Hypoxia/HIF Modulates Immune Responses. Biomedicines 2021; 9:biomedicines9030260. [PMID: 33808042 PMCID: PMC8000289 DOI: 10.3390/biomedicines9030260] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
Oxygen availability varies throughout the human body in health and disease. Under physiological conditions, oxygen availability drops from the lungs over the blood stream towards the different tissues into the cells and the mitochondrial cavities leading to physiological low oxygen conditions or physiological hypoxia in all organs including primary lymphoid organs. Moreover, immune cells travel throughout the body searching for damaged cells and foreign antigens facing a variety of oxygen levels. Consequently, physiological hypoxia impacts immune cell function finally controlling innate and adaptive immune response mainly by transcriptional regulation via hypoxia-inducible factors (HIFs). Under pathophysiological conditions such as found in inflammation, injury, infection, ischemia and cancer, severe hypoxia can alter immune cells leading to dysfunctional immune response finally leading to tissue damage, cancer progression and autoimmunity. Here we summarize the effects of physiological and pathophysiological hypoxia on innate and adaptive immune activity, we provide an overview on the control of immune response by cellular hypoxia-induced pathways with focus on the role of HIFs and discuss the opportunity to target hypoxia-sensitive pathways for the treatment of cancer and autoimmunity.
Collapse
Affiliation(s)
- Yuling Chen
- Charité—Universitätsmedizin Berlin, Corporate Ember of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany;
| | - Timo Gaber
- Charité—Universitätsmedizin Berlin, Corporate Ember of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany;
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-513364
| |
Collapse
|
17
|
Mechanisms controlling bacterial infection in myeloid cells under hypoxic conditions. Cell Mol Life Sci 2020; 78:1887-1907. [PMID: 33125509 PMCID: PMC7966188 DOI: 10.1007/s00018-020-03684-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/08/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
Various factors of the tissue microenvironment such as the oxygen concentration influence the host-pathogen interaction. During the past decade, hypoxia-driven signaling via hypoxia-inducible factors (HIF) has emerged as an important factor that affects both the pathogen and the host. In this chapter, we will review the current knowledge of this complex interplay, with a particular emphasis given to the impact of hypoxia and HIF on the inflammatory and antimicrobial activity of myeloid cells, the bacterial responses to hypoxia and the containment of bacterial infections under oxygen-limited conditions. We will also summarize how low oxygen concentrations influence the metabolism of neutrophils, macrophages and dendritic cells. Finally, we will discuss the consequences of hypoxia and HIFα activation for the invading pathogen, with a focus on Pseudomonas aeruginosa, Mycobacterium tuberculosis, Coxiella burnetii, Salmonella enterica and Staphylococcus aureus. This includes a description of the mechanisms and microbial factors, which the pathogens use to sense and react to hypoxic conditions.
Collapse
|
18
|
Van den Bossche S, Vandeplassche E, Ostyn L, Coenye T, Crabbé A. Bacterial Interference With Lactate Dehydrogenase Assay Leads to an Underestimation of Cytotoxicity. Front Cell Infect Microbiol 2020; 10:494. [PMID: 33042868 PMCID: PMC7523407 DOI: 10.3389/fcimb.2020.00494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Models to study host-pathogen interactions in vitro are an important tool for investigating the infectious disease process and evaluating the efficacy of antimicrobial compounds. In these models, the viability of mammalian cells is often determined using the lactate dehydrogenase (LDH) cytotoxicity assay. In the present study we evaluated whether bacteria could interfere with the LDH assay. As a model for host-pathogen interactions, we co-cultured lung epithelial cells with eight bacteria encountered in the lower respiratory tract. We show that LDH activity is affected by Pseudomonas aeruginosa, Klebsiella pneumoniae, Stenotrophomonas maltophilia, and Streptococcus pneumoniae, and that this depends on the density of the start inoculum and the duration of infection. Two different mechanisms were discovered through which bacteria interfered with LDH activity, i.e., acidification of the cell culture medium (by K. pneumoniae and S. pneumoniae) and protease production (by P. aeruginosa and S. maltophilia). In addition, we developed and validated a modified protocol to evaluate cytotoxicity using the LDH assay, where bacterial interference with LDH quantification is avoided.
Collapse
Affiliation(s)
| | - Eva Vandeplassche
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Lisa Ostyn
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
19
|
Bertelsen A, Elborn SJ, Schock BC. Toll like Receptor signalling by Prevotella histicola activates alternative NF-κB signalling in Cystic Fibrosis bronchial epithelial cells compared to P. aeruginosa. PLoS One 2020; 15:e0235803. [PMID: 33031374 PMCID: PMC7544055 DOI: 10.1371/journal.pone.0235803] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cystic Fibrosis (CF), caused by mutations affecting the CFTR gene, is characterised by viscid secretions in multiple organ systems. CF airways contain thick mucus, creating a gradient of hypoxia, which promotes the establishment of polymicrobial infection. Such inflammation predisposes to further infection, a self-perpetuating cycle in mediated by NF-κB. Anaerobic Gram-negative Prevotella spp. are found in sputum from healthy volunteers and CF patients and in CF lungs correlate with reduced levels of inflammation. Prevotella histicola (P. histicola) can suppress murine lung inflammation, however, no studies have examined the role of P. histicola in modulating infection and inflammation in the CF airways. We investigated innate immune signalling and NF-kB activation in CF epithelial cells CFBE41o- in response to clinical stains of P. histicola and Pseudomonas aeruginosa (P. aeruginosa). Toll-Like Receptor (TLR) expressing HEK-293 cells and siRNA assays for TLRs and IKKα were used to confirm signalling pathways. We show that P. histicola infection activated the alternative NF-kB signalling pathway in CF bronchial epithelial cells inducing HIF-1α protein. TLR5 signalling was responsible for the induction of the alternative NF-kB pathway through phosphorylation of IKKα. The induction of transcription factor HIF-1α was inversely associated with the induction of the alternative NF-kB pathway and knockdown of IKKα partially restored canonical NF-kB activation in response to P. histicola. This study demonstrates that different bacterial species in the respiratory microbiome can contribute differently to inflammation, either by activating inflammatory cascades (P. aeruginosa) or by muting the inflammatory response by modulating similar or related pathways (P. histicola). Further work is required to assess the complex interactions of the lung microbiome in response to mixed bacterial infections and their effects in people with CF.
Collapse
Affiliation(s)
- Anne Bertelsen
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Stuart J. Elborn
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, United Kingdom
- Imperial College London, London, United Kingdom
| | - Bettina C. Schock
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Yang YY, Lin CJ, Wang CC, Chen CM, Kao WJ, Chen YH. Consecutive Hypoxia Decreases Expression of NOTCH3, HEY1, CC10, and FOXJ1 via NKX2-1 Downregulation and Intermittent Hypoxia-Reoxygenation Increases Expression of BMP4, NOTCH1, MKI67, OCT4, and MUC5AC via HIF1A Upregulation in Human Bronchial Epithelial Cells. Front Cell Dev Biol 2020; 8:572276. [PMID: 33015064 PMCID: PMC7500169 DOI: 10.3389/fcell.2020.572276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/17/2020] [Indexed: 01/11/2023] Open
Abstract
Previous studies have shown that the experimental models of hypoxia-reoxygenation (H/R) mimics the physiological conditions of ischemia-reperfusion and induce oxidative stress and injury in various types of organs, tissues, and cells, both in vivo and in vitro, including human lung adenocarcinoma epithelial cells. Nonetheless, it had not been reported whether H/R affected proliferation, apoptosis, and expression of stem/progenitor cell markers in the bronchial epithelial cells. In this study, we investigated differential effects of consecutive hypoxia and intermittent 24/24-h cycles of H/R on human bronchial epithelial (HBE) cells derived from the same-race and age-matched healthy subjects (i.e., NHBE) and subjects with chronic obstructive pulmonary disease (COPD) (i.e., DHBE). To analyze gene/protein expression during differentiation, both the NHBE and DHBE cells at the 2nd passage were cultured at the air-liquid interface (ALI) in the differentiation medium under normoxia for 3 days, followed by either culturing under hypoxia (1% O2) for consecutively 9 days and then returning to normoxia for another 9 days, or culturing under 24/24-h cycles of H/R (i.e., 24 h of 1% O2 followed by 24 h of 21% O2, repetitively) for 18 days in total, so that all differentiating HBE cells were exposed to hypoxia for a total of 9 days. In both the normal and diseased HBE cells, intermittent H/R significantly increased HIF1A, BMP4, NOTCH1, MKI67, OCT4, and MUC5AC expression, while consecutive hypoxia significantly decreased NKX2-1, NOTCH3, HEY1, CC10, and FOXJ1 expression. Inhibition of HIF1A or NKX2-1 expression by siRNA transfection respectively decreased BMP4/NOTCH1/MKI67/OCT4/MUC5AC and NOTCH3/HEY1/CC10/FOXJ1 expression in the HBE cells cultured under intermittent H/R to the same levels under normoxia. Overexpression of NKX2-1 via cDNA transfection caused more than 2.8-fold increases in NOTCH3, HEY1, and FOXJ1 mRNA levels in the HBE cells cultured under consecutive hypoxia compared to the levels under normoxia. Taken together, our results show for the first time that consecutive hypoxia decreased expression of the co-regulated gene module NOTCH3/HEY1/CC10 and the ciliogenesis-inducing transcription factor gene FOXJ1 via NKX2-1 mRNA downregulation, while intermittent H/R increased expression of the co-regulated gene module BMP4/NOTCH1/MKI67/OCT4 and the predominant airway mucin gene MUC5AC via HIF1A mRNA upregulation.
Collapse
Affiliation(s)
- Yung-Yu Yang
- Department of General Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Ju Lin
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Chin Wang
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.,Section of Respiratory Therapy, Rueifang Miner Hospital, New Taipei City, Taiwan
| | - Chieh-Min Chen
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Jen Kao
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Hui Chen
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
21
|
Molecular Action of Hydroxytyrosol in Wound Healing: An In Vitro Evidence-Based Review. Biomolecules 2020; 10:biom10101397. [PMID: 33008084 PMCID: PMC7600962 DOI: 10.3390/biom10101397] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 01/09/2023] Open
Abstract
Hydroxytyrosol (HT) is an essential molecule isolated from the phenolic fraction of olive (Olea europaea). HT has been implicated for its health-stimulating effect mainly due to its antioxidative capacity. The current review summarises and discusses the available evidence, related to HT activities in wound healing enhancement. The literature search of related articles published within the year 2010 to 2020 was conducted using Medline via Ebscohost, Scopus, and Google Scholar databases. Studies were limited to in vitro research regarding the role of HT in wound closure, including anti-inflammation, antimicrobial, antioxidative, and its direct effect to the cells involved in wound healing. The literature search revealed 7136 potentially relevant records were obtained from the database search. Through the screening process, 13 relevant in vitro studies investigating the role of HT in wound repair were included. The included studies reported a proangiogenic, antioxidative, antiaging, anti-inflammatory and antimicrobial effect of HT. The current in vitro evidence-based review highlights the cellular and molecular action of HT in influencing positive outcomes toward wound healing. Based on this evidence, HT is a highly recommended bioactive compound to be used as a pharmaceutical product for wound care applications.
Collapse
|
22
|
Abstract
Recent years have witnessed an emergence of interest in understanding metabolic changes associated with immune responses, termed immunometabolism. As oxygen is central to all aerobic metabolism, hypoxia is now recognized to contribute fundamentally to inflammatory and immune responses. Studies from a number of groups have implicated a prominent role for oxygen metabolism and hypoxia in innate immunity of healthy tissue (physiologic hypoxia) and during active inflammation (inflammatory hypoxia). This inflammatory hypoxia emanates from a combination of recruited inflammatory cells (e.g., neutrophils, eosinophils, and monocytes), high rates of oxidative metabolism, and the activation of multiple oxygen-consuming enzymes during inflammation. These localized shifts toward hypoxia have identified a prominent role for the transcription factor hypoxia-inducible factor (HIF) in the regulation of innate immunity. Such studies have provided new and enlightening insight into our basic understanding of immune mechanisms, and extensions of these findings have identified potential therapeutic targets. In this review, we summarize recent literature around the topic of innate immunity and mucosal hypoxia with a focus on transcriptional responses mediated by HIF.
Collapse
Affiliation(s)
- Sean P Colgan
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA;
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Glenn T Furuta
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
23
|
Osada-Oka M, Goda N, Saiga H, Yamamoto M, Takeda K, Ozeki Y, Yamaguchi T, Soga T, Tateishi Y, Miura K, Okuzaki D, Kobayashi K, Matsumoto S. Metabolic adaptation to glycolysis is a basic defense mechanism of macrophages for Mycobacterium tuberculosis infection. Int Immunol 2020; 31:781-793. [PMID: 31201418 PMCID: PMC6839748 DOI: 10.1093/intimm/dxz048] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 06/26/2019] [Indexed: 12/17/2022] Open
Abstract
Macrophages are major components of tuberculosis (TB) granulomas and are responsible for host defenses against the intracellular pathogen, Mycobacterium tuberculosis. We herein showed the strong expression of hypoxia-inducible factor-1α (HIF-1α) in TB granulomas and more rapid death of HIF-1α-conditional knockout mice than wild-type (WT) mice after M. tuberculosis infection. Although interferon-γ (IFN-γ) is a critical host-protective cytokine against intracellular pathogens, HIF-1-deficient macrophages permitted M. tuberculosis growth even after activation with IFN-γ. These results prompted us to investigate the role of HIF-1α in host defenses against infection. We found that the expression of lactate dehydrogenase-A (LDH-A) was controlled by HIF-1α in M. tuberculosis-infected macrophages IFN-γ independently. LDH-A is an enzyme that converts pyruvate to lactate and we found that the intracellular level of pyruvate in HIF-1α-deficient bone marrow-derived macrophages (BMDMs) was significantly higher than in WT BMDMs. Intracellular bacillus replication was enhanced by an increase in intracellular pyruvate concentrations, which were decreased by LDH-A. Mycobacteria in phagosomes took up exogenous pyruvate more efficiently than glucose, and used it as the feasible carbon source for intracellular growth. These results demonstrate that HIF-1α prevents the hijacking of pyruvate in macrophages, making it a fundamental host-protective mechanism against M. tuberculosis.
Collapse
Affiliation(s)
- Mayuko Osada-Oka
- Food Hygiene and Environmental Health, Graduate School of Life and Environmental Science, Kyoto Prefectural University, Kyoto, Kyoto, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical BioScience, Waseda University School of Advanced Science and Engineering, Shinjuku-ku, Tokyo, Japan
| | - Hiroyuki Saiga
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuriko Ozeki
- Department of Bacteriology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Takehiro Yamaguchi
- Department of Bacteriology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Yu Tateishi
- Department of Applied Pharmacology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Osaka, Japan
| | - Katsuyuki Miura
- Department of Applied Pharmacology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kazuo Kobayashi
- Division of Public Health, Osaka Institute of Public Health, Osaka, Osaka, Japan
| | - Sohkichi Matsumoto
- Department of Bacteriology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| |
Collapse
|
24
|
Vanderhaeghen T, Vandewalle J, Libert C. Hypoxia-inducible factors in metabolic reprogramming during sepsis. FEBS J 2020; 287:1478-1495. [PMID: 31970890 DOI: 10.1111/febs.15222] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
Sepsis is a highly heterogeneous syndrome that is caused by an imbalanced host response to infection. Despite huge investments, sepsis remains a contemporary threat with significant burden on health systems. Vascular dysfunction and elevated oxygen consumption by highly metabolically active immune cells result in tissue hypoxia during inflammation. The transcription factor hypoxia-inducible factor-1a (HIF1α), and its family members, plays an important role in cellular metabolism and adaptation to cellular stress caused by hypoxia. In this review, we discuss the role of HIF in sepsis. We show possible mechanisms by which the inflammatory response activated during sepsis affects the HIF pathway. The activated HIF pathway in turn changes the metabolism of both innate and adaptive immune cells. As HIF expression in leukocytes of septic patients can be directly linked with mortality, we discuss multiple ways of interfering with the HIF signaling pathway.
Collapse
Affiliation(s)
- Tineke Vanderhaeghen
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Jolien Vandewalle
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Belgium
| |
Collapse
|
25
|
Schaible B, Crifo B, Schaffer K, Taylor CT. The putative bacterial oxygen sensor Pseudomonas prolyl hydroxylase (PPHD) suppresses antibiotic resistance and pathogenicity in Pseudomonas aeruginosa. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49879-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
26
|
Schaible B, Crifo B, Schaffer K, Taylor CT. The putative bacterial oxygen sensor Pseudomonas prolyl hydroxylase (PPHD) suppresses antibiotic resistance and pathogenicity in Pseudomonas aeruginosa. J Biol Chem 2019; 295:1195-1201. [PMID: 31826919 DOI: 10.1074/jbc.ra119.010033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/01/2019] [Indexed: 01/27/2023] Open
Abstract
Pseudomonas aeruginosa is an extracellular opportunistic bacterial pathogen commonly associated with infectious complications in susceptible individuals, such as those with underlying diseases including HIV/AIDS and cystic fibrosis. Antibiotic resistance in multiple strains of P. aeruginosa is a rapidly developing clinical problem. We have previously demonstrated that the oxygen levels at the site of P. aeruginosa infection can strongly influence virulence and antibiotic resistance in this pathogen, although the oxygen-sensing and -signaling mechanisms underpinning this response have remained unknown. In this study, we investigated the potential role of the putative oxygen sensor Pseudomonas prolyl hydroxylase (PPHD) in the control of virulence and antibiotic resistance in P. aeruginosa We found that a P. aeruginosa strain lacking PPHD (PAO310) exhibits increased virulence associated with increased bacterial motility. Furthermore, PPHD-deficient P. aeruginosa displayed enhanced antibiotic resistance against tetracycline through increased expression of the xenobiotic transporters mexEF-oprN and MexXY. Of note, the effect of the PPHD knockout on antibiotic resistance was phenocopied in bacteria exposed to atmospheric hypoxia. We conclude that PPHD is a putative bacterial oxygen sensor that may link microenvironmental oxygen levels to virulence and antibiotic resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Bettina Schaible
- Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Bianca Crifo
- Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kirsten Schaffer
- Department of Clinical Microbiology, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Cormac T Taylor
- Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
27
|
Renga G, Oikonomou V, Moretti S, Stincardini C, Bellet MM, Pariano M, Bartoli A, Brancorsini S, Mosci P, Finocchi A, Rossi P, Costantini C, Garaci E, Goldstein AL, Romani L. Thymosin β4 promotes autophagy and repair via HIF-1α stabilization in chronic granulomatous disease. Life Sci Alliance 2019; 2:2/6/e201900432. [PMID: 31719116 PMCID: PMC6851533 DOI: 10.26508/lsa.201900432] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
This study demonstrates that thymosin β4 stabilizes HIF-1a to promote autophagy and up-regulate genes involved in tissue and mucosal barrier protection in chronic granulomatous disease. Chronic granulomatous disease (CGD) is a genetic disorder of the NADPH oxidase characterized by increased susceptibility to infections and hyperinflammation associated with defective autophagy and increased inflammasome activation. Herein, we demonstrate that thymosin β4 (Tβ4), a g-actin sequestering peptide with multiple and diverse intracellular and extracellular activities affecting inflammation, wound healing, fibrosis, and tissue regeneration, promoted in human and murine cells noncanonical autophagy, a form of autophagy associated with phagocytosis and limited inflammation via the death-associated protein kinase 1. We further show that the hypoxia inducible factor-1 (HIF-1)α was underexpressed in CGD but normalized by Tβ4 to promote autophagy and up-regulate genes involved in mucosal barrier protection. Accordingly, inflammation and granuloma formation were impaired and survival increased in CGD mice with colitis or aspergillosis upon Tβ4 treatment or HIF-1α stabilization. Thus, the promotion of endogenous pathways of inflammation resolution through HIF-1α stabilization is druggable in CGD by Tβ4.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Paolo Mosci
- Internal Medicine, Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Andrea Finocchi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Paolo Rossi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Rome, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, the George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
28
|
Talla U, Bozonet SM, Parker HA, Hampton MB, Vissers MCM. Prolonged exposure to hypoxia induces an autophagy-like cell survival program in human neutrophils. J Leukoc Biol 2019; 106:1367-1379. [PMID: 31412152 DOI: 10.1002/jlb.4a0319-079rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/14/2019] [Accepted: 07/14/2019] [Indexed: 12/13/2022] Open
Abstract
Neutrophils contribute to low oxygen availability at inflammatory sites through the generation of reactive oxidants. They are also functionally affected by hypoxia, which delays neutrophil apoptosis. However, the eventual fate of neutrophils in hypoxic conditions is unknown and this is important for their effective clearance and the resolution of inflammation. We have monitored the survival and function of normal human neutrophils exposed to hypoxia over a 48 h period. Apoptosis was delayed, and the cells remained intact even at 48 h. However, hypoxia promoted significant changes in neutrophil morphology with the appearance of many new cytoplasmic vesicles, often containing cell material, within 5 hours of exposure to low O2 . This coincided with an increase in LC3B-II expression, indicative of autophagosome formation and an autophagy-like process. In hypoxic conditions, neutrophils preferentially lost myeloperoxidase, a marker of azurophil granules. Short-term (2 h) hypoxic exposure resulted in sustained potential to generate superoxide when O2 was restored, but the capacity for oxidant production was lost with longer periods of hypoxia. Phagocytic ability was unchanged by hypoxia, and bacterial killing by neutrophils in both normoxic and hypoxic conditions was substantially diminished after 24 hours. However, pre-exposure to hypoxia resulted in an enhanced ability to kill bacteria by oxidant-independent mechanisms. Our data provide the first evidence for hypoxia as a driver of neutrophil autophagy that can influence the function and ultimate fate of these cells, including their eventual clearance and the resolution of inflammation.
Collapse
Affiliation(s)
- Usharani Talla
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Stephanie M Bozonet
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Heather A Parker
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Margreet C M Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
29
|
Crifo B, Schaible B, Brown E, Halligan DN, Scholz CC, Fitzpatrick SF, Kirwan A, Roche HM, Criscuoli M, Naldini A, Giffney H, Crean D, Blanco A, Cavadas MA, Cummins EP, Fabian Z, Taylor CT. Hydroxylase Inhibition Selectively Induces Cell Death in Monocytes. THE JOURNAL OF IMMUNOLOGY 2019; 202:1521-1530. [DOI: 10.4049/jimmunol.1800912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022]
|
30
|
Shang L, Wang T, Tong D, Kang W, Liang Q, Ge S. Prolyl hydroxylases positively regulated LPS-induced inflammation in human gingival fibroblasts via TLR4/MyD88-mediated AKT/NF-κB and MAPK pathways. Cell Prolif 2018; 51:e12516. [PMID: 30091492 PMCID: PMC6528886 DOI: 10.1111/cpr.12516] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Prolyl hydroxylases (PHDs) play essential roles in oxygen-sensing system, whereas the effects of PHDs on inflammation have not been totally uncovered. Our study aimed to investigate the role of PHDs in lipopolysaccharide (LPS)-induced inflammation of human gingival fibroblasts (HGFs) and clarify the potential mechanisms. MATERIALS AND METHODS A pan hydroxylase inhibitor, dimethyloxallyl glycine (DMOG), and RNA interference were used to explore the role of PHDs in inflammation. Cytotoxic effect of DMOG was determined by cell-counting kit-8 and flow cytometry respectively. The secretion levels of IL-6 and IL-8 were assessed by ELISA. The mRNA levels of inflammatory cytokines, Toll-like receptor (TLR) 4 and MyD88 were evaluated by quantitative real-time PCR. The activation of NF-κB, mitogen-activated protein kinase (MAPK) and PI3K/AKT pathways were detected by western blot and the nuclear translocation of NF-κB p65 was examined by immunofluorescence. Downregulation of PHD1 and PHD2 was performed with siRNA transfection. RESULTS Dimethyloxallyl glycine inhibited LPS-induced inflammatory cytokine, TLR4 and MyD88 expression in gene level and the elevated secretion of IL-6 and IL-8 was also downregulated. Additionally, LPS-induced activation of NF-κB, MAPK and AKT pathways was abolished by DMOG treatment. Importantly, LPS-induced inflammatory cytokine expression was merely suppressed by PHD2 knockdown. CONCLUSIONS Prolyl hydroxylases acted as a positive regulator in LPS-induced inflammation of HGFs via TLR4/MyD88-mediated NF-κB, MAPK and AKT signalling pathways and PHD2 among three isoforms was principally responsible for the effects.
Collapse
Affiliation(s)
- Lingling Shang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Ting Wang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Dongdong Tong
- Department of Oral maxillofacial SurgerySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Wenyan Kang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Qianyu Liang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Shaohua Ge
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| |
Collapse
|
31
|
Prolyl hydroxylase inhibitor DMOG suppressed inflammatory cytokine production in human gingival fibroblasts stimulated with Fusobacterium nucleatum. Clin Oral Investig 2018; 23:3123-3132. [PMID: 30411281 DOI: 10.1007/s00784-018-2733-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Fusobacterium nucleatum (F. nucleatum) is one of the most common bacteria involved in the initiation and progression of periodontal diseases. Pharmacological inhibitor of prolyl hydroxylases (PHDs), dimethyloxallyl glycine (DMOG), has been reported to exert anti-inflammatory effects. The aim of this investigation was to evaluate the role of DMOG in inflammatory cytokine production of human gingival fibroblasts (HGFs) stimulated with F. nucleatum. MATERIAL AND METHODS HGFs were pretreated with 10, 50, and 100 μM DMOG for 24 h before infected with F. nucleatum (MOI = 100). Cell morphology and survival after infection with F. nucleatum were determined by crystal violet staining assay. The mRNA levels of interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and IL-1β were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). The production of IL-6, IL-8, TNF-α, and IL-1β was assessed by enzyme-linked immunosorbent assay (ELISA). RESULTS F. nucleatum did not affect the morphology and survival of HGFs by the concentrations of MOI (multiplicity of infection) = 10, 50, and 100. The mRNA levels of IL-6, IL-8, TNF-α, and IL-1β were significantly enhanced with the stimulation of F. nucleatum, and the maximal effect reached at 6 h. The secretion of IL-6, IL-8, and TNF-α was significantly upregulated by the infection of F. nucleatum while the production of IL-1β was nearly unchanged. Above all, DMOG suppressed F. nucleatum-stimulated IL-6, IL-8, TNF-α, and IL-1β expressions. CONCLUSIONS These data indicate that prolyl hydroxylase inhibitor DMOG partly downregulates inflammatory cytokine expression in F. nucleatum-infected HGFs. CLINICAL RELEVANCE DMOG may provide a novel strategy for the therapy of periodontitis.
Collapse
|
32
|
Effect of Hypoxia on the Pathogenesis of Acinetobacter baumannii and Pseudomonas aeruginosa In Vitro and in Murine Experimental Models of Infection. Infect Immun 2018; 86:IAI.00543-18. [PMID: 30082478 PMCID: PMC6204731 DOI: 10.1128/iai.00543-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022] Open
Abstract
Hypoxia modulates bacterial virulence and the inflammation response through hypoxia-inducible factor 1α (HIF-1α). Here we study the influence of hypoxia on Acinetobacter baumannii and Pseudomonas aeruginosa infections. In vitro, hypoxia increases the bactericidal activities of epithelial cells against A. baumannii and P. aeruginosa, reducing extracellular bacterial concentrations to 50.5% ± 7.5% and 90.8% ± 13.9%, respectively, at 2 h postinfection. The same phenomenon occurs in macrophages (67.6% ± 18.2% for A. baumannii at 2 h and 50.3% ± 10.9% for P. aeruginosa at 24 h). Hypoxia decreases the adherence of A. baumannii to epithelial cells (42.87% ± 8.16% at 2 h) and macrophages (52.0% ± 18.7% at 24 h), as well as that of P. aeruginosa (24.9% ± 4.5% in epithelial cells and 65.7% ± 5.5% in macrophages at 2 h). Moreover, hypoxia decreases the invasion of epithelial cells (48.6% ± 3.8%) and macrophages (8.7% ± 6.9%) by A. baumannii at 24 h postinfection and by P. aeruginosa at 2 h postinfection (75.0% ± 16.3% and 63.4% ± 5.4%, respectively). In vivo, hypoxia diminishes bacterial loads in fluids and tissues in animal models of infection by both pathogens. In contrast, mouse survival time was shorter under hypoxia (23.92 versus 36.42 h) with A. baumannii infection. No differences in the production of cytokines or HIF-1α were found between hypoxia and normoxia in vitro or in vivo We conclude that hypoxia increases the bactericidal activities of host cells against both pathogens and reduces the interaction of pathogens with host cells. Moreover, hypoxia accelerates the rate at which animals die despite the lower bacterial concentrations in vivo.
Collapse
|
33
|
Yamamoto T, Ugawa Y, Kawamura M, Yamashiro K, Kochi S, Ideguchi H, Takashiba S. Modulation of microenvironment for controlling the fate of periodontal ligament cells: the role of Rho/ROCK signaling and cytoskeletal dynamics. J Cell Commun Signal 2018; 12:369-378. [PMID: 29086204 PMCID: PMC5842188 DOI: 10.1007/s12079-017-0425-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022] Open
Abstract
Cells behave in a variety of ways when they perceive changes in their microenvironment; the behavior of cells is guided by their coordinated interactions with growth factors, niche cells, and extracellular matrix (ECM). Modulation of the microenvironment affects the cell morphology and multiple gene expressions. Rho/Rho-associated coiled-coil-containing protein kinase (ROCK) signaling is one of the key regulators of cytoskeletal dynamics and actively and/or passively determines the cell fate, such as proliferation, migration, differentiation, and apoptosis, by reciprocal communication with the microenvironment. During periodontal wound healing, it is important to recruit the residential stem cells into the defect site for regeneration and homeostasis of the periodontal tissue. Periodontal ligament (PDL) cells contain a heterogeneous fibroblast population, including mesenchymal stem cells, and contribute to the reconstruction of tooth-supporting tissues. Therefore, bio-regeneration of PDL cells has been the ultimate goal of periodontal therapy for decades. Recent stem cell researches have shed light on intrinsic ECM properties, providing paradigm shifts in cell fate determination. This review focuses on the role of ROCK activity and the effects of Y-27632, a specific inhibitor of ROCK, in the modulation of ECM-microenvironment. Further, it presents the current understanding of how Rho/ROCK signaling affects the fate determination of stem cells, especially PDL cells. In addition, we have also discussed in detail the underlying mechanisms behind the reciprocal response to the microenvironment.
Collapse
Affiliation(s)
- Tadashi Yamamoto
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Yuki Ugawa
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Mari Kawamura
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Keisuke Yamashiro
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Shinsuke Kochi
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Hidetaka Ideguchi
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Shogo Takashiba
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| |
Collapse
|
34
|
Ebersole JL, Novak MJ, Orraca L, Martinez-Gonzalez J, Kirakodu S, Chen KC, Stromberg A, Gonzalez OA. Hypoxia-inducible transcription factors, HIF1A and HIF2A, increase in aging mucosal tissues. Immunology 2018; 154:452-464. [PMID: 29338076 DOI: 10.1111/imm.12894] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/22/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023] Open
Abstract
Hypoxia (i.e. oxygen deprivation) activates the hypoxia-signalling pathway, primarily via hypoxia-inducible transcription factors (HIF) for numerous target genes, which mediate angiogenesis, metabolism and coagulation, among other processes to try to replenish tissues with blood and oxygen. Hypoxia signalling dysregulation also commonly occurs during chronic inflammation. We sampled gingival tissues from rhesus monkeys (Macaca mulatta; 3-25 years old) and total RNA was isolated for microarray analysis. HIF1A, HIF1B and HIF2A were significantly different in healthy aged tissues, and both HIF1A and HIF3A were positively correlated with aging. Beyond these transcription factor alterations, analysis of patterns of gene expression involved in hypoxic changes in tissues showed specific increases in metabolic pathway hypoxia-inducible genes, whereas angiogenesis pathway gene changes were more variable in healthy aging tissues across the animals. With periodontitis, aging tissues showed decreases in metabolic gene expression related to carbohydrate/lipid utilization (GBE1, PGAP1, TPI1), energy metabolism and cell cycle regulation (IER3, CCNG2, PER1), with up-regulation of transcription genes and cellular proliferation genes (FOS, EGR1, MET, JMJD6) that are hypoxia-inducible. The potential clinical implications of these results are related to the epidemiological findings of increased susceptibility and expression of periodontitis with aging. More specifically the findings describe that hypoxic stress may exist in aging gingival tissues before documentation of clinical changes of periodontitis and, so, may provide an explanatory molecular risk factor for an elevated capacity of the tissues to express destructive processes in response to changes in the microbial biofilms characteristic of a more pathogenic microbial challenge.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Michael John Novak
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Luis Orraca
- School of Dentistry, University of Puerto Rico, Sabana Seca, PR, USA
| | | | - Sreenatha Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Kuey C Chen
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Arnold Stromberg
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Octavio A Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
35
|
Taylor CT, Colgan SP. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat Rev Immunol 2017; 17:774-785. [PMID: 28972206 PMCID: PMC5799081 DOI: 10.1038/nri.2017.103] [Citation(s) in RCA: 424] [Impact Index Per Article: 60.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunological niches are focal sites of immune activity that can have varying microenvironmental features. Hypoxia is a feature of physiological and pathological immunological niches. The impact of hypoxia on immunity and inflammation can vary depending on the microenvironment and immune processes occurring in a given niche. In physiological immunological niches, such as the bone marrow, lymphoid tissue, placenta and intestinal mucosa, physiological hypoxia controls innate and adaptive immunity by modulating immune cell proliferation, development and effector function, largely via transcriptional changes driven by hypoxia-inducible factor (HIF). By contrast, in pathological immunological niches, such as tumours and chronically inflamed, infected or ischaemic tissues, pathological hypoxia can drive tissue dysfunction and disease development through immune cell dysregulation. Here, we differentiate between the effects of physiological and pathological hypoxia on immune cells and the consequences for immunity and inflammation in different immunological niches. Furthermore, we discuss the possibility of targeting hypoxia-sensitive pathways in immune cells for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, 80045 Colorado, USA
| |
Collapse
|
36
|
Dickinson RS, Murphy F, Doherty C, Williams S, Mirchandani A, Willson J, Scotti JS, Preston G, Schofield CJ, Whyte MK, Walmsley SR. Pseudomonas expression of an oxygen sensing prolyl hydroxylase homologue regulates neutrophil host responses in vitro and in vivo. Wellcome Open Res 2017; 2:104. [PMID: 29387803 PMCID: PMC5701443 DOI: 10.12688/wellcomeopenres.12871.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Pseudomonas species are adapted to evade innate immune responses and can persist at sites of relative tissue hypoxia, including the mucus-plugged airways of patients with cystic fibrosis and bronchiectasis. The ability of these bacteria to directly sense and respond to changes in local oxygen availability is in part consequent upon expression of the 2-oxoglutarate oxygenase, Pseudomonas prolyl hydroxylase (PPHD), which acts on elongation factor Tu (EF-Tu), and is homologous with the human hypoxia inducible factor (HIF) prolyl hydroxylases. We report that PPHD expression regulates the neutrophil response to acute pseudomonal infection. Methods:In vitro co-culture experiments were performed with human neutrophils and PPHD-deficient and wild-type bacteria and supernatants, with viable neutrophil counts determined by flow cytometry. In vivo consequences of infection with PPHD deficient P. aeruginosa were determined in an acute pneumonia mouse model following intra-tracheal challenge. Results: Supernatants of PPHD-deficient bacterial cultures contained higher concentrations of the phenazine exotoxin pyocyanin and induced greater acceleration of neutrophil apoptosis than wild-type PAO1 supernatants in vitro. In vivo infection with PPHD mutants compared to wild-type PAO1 controls resulted in increased levels of neutrophil apoptosis and impaired control of infection, with higher numbers of P. aeruginosa recovered from the lungs of mice infected with the PPHD-deficient strain. This resulted in an overall increase in mortality in mice infected with the PPHD-deficient strain. Conclusions: Our data show that Pseudomonas expression of its prolyl hydroxylase influences the outcome of host-pathogen interactions in vitro and in vivo, demonstrating the importance of considering how both host and pathogen adaptations to hypoxia together define outcomes of infection. Given that inhibitors for the HIF prolyl hydroxylases are in late stage trials for the treatment of anaemia and that the active sites of PPHD and human HIF prolyl hydroxylases are closely related, the results are of current clinical interest.
Collapse
Affiliation(s)
- Rebecca S. Dickinson
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Fiona Murphy
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Catherine Doherty
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Sam Williams
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Ananda Mirchandani
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Joseph Willson
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - John S. Scotti
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Gail Preston
- Department of Plant Sciences, University of Oxford, Oxford, OX1 3RB, UK
| | - Christopher J. Schofield
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Moira K.B. Whyte
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Sarah R. Walmsley
- MRC/University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
37
|
Colgan SP, Campbell EL. Oxygen metabolism and innate immune responses in the gut. J Appl Physiol (1985) 2017; 123:1321-1327. [PMID: 28705991 DOI: 10.1152/japplphysiol.00113.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 01/02/2023] Open
Abstract
Epithelial cells of the mucosa provide a first line of defense to prevent the inappropriate translocation of luminal antigens, and therefore contribute significantly to nonspecific innate immunity. In the gastrointestinal (GI) tract, barrier is provided by multiple components of the mucosa, including mucus production, epithelial junctional complexes, and the production of antimicrobial molecules. In recent years, it is better appreciated that tissue oxygen metabolism is key to homeostasis in the mucosa. The intestine, for example, maintains a low baseline Po2 level due to high rates of metabolism, countercurrent blood flow, and the presence of a steep oxygen gradient across the luminal aspect of tissue surface. As a result, hypoxia and hypoxia-inducible factor (HIF)-dependent signaling exists even in the healthy, unperturbed intestinal mucosa. In a number of examples, HIF has been demonstrated both to promote barrier function during homeostasis and to promote resolution of active inflammation. Hypoxia-elicited factors that contribute to innate responses in the mucosa include the transcriptional regulation of mucin genes, junction proteins, and autophagic flux. Here, we review current literature related to hypoxia and innate immunity in health and during mucosal inflammation.
Collapse
Affiliation(s)
- Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado; and
| | - Eric L Campbell
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
38
|
Marteyn BS, Burgel PR, Meijer L, Witko-Sarsat V. Harnessing Neutrophil Survival Mechanisms during Chronic Infection by Pseudomonas aeruginosa: Novel Therapeutic Targets to Dampen Inflammation in Cystic Fibrosis. Front Cell Infect Microbiol 2017; 7:243. [PMID: 28713772 PMCID: PMC5492487 DOI: 10.3389/fcimb.2017.00243] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
More than two decades after cloning the cystic fibrosis transmembrane regulator (CFTR) gene, the defective gene in cystic fibrosis (CF), we still do not understand how dysfunction of this ion channel causes lung disease and the tremendous neutrophil burden which persists within the airways; nor why chronic colonization by Pseudomonas aeruginosa develops in CF patients who are thought to be immunocompetent. It appears that the microenvironment within the lung of CF patients provides favorable conditions for both P. aeruginosa colonization and neutrophil survival. In this context, the ability of bacteria to induce hypoxia, which in turn affects neutrophil survival is an additional level of complexity that needs to be accounted for when controlling neutrophil fate in CF. Recent studies have underscored the importance of neutrophils in innate immunity and their functions appear to extend far beyond their well-described role in antibacterial defense. Perhaps a disturbance in neutrophil reprogramming during the course of an infection severely modulates the inflammatory response in CF. Furthermore there is an emerging concept that the CFTR itself may be an immune modulator and stimulating CFTR function in CF patients could promote neutrophil and macrophages antimicrobial function. Fostering the resolution of inflammation by favoring neutrophil apoptosis could preserve their microbicidal activities but decrease their proinflammatory potential. In this context, triggering neutrophil apoptosis with roscovitine may be a potential therapeutic option and this is currently being evaluated in CF patients. In the present review we discuss how neutrophils functions are disturbed in CF and how this may relate to chronic infection with P. aeuginosa and we propose novel research directions aimed at modulating neutrophil survival, dampening lung inflammation and ultimately leading to an amelioration of the lung disease.
Collapse
Affiliation(s)
- Benoît S Marteyn
- Unité de Pathogénie Microbienne Moléculaire, Institut PasteurParis, France.,Institut National de la Santé et de la Recherche Médicale, U12021202Paris, France.,Institut Gustave RoussyVillejuif, France
| | - Pierre-Régis Burgel
- Université Paris Descartes, Sorbonne Paris CitéParis, France.,Pneumology Department, Hôpital CochinParis, France
| | | | - Véronique Witko-Sarsat
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut CochinParis, France.,Centre National de la Recherche Scientifique-UMR 8104Paris, France.,Center of Excellence, Labex InflamexParis, France
| |
Collapse
|
39
|
Schaible B, Rodriguez J, Garcia A, von Kriegsheim A, McClean S, Hickey C, Keogh CE, Brown E, Schaffer K, Broquet A, Taylor CT. Hypoxia Reduces the Pathogenicity of Pseudomonas aeruginosa by Decreasing the Expression of Multiple Virulence Factors. J Infect Dis 2017; 215:1459-1467. [PMID: 28368464 DOI: 10.1093/infdis/jix139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/17/2017] [Indexed: 12/16/2023] Open
Abstract
Our understanding of how the course of opportunistic bacterial infection is influenced by the microenvironment is limited. We demonstrate that the pathogenicity of Pseudomonas aeruginosa strains derived from acute clinical infections is higher than that of strains derived from chronic infections, where tissues are hypoxic. Exposure to hypoxia attenuated the pathogenicity of strains from acute (but not chronic) infections, implicating a role for hypoxia in regulating bacterial virulence. Mass spectrometric analysis of the secretome of P. aeruginosa derived from an acute infection revealed hypoxia-induced repression of multiple virulence factors independent of altered bacterial growth. Pseudomonas aeruginosa lacking the Pseudomonas prolyl-hydroxylase domain-containing protein, which has been implicated in bacterial oxygen sensing, displays reduced virulence factor expression. Furthermore, pharmacological hydroxylase inhibition reduces virulence factor expression and pathogenicity in a murine model of pneumonia. We hypothesize that hypoxia reduces P. aeruginosa virulence at least in part through the regulation of bacterial hydroxylases.
Collapse
Affiliation(s)
| | | | - Amaya Garcia
- Systems Biology Ireland, University College Dublin
| | | | - Siobhán McClean
- Centre for Microbial Host Interactions, Department of Science, Institute of Technology Tallaght-Dublin, and
| | | | | | | | - Kirsten Schaffer
- Department of Clinical Microbiology, St Vincent's University Hospital, Dublin, Ireland; and
| | | | - Cormac T Taylor
- Conway Institute and
- Systems Biology Ireland, University College Dublin
| |
Collapse
|
40
|
Polke M, Seiler F, Lepper PM, Kamyschnikow A, Langer F, Monz D, Herr C, Bals R, Beisswenger C. Hypoxia and the hypoxia-regulated transcription factor HIF-1α suppress the host defence of airway epithelial cells. Innate Immun 2017; 23:373-380. [PMID: 28409544 DOI: 10.1177/1753425917698032] [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] [Indexed: 01/24/2023] Open
Abstract
Chronic diseases of the respiratory tract, such as cystic fibrosis, are associated with mucosal and systemic hypoxia. Innate immune functions of airway epithelial cells are required to prevent and control infections of the lung parenchyma. The transcription factor hypoxia-inducible factor 1α (HIF-1α) regulates cellular adaptation to low oxygen conditions. Here, we show that hypoxia and HIF-1α regulate innate immune mechanisms of cultured human bronchial epithelial cells (HBECs). Exposure of primary HBECs to hypoxia or the prolyl hydroxylase inhibitor dimethyloxaloylglycine (DMOG) resulted in a significantly decreased expression of inflammatory mediators (IL-6, IFN-γ-induced protein 10) in response to ligands for TLRs (flagellin, polyI:C) and Pseudomonas aeruginosa, whereas the expression of inflammatory mediators was not affected by hypoxia or DMOG in the absence of microbial factors. Small interfering RNA-mediated knockdown of HIF-1α in HBECs and in the bronchial epithelial cell line Calu-3 resulted in increased expression of inflammatory mediators. The inflammatory response was decreased in lungs of mice stimulated with inactivated P. aeruginosa under hypoxia. These data suggest that hypoxia suppresses the innate immune response of airway epithelial cells via HIF-1α.
Collapse
Affiliation(s)
- Markus Polke
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Frederik Seiler
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Philipp M Lepper
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Andreas Kamyschnikow
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Frank Langer
- 2 Department of Thoracic and Cardiovascular Surgery, Saarland University, Homburg, Germany
| | - Dominik Monz
- 3 Department of Pediatrics and Neonatology, Saarland University, Homburg, Germany
| | - Christian Herr
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Robert Bals
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christoph Beisswenger
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
41
|
Zeitouni NE, Chotikatum S, von Köckritz-Blickwede M, Naim HY. The impact of hypoxia on intestinal epithelial cell functions: consequences for invasion by bacterial pathogens. Mol Cell Pediatr 2016; 3:14. [PMID: 27002817 PMCID: PMC4803720 DOI: 10.1186/s40348-016-0041-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/13/2016] [Indexed: 02/06/2023] Open
Abstract
The maintenance of oxygen homeostasis in human tissues is mediated by several cellular adaptations in response to low-oxygen stress, called hypoxia. A decrease in tissue oxygen levels is initially counteracted by increasing local blood flow to overcome diminished oxygenation and avoid hypoxic stress. However, studies have shown that the physiological oxygen concentrations in several tissues are much lower than atmospheric (normoxic) conditions, and the oxygen supply is finely regulated in individual cell types. The gastrointestinal tract has been described to subsist in a state of physiologically low oxygen level and is thus depicted as a tissue in the state of constant low-grade inflammation. The intestinal epithelial cell layer plays a vital role in the immune response to inflammation and infections that occur within the intestinal tissue and is involved in many of the adaptation responses to hypoxic stress. This is especially relevant in the context of inflammatory disorders, such as inflammatory bowel disease (IBD). Therefore, this review aims to describe the intestinal epithelial cellular response to hypoxia and the consequences for host interactions with invading gastrointestinal bacterial pathogens.
Collapse
Affiliation(s)
- Nathalie E. Zeitouni
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sucheera Chotikatum
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hassan Y. Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
42
|
Devraj G, Beerlage C, Brüne B, Kempf VAJ. Hypoxia and HIF-1 activation in bacterial infections. Microbes Infect 2016; 19:144-156. [PMID: 27903434 DOI: 10.1016/j.micinf.2016.11.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022]
Abstract
For most of the living beings, oxygen is one of the essential elements required to sustain life. Deprivation of oxygen causes tissue hypoxia and this severely affects host cell and organ functions. Tissue hypoxia is a prominent microenvironmental condition occurring in infections and there is a body of evidence that hypoxia and inflammation are interconnected with each other. The primary key factor mediating the mammalian hypoxic response is hypoxia inducible factor (HIF)-1, which regulates oxygen homeostasis on cellular, tissue and organism level. Recent studies show that HIF-1 plays a central role in angiogenesis, cancer and cardiovascular disease but also in bacterial infections. Activation of HIF-1 depends on the nature of the pathogen and the characteristics of infections in certain hosts. Up to date, it is not completely clear whether the phenomenon of HIF-1 activation in infections has a protective or detrimental effect on the host. In this review, we give an overview of whether and how hypoxia and HIF-1 affect the course of infections.
Collapse
Affiliation(s)
- Gayatri Devraj
- Institute of Medical Microbiology and Infection Control, Goethe-University, Paul-Ehrlich-Str. 40, D-60596 Frankfurt am Main, Germany
| | - Christiane Beerlage
- Institute of Medical Microbiology and Infection Control, Goethe-University, Paul-Ehrlich-Str. 40, D-60596 Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I - Pathobiochemistry, Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Volkhard A J Kempf
- Institute of Medical Microbiology and Infection Control, Goethe-University, Paul-Ehrlich-Str. 40, D-60596 Frankfurt am Main, Germany.
| |
Collapse
|
43
|
Negative Impact of Hypoxia on Tryptophan 2,3-Dioxygenase Function. Mediators Inflamm 2016; 2016:1638916. [PMID: 27563172 PMCID: PMC4985583 DOI: 10.1155/2016/1638916] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/14/2016] [Accepted: 06/26/2016] [Indexed: 02/01/2023] Open
Abstract
Tryptophan is an essential amino acid for hosts and pathogens. The liver enzyme tryptophan 2,3-dioxygenase (TDO) provokes, by its ability to degrade tryptophan to N-formylkynurenine, the precursor of the immune-relevant kynurenines, direct and indirect antimicrobial and immunoregulatory states. Up to now these TDO-mediated broad-spectrum effector functions have never been observed under hypoxia in vitro, although physiologic oxygen concentrations in liver tissue are low, especially in case of infection. Here we analysed recombinant expressed human TDO and ex vivo murine TDO functions under different oxygen conditions and show that TDO-induced restrictions of clinically relevant pathogens (bacteria, parasites) and of T cell proliferation are abrogated under hypoxic conditions. We pinpointed the loss of TDO efficiency to the reduction of TDO activity, since cell survival and TDO protein levels were unaffected. In conclusion, the potent antimicrobial as well as immunoregulatory effects of TDO were substantially impaired under hypoxic conditions that pathophysiologically occur in vivo. This might be detrimental for the appropriate host immune response towards relevant pathogens.
Collapse
|
44
|
Taylor CT, Doherty G, Fallon PG, Cummins EP. Hypoxia-dependent regulation of inflammatory pathways in immune cells. J Clin Invest 2016; 126:3716-3724. [PMID: 27454299 DOI: 10.1172/jci84433] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Uncontrolled inflammation underpins a diverse range of diseases where effective therapy remains an unmet clinical need. Hypoxia is a prominent feature of the inflammatory microenvironment that regulates key transcription factors including HIF and NF-κB in both innate and adaptive immune cells. In turn, altered activity of the pathways controlled by these factors can affect the course of inflammation through the regulation of immune cell development and function. In this review, we will discuss these pathways and the oxygen sensors that confer hypoxic sensitivity in immune cells. Furthermore, we will describe how hypoxia-dependent pathways contribute to immunity and discuss their potential as therapeutic targets in inflammatory and infectious disease.
Collapse
|
45
|
Guillard T, Pons S, Roux D, Pier GB, Skurnik D. Antibiotic resistance and virulence: Understanding the link and its consequences for prophylaxis and therapy. Bioessays 2016; 38:682-93. [PMID: 27248008 DOI: 10.1002/bies.201500180] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
"Antibiotic resistance is usually associated with a fitness cost" is frequently accepted as common knowledge in the field of infectious diseases. However, with the advances in high-throughput DNA sequencing that allows for a comprehensive analysis of bacterial pathogenesis at the genome scale, including antibiotic resistance genes, it appears that this paradigm might not be as solid as previously thought. Recent studies indicate that antibiotic resistance is able to enhance bacterial fitness in vivo with a concomitant increase in virulence during infections. As a consequence, strategies to minimize antibiotic resistance turn out to be not as simple as initially believed. Indeed, decreased antibiotic use may not be sufficient to let susceptible strains outcompete the resistant ones. Here, we put in perspective these findings and review alternative approaches, such as preventive and therapeutic anti-bacterial immunotherapies that have the potential to by-pass the classic antibiotics.
Collapse
Affiliation(s)
- Thomas Guillard
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Laboratoire de Bactériologie-Virologie-Hygiène hospitalière, Hôpital Robert Debré - CHU de Reims, UFR de Médecine, Université de Reims Champagne-Ardenne, Reims, France
| | - Stéphanie Pons
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Damien Roux
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,INSERM, IAME, UMR 1137, Paris, France.,Univ Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France.,AP-HP, Hôpital Louis Mourier, Service de Réanimation Médico-Chirurgicale, Colombes, France
| | - Gerald B Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Skurnik
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Selfridge AC, Cavadas MAS, Scholz CC, Campbell EL, Welch LC, Lecuona E, Colgan SP, Barrett KE, Sporn PHS, Sznajder JI, Cummins EP, Taylor CT. Hypercapnia Suppresses the HIF-dependent Adaptive Response to Hypoxia. J Biol Chem 2016; 291:11800-8. [PMID: 27044749 DOI: 10.1074/jbc.m116.713941] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 01/18/2023] Open
Abstract
Molecular oxygen and carbon dioxide are the primary gaseous substrate and product of oxidative metabolism, respectively. Hypoxia (low oxygen) and hypercapnia (high carbon dioxide) are co-incidental features of the tissue microenvironment in a range of pathophysiologic states, including acute and chronic respiratory diseases. The hypoxia-inducible factor (HIF) is the master regulator of the transcriptional response to hypoxia; however, little is known about the impact of hypercapnia on gene transcription. Because of the relationship between hypoxia and hypercapnia, we investigated the effect of hypercapnia on the HIF pathway. Hypercapnia suppressed HIF-α protein stability and HIF target gene expression both in mice and cultured cells in a manner that was at least in part independent of the canonical O2-dependent HIF degradation pathway. The suppressive effects of hypercapnia on HIF-α protein stability could be mimicked by reducing intracellular pH at a constant level of partial pressure of CO2 Bafilomycin A1, a specific inhibitor of vacuolar-type H(+)-ATPase that blocks lysosomal degradation, prevented the hypercapnic suppression of HIF-α protein. Based on these results, we hypothesize that hypercapnia counter-regulates activation of the HIF pathway by reducing intracellular pH and promoting lysosomal degradation of HIF-α subunits. Therefore, hypercapnia may play a key role in the pathophysiology of diseases where HIF is implicated.
Collapse
Affiliation(s)
| | - Miguel A S Cavadas
- Conway Institute, and Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland
| | - Carsten C Scholz
- From the School of Medicine and Medical Science, Conway Institute, and Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland, the Institute of Physiology, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Eric L Campbell
- the University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Lynn C Welch
- the Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Emilia Lecuona
- the Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Sean P Colgan
- the University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Kim E Barrett
- From the School of Medicine and Medical Science, Conway Institute, and
| | - Peter H S Sporn
- the Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and the Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Jacob I Sznajder
- the Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Eoin P Cummins
- From the School of Medicine and Medical Science, Conway Institute, and
| | - Cormac T Taylor
- From the School of Medicine and Medical Science, Conway Institute, and Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland,
| |
Collapse
|
47
|
Zeitouni NE, Dersch P, Naim HY, von Köckritz-Blickwede M. Hypoxia Decreases Invasin-Mediated Yersinia enterocolitica Internalization into Caco-2 Cells. PLoS One 2016; 11:e0146103. [PMID: 26731748 PMCID: PMC4701670 DOI: 10.1371/journal.pone.0146103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/14/2015] [Indexed: 12/20/2022] Open
Abstract
Yersinia enterocolitica is a major cause of human yersiniosis, with enterocolitis being a typical manifestation. These bacteria can cross the intestinal mucosa, and invade eukaryotic cells by binding to host β1 integrins, a process mediated by the bacterial effector protein invasin. This study examines the role of hypoxia on the internalization of Y. enterocolitica into intestinal epithelial cells, since the gastrointestinal tract has been shown to be physiologically deficient in oxygen levels (hypoxic), especially in cases of infection and inflammation. We show that hypoxic pre-incubation of Caco-2 cells resulted in significantly decreased bacterial internalization compared to cells grown under normoxia. This phenotype was absent after functionally blocking host β1 integrins as well as upon infection with an invasin-deficient Y. enterocolitica strain. Furthermore, downstream phosphorylation of the focal adhesion kinase was also reduced under hypoxia after infection. In good correlation to these data, cells grown under hypoxia showed decreased protein levels of β1 integrins at the apical cell surface whereas the total protein level of the hypoxia inducible factor (HIF-1) alpha was elevated. Furthermore, treatment of cells with the HIF-1 α stabilizer dimethyloxalylglycine (DMOG) also reduced invasion and decreased β1 integrin protein levels compared to control cells, indicating a potential role for HIF-1α in this process. These results suggest that hypoxia decreases invasin-integrin-mediated internalization of Y. enterocolitica into intestinal epithelial cells by reducing cell surface localization of host β1 integrins.
Collapse
Affiliation(s)
- Nathalie E. Zeitouni
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Petra Dersch
- Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Hassan Y. Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
- * E-mail:
| |
Collapse
|
48
|
Pseudomonas aeruginosa Transmigrates at Epithelial Cell-Cell Junctions, Exploiting Sites of Cell Division and Senescent Cell Extrusion. PLoS Pathog 2016; 12:e1005377. [PMID: 26727615 PMCID: PMC4699652 DOI: 10.1371/journal.ppat.1005377] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/09/2015] [Indexed: 12/19/2022] Open
Abstract
To achieve systemic infection, bacterial pathogens must overcome the critical and challenging step of transmigration across epithelial barriers. This is particularly true for opportunistic pathogens such as Pseudomonas aeruginosa, an agent which causes nosocomial infections. Despite extensive study, details on the mechanisms used by this bacterium to transmigrate across epithelial tissues, as well as the entry sites it uses, remain speculative. Here, using real-time microscopy and a model epithelial barrier, we show that P. aeruginosa employs a paracellular transmigration route, taking advantage of altered cell-cell junctions at sites of cell division or when senescent cells are expelled from the cell layer. Once a bacterium transmigrates, it is followed by a cohort of bacteria using the same entry point. The basal compartment is then invaded radially from the initial penetration site. Effective transmigration and propagation require type 4 pili, the type 3 secretion system (T3SS) and a flagellum, although flagellum-deficient bacteria can occasionally invade the basal compartment from wounded areas. In the basal compartment, the bacteria inject the T3SS toxins into host cells, disrupting the cytoskeleton and focal contacts to allow their progression under the cells. Thus, P. aeruginosa exploits intrinsic host cell processes to breach the epithelium and invade the subcellular compartment. In normal situations, the mucosae constitute efficient barriers against the invasion of opportunistic pathogens. The bacteria inducing nosocomial infections take advantage of pre-existing pathological situations to cross the epithelium and spread in deeper tissues. The conditions on the host side permitting transmigration and the combination of virulence factors used by the bacteria to transmigrate are mostly speculative. Here, we studied the transmigration process of Pseudomonas aeruginosa, a bacterium causing hospital-acquired acute and chronic infections. We found that bacteria exploits weakened cell-cell junctions of the epithelium, such as those generated at sites of cell division or when dying cells are extruded from the cell layer, to breach the cell layer, using specific virulence factors and motility appendages.
Collapse
|
49
|
Expression of hypoxia inducible factor-1α and vascular endothelial growth factor-C in human chronic periodontitis. J Dent Sci 2015. [DOI: 10.1016/j.jds.2014.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
50
|
Schaffer K, Taylor CT. The impact of hypoxia on bacterial infection. FEBS J 2015; 282:2260-6. [PMID: 25786849 DOI: 10.1111/febs.13270] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/19/2015] [Accepted: 03/16/2015] [Indexed: 12/28/2022]
Abstract
Tissue hypoxia is a common microenvironmental feature during inflammation associated with bacterial infection. Hypoxia has recently been shown to play an important role in both innate and adaptive host immunity through the regulation of transcription factors, including hypoxia-inducible factor and nuclear factor-κB, in both infiltrating immunocytes and inflamed resident cells. Recent studies have suggested that, by regulating these important immune effector pathways in host tissues, hypoxia can significantly alter the process of bacterial infection and subsequent disease progression. Although hypoxia is often beneficial in terms of reducing the development of infection, its net effect depends on a number of factors, including the nature of the pathogen and the characteristics of the infection encountered. In this minireview, we will discuss the impact of local tissue hypoxia and the resulting activation of hypoxia-sensitive pathways on bacterial infection by a range of pathogens. Furthermore, we will review how this knowledge may be used to develop new approaches to anti-infective therapeutics.
Collapse
Affiliation(s)
- Kirsten Schaffer
- Department of Microbiology, St. Vincent's University Hospital, Dublin 4, Ireland
| | - Cormac T Taylor
- School of Medicine and Medical Science & The Conway Institute, University College Dublin, Dublin 4, Ireland
| |
Collapse
|