1
|
Fredman G, Serhan CN. Specialized pro-resolving mediators in vascular inflammation and atherosclerotic cardiovascular disease. Nat Rev Cardiol 2024; 21:808-823. [PMID: 38216693 DOI: 10.1038/s41569-023-00984-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/14/2024]
Abstract
Timely resolution of the acute inflammatory response (or inflammation resolution) is an active, highly coordinated process that is essential to optimal health. Inflammation resolution is regulated by specific endogenous signalling molecules that function as 'stop signals' to terminate the inflammatory response when it is no longer needed; to actively promote healing, regeneration and tissue repair; and to limit pain. Specialized pro-resolving mediators are a superfamily of signalling molecules that initiate anti-inflammatory and pro-resolving actions. Without an effective and timely resolution response, inflammation can become chronic, a pathological state that is associated with many widely occurring human diseases, including atherosclerotic cardiovascular disease. Uncovering the mechanisms of inflammation resolution failure in cardiovascular diseases and identifying useful biomarkers for non-resolving inflammation are unmet needs. In this Review, we discuss the accumulating evidence that supports the role of non-resolving inflammation in atherosclerosis and the use of specialized pro-resolving mediators as therapeutic tools for the treatment of atherosclerotic cardiovascular disease. We highlight open questions about therapeutic strategies and mechanisms of disease to provide a framework for future studies on the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anaesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Serhan CN, Chiang N, Nshimiyimana R. Low-dose pro-resolving mediators temporally reset the resolution response to microbial inflammation. Mol Med 2024; 30:153. [PMID: 39294573 PMCID: PMC11411770 DOI: 10.1186/s10020-024-00877-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/15/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Specialized pro-resolving mediators (SPMs) promote resolution of inflammation, clear infections and stimulate tissue regeneration. These include resolvins, protectins, and maresins. During self-resolving acute inflammation, SPMs are produced and have key functions activating endogenous resolution response for returning to homeostasis. Herein, we addressed whether infections initiated with ongoing inflammation alter resolution programs, and if low-dose repetitive SPM regimen re-programs the resolution response. METHODS Inflammation was initiated with zymosan (1 mg/mouse) followed by E. coli (105 CFU/mouse) infections carried out in murine peritonitis, and exudates collected at 4-72 h. Leukocytes were enumerated using light microscopy, percentages of PMN, monocytes and macrophages were determined using flow cytometry, and resolution indices calculated. Lipid mediators and SPM profiles were established using mass spectrometry-based metabololipidomics. Repetitive dosing with a SPM panel consisting of RvD1, RvD2, RvD5, MaR1 and RvE2 (0.1 ng/mouse each, i.p.) was given to mice, followed by zymosan challenge. Leukocyte composition, resolution indices and RNA-sequencing were carried out for the repetitive SPM treatments. RESULTS E. coli infections initiated acute inflammation-resolution programs with temporal SPM production in the infectious exudates. Zymosan-induced inflammation prior to E. coli peritonitis shifted exudate resolution indices and delayed E. coli clearance. Lipid mediator metabololipidomics demonstrated that E. coli infection with ongoing zymosan-induced inflammation shifted the time course of exudate SPMs, activating a SPM cluster that included RvD1, RvD5 and MaR1 during the initiation phase of infectious inflammation (0-4 h); RvD5 and MaR1 were present also in the resolution phase (24-48 h). To emulate daily SPM regimens used in humans, a repetitive subthreshold dosing of the SPM panel RvD1, RvD2, RvD5, MaR1 and RvE2 each at 0.1 ng per mouse was administered. This low-dose SPM regimen accelerated exudate PMN clearance following zymosan-induced inflammation, and shortened the resolution interval by > 70%. These low-dose SPMs regulated genes and pathways related to immune response, chemokine clearance and tissue repair, as demonstrated by using RNA-sequencing. CONCLUSIONS Infections encountered during ongoing inflammation in mice reset the resolution mechanisms of inflammation via SPM clusters. Low-dose SPMs activate innate immune responses and pathways towards the resolution response that can be reprogrammed.
Collapse
Affiliation(s)
- Charles N Serhan
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Mass General Brigham and Harvard Medical School, 60 Fenwood Rd., Hale Building for Transformative Medicine 3-016, Boston, MA, 02115, USA.
| | - Nan Chiang
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Mass General Brigham and Harvard Medical School, 60 Fenwood Rd., Hale Building for Transformative Medicine 3-016, Boston, MA, 02115, USA
| | - Robert Nshimiyimana
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Mass General Brigham and Harvard Medical School, 60 Fenwood Rd., Hale Building for Transformative Medicine 3-016, Boston, MA, 02115, USA
| |
Collapse
|
3
|
Wang YH, Lin CW, Huang CW. Polyunsaturated Fatty Acids as Potential Treatments for COVID-19-Induced Anosmia. Biomedicines 2024; 12:2085. [PMID: 39335598 PMCID: PMC11428228 DOI: 10.3390/biomedicines12092085] [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: 07/28/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Some individuals with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) experience anosmia, or loss of smell. Although the prevalence of anosmia has decreased with the emergence of the Omicron variant, it remains a significant concern. This review examines the potential role of polyunsaturated fatty acids (PUFAs), particularly omega-3 PUFAs, in treating COVID-19-induced anosmia by focusing on the underlying mechanisms of the condition. Omega-3 PUFAs are known for their anti-inflammatory, neuroprotective, and neurotransmission-enhancing properties, which could potentially aid in olfactory recovery. However, study findings are inconsistent. For instance, a placebo-controlled randomized clinical trial found no significant effect of omega-3 PUFA supplementation on olfactory recovery in patients with COVID-19-induced anosmia. These mixed results highlight the limitations of existing research, including small sample sizes, lack of placebo controls, short follow-up periods, and combined treatments. Therefore, more rigorous, large-scale studies are urgently needed to definitively assess the therapeutic potential of omega-3 PUFAs for olfactory dysfunction. Further research is also crucial to explore the broader role of PUFAs in managing viral infections and promoting sensory recovery.
Collapse
Affiliation(s)
- Yu-Han Wang
- Department of Education, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chung-Wei Lin
- Department of Education, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chiung-Wei Huang
- Department of Physiology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
4
|
Rago F, Melo EM, Miller LM, Duray AM, Batista Felix F, Vago JP, de Faria Gonçalves AP, Angelo ALPM, Cassali GD, de Gaetano M, Brennan E, Owen B, Guiry P, Godson C, Alcorn JF, Teixeira MM. Treatment with lipoxin A 4 improves influenza A infection outcome, induces macrophage reprogramming, anti-inflammatory and pro-resolutive responses. Inflamm Res 2024:10.1007/s00011-024-01939-9. [PMID: 39214890 DOI: 10.1007/s00011-024-01939-9] [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: 05/28/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Influenza A is a virus from the Orthomixoviridae family responsible for high lethality rates and morbidity, despite clinically proven vaccination strategies and some anti-viral therapies. The eicosanoid Lipoxin A4 (LXA4) promotes the resolution of inflammation by decreasing cell recruitment and pro-inflammatory cytokines release, but also for inducing activation of apoptosis, efferocytosis, and macrophage reprogramming. OBJECTIVE Here, we evaluated whether a synthetic lipoxin mimetic, designated AT-01-KG, would improve the course of influenza A infection in a murine model. METHOD Mice were infected with influenza A/H1N1 and treated with AT-01-KG (1.7 μg/kg/day, i.p.) at day 3 post-infection. RESULTS AT-01-KG attenuated mortality, reducing leukocyte infiltration and lung damage at day 5 and day 7 post-infection. AT-01-KG is a Formyl Peptide Receptor 2 (designated FPR2/3 in mice) agonist, and the protective responses were not observed in fpr2/3 -/- animals. In mice treated with LXA4 (50 μg/kg/day, i.p., days 3-6 post-infection), at day 7, macrophage reprogramming was observed, as seen by a decrease in classically activated macrophages and an increase in alternatively activated macrophages in the lungs. Furthermore, the number of apoptotic cells and cells undergoing efferocytosis was increased in the lavage of treated mice. Treatment also modulated the adaptive immune response, increasing the number of T helper 2 cells (Th2) and regulatory T (Tregs) cells in the lungs of the treated mice. CONCLUSION Therefore, treatment with a lipoxin A4 analog was beneficial in a model of influenza A infection in mice. The drug decreased inflammation and promoted resolution and beneficial immune responses, suggesting it may be useful in patients with severe influenza.
Collapse
Affiliation(s)
- Flavia Rago
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil.
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
| | - Eliza Mathias Melo
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil
| | - Leigh M Miller
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Alexis M Duray
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Franciel Batista Felix
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana Priscila Vago
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Paula de Faria Gonçalves
- Immunology of Viral Diseases, René Rachou Research Center, Oswaldo Cruz Foundation (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | | | - Geovanni D Cassali
- Comparative Pathology Laboratory, Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Monica de Gaetano
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Benjamin Owen
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Patrick Guiry
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - John F Alcorn
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil.
| |
Collapse
|
5
|
Franks SJ, Gowler PRW, Dunster JL, Turnbull J, Gohir SA, Kelly A, Valdes AM, King JR, Barrett DA, Chapman V, Preston S. Modelling the role of enzymatic pathways in the metabolism of docosahexaenoic acid by monocytes and its association with osteoarthritic pain. Math Biosci 2024; 374:109228. [PMID: 38851528 DOI: 10.1016/j.mbs.2024.109228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Chronic pain is a major cause of disability and suffering in osteoarthritis (OA) patients. Endogenous specialised pro-resolving molecules (SPMs) curtail pro-inflammatory responses. One of the SPM intermediate oxylipins, 17-hydroxydocasahexaenoic acid (17-HDHA, a metabolite of docosahexaenoic acid (DHA)), is significantly associated with OA pain. The aim of this multidisciplinary work is to develop a mathematical model to describe the contributions of enzymatic pathways (and the genes that encode them) to the metabolism of DHA by monocytes and to the levels of the down-stream metabolites, 17-HDHA and 14-hydroxydocasahexaenoic acid (14-HDHA), motivated by novel clinical data from a study involving 30 participants with OA. The data include measurements of oxylipin levels, mRNA levels, measures of OA severity and self-reported pain scores. We propose a system of ordinary differential equations to characterise associations between the different datasets, in order to determine the homeostatic concentrations of DHA, 17-HDHA and 14-HDHA, dependent upon the gene expression of the associated metabolic enzymes. Using parameter-fitting methods, local sensitivity and uncertainty analysis, the model is shown to fit well qualitatively to experimental data. The model suggests that up-regulation of some ALOX genes may lead to the down-regulation of 17-HDHA and that dosing with 17-HDHA increases the production of resolvins, which helps to down-regulate the inflammatory response. More generally, we explore the challenges and limitations of modelling real data, in particular individual variability, and also discuss the value of gathering additional experimental data motivated by the modelling insights.
Collapse
Affiliation(s)
- S J Franks
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - P R W Gowler
- Pain Centre Versus Arthritis, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - J L Dunster
- Institute for Cardiovascular and Metabolic Research, University of Reading, UK
| | - J Turnbull
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK; NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - S A Gohir
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - A Kelly
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - A M Valdes
- Pain Centre Versus Arthritis, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK; NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - J R King
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - D A Barrett
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - V Chapman
- Pain Centre Versus Arthritis, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK; NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - S Preston
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
6
|
Biji CA, Balde A, Nazeer RA. Anti-inflammatory peptide therapeutics and the role of sulphur containing amino acids (cysteine and methionine) in inflammation suppression: A review. Inflamm Res 2024; 73:1203-1221. [PMID: 38769154 DOI: 10.1007/s00011-024-01893-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Inflammation serves as our body's immune response to combat infections, pathogens, viruses, and external stimuli. Inflammation can be classified into two types: acute inflammation and chronic inflammation. Non-steroidal anti-inflammatory medications (NSAIDs) are used to treat both acute and chronic inflammatory disorders. However, these treatments have various side effects such as reduced healing efficiency, peptic ulcers, gastrointestinal toxicities, etc. METHOD: This review assesses the potential of anti-inflammatory peptides (AIPs) derived from various natural sources, such as algae, fungi, plants, animals, and marine organisms. Focusing on peptides rich in cysteines and methionine, sulphur-containing amino acids known for their role in suppression of inflammation. RESULT Due to their varied biological activity, ability to penetrate cells, and low cytotoxicity, bioactive peptides have garnered interest as possible therapeutic agents. The utilisation of AIPs has shown great potential in the treatment of disorders associated with inflammation. AIPs can be obtained from diverse natural sources such as algae, fungi, plants, and animals. Cysteine and methionine are sulphur-containing amino acids that aid in the elimination of free radicals, hence assisting in the treatment of inflammatory diseases. CONCLUSION This review specifically examines several sources of AIPs including peptides that contain numerous cysteines and methionine. In addition, the biological characteristics of these amino acids and advancements in peptide delivery are also discussed.
Collapse
Affiliation(s)
- Catherin Ann Biji
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India.
| |
Collapse
|
7
|
Rago F, Melo EM, Miller LM, Duray AM, Felix FB, Vago JP, Gonçalves APF, Angelo ALPM, Cassali GD, Gaetano M, Brennan E, Owen B, Guiry P, Godson C, Alcorn JF, Teixeira MM. Treatment with lipoxin A 4 improves influenza A infection outcome through macrophage reprogramming, anti-inflammatory and pro-resolutive responses. RESEARCH SQUARE 2024:rs.3.rs-4491036. [PMID: 38947034 PMCID: PMC11213203 DOI: 10.21203/rs.3.rs-4491036/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Objective and design Here, we evaluated whether a synthetic lipoxin mimetic, designated AT-01-KG, would improve the course of influenza A infection in a murine model. Treatment Mice were infected with influenza A/H1N1 and treated with AT-01-KG (1.7 mg/kg/day, i.p.) at day 3 post-infection. Methods Mortality rate was assessed up to day 21 and inflammatory parameters were assessed at days 5 and 7. Results AT-01-KG attenuated mortality, reducing leukocyte infiltration and lung damage at day 5 and day 7 post-infection. AT-01-KG is a Formyl Peptide Receptor 2 (designated FPR2/3 in mice) agonist, and the protective responses were not observed in FPR2/3 -/- animals. In mice treated with LXA4 (50mg/kg/day, i.p., days 3-6 post-infection), at day 7, macrophage reprogramming was observed, as seen by a decrease in classically activated macrophages and an increase in alternatively activated macrophages in the lungs. Furthermore, the number of apoptotic cells and cells undergoing efferocytosis was increased in the lavage of treated mice. Treatment also modulated the adaptive immune response, increasing the number of anti-inflammatory T cells (Th2) and regulatory T (Tregs) cells in the lungs of the treated mice. Conclusions Therefore, treatment with a lipoxin A4 analog was beneficial in a model of influenza A infection in mice. The drug decreased inflammation and promoted resolution and beneficial immune responses, suggesting it may be useful in patients with severe influenza.
Collapse
|
8
|
Li YH, Yang YS, Xue YB, Lei H, Zhang SS, Qian J, Yao Y, Zhou R, Huang L. G protein subunit G γ13-mediated signaling pathway is critical to the inflammation resolution and functional recovery of severely injured lungs. eLife 2024; 12:RP92956. [PMID: 38836551 DOI: 10.7554/elife.92956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
Tuft cells are a group of rare epithelial cells that can detect pathogenic microbes and parasites. Many of these cells express signaling proteins initially found in taste buds. It is, however, not well understood how these taste signaling proteins contribute to the response to the invading pathogens or to the recovery of injured tissues. In this study, we conditionally nullified the signaling G protein subunit Gγ13 and found that the number of ectopic tuft cells in the injured lung was reduced following the infection of the influenza virus H1N1. Furthermore, the infected mutant mice exhibited significantly larger areas of lung injury, increased macrophage infiltration, severer pulmonary epithelial leakage, augmented pyroptosis and cell death, greater bodyweight loss, slower recovery, worsened fibrosis and increased fatality. Our data demonstrate that the Gγ13-mediated signal transduction pathway is critical to tuft cells-mediated inflammation resolution and functional repair of the damaged lungs.To our best knowledge, it is the first report indicating subtype-specific contributions of tuft cells to the resolution and recovery.
Collapse
Affiliation(s)
- Yi-Hong Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Sen Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan-Bo Xue
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Hao Lei
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sai-Sai Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yushi Yao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruhong Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
| | - Liquan Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
- Monell Chemical Senses Center, Philadelphia, United States
| |
Collapse
|
9
|
Mir RA, Tyagi A, Hussain SJ, Almalki MA, Zeyad MT, Deshmukh R, Ali S. Saffron, a Potential Bridge between Nutrition and Disease Therapeutics: Global Health Challenges and Therapeutic Opportunities. PLANTS (BASEL, SWITZERLAND) 2024; 13:1467. [PMID: 38891276 PMCID: PMC11174376 DOI: 10.3390/plants13111467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/12/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024]
Abstract
Plants are an important source of essential bioactive compounds that not only have a beneficial role in human health and nutrition but also act as drivers for shaping gut microbiome. However, the mechanism of their functional attributes is not fully understood despite their significance. One such important plant is Crocus sativus, also known as saffron, which possesses huge medicinal, nutritional, and industrial applications like food and cosmetics. The importance of this plant is grossly attributed to its incredible bioactive constituents such as crocins, crocetin, safranal, picrocrocin, and glycosides. These bioactive compounds possess a wide range of therapeutic activities against multiple human ailments. Since a huge number of studies have revealed negative unwanted side effects of modern-day drugs, the scientific communities at the global level are investigating a large number of medicinal plants to explore natural products as the best alternatives. Taken into consideration, the available research findings indicate that saffron has a huge scope to be further explored to establish alternative natural-product-based drugs for health benefits. In this review, we are providing an update on the role of bioactive compounds of saffron as therapeutic agents (human disorders and antimicrobial activity) and its nutritional values. We also highlighted the role of omics and metabolic engineering tools for increasing the content of key saffron bioactive molecules for its mass production. Finally, pre-clinical and clinical studies seem to be necessary to establish its therapeutic potential against human diseases.
Collapse
Affiliation(s)
- Rakeeb Ahmad Mir
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India
| | - Anshika Tyagi
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Sofi Javed Hussain
- Department of Botany, Central University of Kashmir, Ganderbal 191201, India;
| | - Mohammed A. Almalki
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Mohammad Tarique Zeyad
- Department of Agricultural Microbiology, Faculty of Agriculture Sciences, Aligarh Muslim University, Aligarh 202002, India;
| | - Rupesh Deshmukh
- Department of Biotechnology, Central University of Haryana, Mahendragarh 123031, India;
| | - Sajad Ali
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
10
|
Gottumukkala SB, Ganesan TS, Palanisamy A. Comprehensive molecular interaction map of TGFβ induced epithelial to mesenchymal transition in breast cancer. NPJ Syst Biol Appl 2024; 10:53. [PMID: 38760412 PMCID: PMC11101644 DOI: 10.1038/s41540-024-00378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
Breast cancer is one of the prevailing cancers globally, with a high mortality rate. Metastatic breast cancer (MBC) is an advanced stage of cancer, characterised by a highly nonlinear, heterogeneous process involving numerous singling pathways and regulatory interactions. Epithelial-mesenchymal transition (EMT) emerges as a key mechanism exploited by cancer cells. Transforming Growth Factor-β (TGFβ)-dependent signalling is attributed to promote EMT in advanced stages of breast cancer. A comprehensive regulatory map of TGFβ induced EMT was developed through an extensive literature survey. The network assembled comprises of 312 distinct species (proteins, genes, RNAs, complexes), and 426 reactions (state transitions, nuclear translocations, complex associations, and dissociations). The map was developed by following Systems Biology Graphical Notation (SBGN) using Cell Designer and made publicly available using MINERVA ( http://35.174.227.105:8080/minerva/?id=Metastatic_Breast_Cancer_1 ). While the complete molecular mechanism of MBC is still not known, the map captures the elaborate signalling interplay of TGFβ induced EMT-promoting MBC. Subsequently, the disease map assembled was translated into a Boolean model utilising CaSQ and analysed using Cell Collective. Simulations of these have captured the known experimental outcomes of TGFβ induced EMT in MBC. Hub regulators of the assembled map were identified, and their transcriptome-based analysis confirmed their role in cancer metastasis. Elaborate analysis of this map may help in gaining additional insights into the development and progression of metastatic breast cancer.
Collapse
Affiliation(s)
| | - Trivadi Sundaram Ganesan
- Department of Medical Oncology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Anbumathi Palanisamy
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, India.
| |
Collapse
|
11
|
Tubeeckx MRL, De Keulenaer GW, Heidbuchel H, Segers VFM. Pathophysiology and clinical relevance of atrial myopathy. Basic Res Cardiol 2024; 119:215-242. [PMID: 38472506 DOI: 10.1007/s00395-024-01038-0] [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: 09/30/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
Atrial myopathy is a condition that consists of electrical, structural, contractile, and autonomic remodeling of the atria and is the substrate for development of atrial fibrillation, the most common arrhythmia. Pathophysiologic mechanisms driving atrial myopathy are inflammation, oxidative stress, atrial stretch, and neurohormonal signals, e.g., angiotensin-II and aldosterone. These mechanisms initiate the structural and functional remodeling of the atrial myocardium. Novel therapeutic strategies are being developed that target the pathophysiologic mechanisms of atrial myopathy. In this review, we will discuss the pathophysiology of atrial myopathy, as well as diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Michiel R L Tubeeckx
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium.
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, ZNA Middelheim Hospital Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| |
Collapse
|
12
|
Niarakis A, Ostaszewski M, Mazein A, Kuperstein I, Kutmon M, Gillespie ME, Funahashi A, Acencio ML, Hemedan A, Aichem M, Klein K, Czauderna T, Burtscher F, Yamada TG, Hiki Y, Hiroi NF, Hu F, Pham N, Ehrhart F, Willighagen EL, Valdeolivas A, Dugourd A, Messina F, Esteban-Medina M, Peña-Chilet M, Rian K, Soliman S, Aghamiri SS, Puniya BL, Naldi A, Helikar T, Singh V, Fernández MF, Bermudez V, Tsirvouli E, Montagud A, Noël V, Ponce-de-Leon M, Maier D, Bauch A, Gyori BM, Bachman JA, Luna A, Piñero J, Furlong LI, Balaur I, Rougny A, Jarosz Y, Overall RW, Phair R, Perfetto L, Matthews L, Rex DAB, Orlic-Milacic M, Gomez LCM, De Meulder B, Ravel JM, Jassal B, Satagopam V, Wu G, Golebiewski M, Gawron P, Calzone L, Beckmann JS, Evelo CT, D’Eustachio P, Schreiber F, Saez-Rodriguez J, Dopazo J, Kuiper M, Valencia A, Wolkenhauer O, Kitano H, Barillot E, Auffray C, Balling R, Schneider R. Drug-target identification in COVID-19 disease mechanisms using computational systems biology approaches. Front Immunol 2024; 14:1282859. [PMID: 38414974 PMCID: PMC10897000 DOI: 10.3389/fimmu.2023.1282859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/22/2023] [Indexed: 02/29/2024] Open
Abstract
Introduction The COVID-19 Disease Map project is a large-scale community effort uniting 277 scientists from 130 Institutions around the globe. We use high-quality, mechanistic content describing SARS-CoV-2-host interactions and develop interoperable bioinformatic pipelines for novel target identification and drug repurposing. Methods Extensive community work allowed an impressive step forward in building interfaces between Systems Biology tools and platforms. Our framework can link biomolecules from omics data analysis and computational modelling to dysregulated pathways in a cell-, tissue- or patient-specific manner. Drug repurposing using text mining and AI-assisted analysis identified potential drugs, chemicals and microRNAs that could target the identified key factors. Results Results revealed drugs already tested for anti-COVID-19 efficacy, providing a mechanistic context for their mode of action, and drugs already in clinical trials for treating other diseases, never tested against COVID-19. Discussion The key advance is that the proposed framework is versatile and expandable, offering a significant upgrade in the arsenal for virus-host interactions and other complex pathologies.
Collapse
Affiliation(s)
- Anna Niarakis
- Université Paris-Saclay, Laboratoire Européen de Recherche pour la Polyarthrite rhumatoïde - Genhotel, Univ Evry, Evry, France
- Lifeware Group, Inria, Saclay-île de France, Palaiseau, France
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexander Mazein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Inna Kuperstein
- Institut Curie, P.S.L. Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, Netherlands
| | - Marc E. Gillespie
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- St. John’s University, Queens, NY, United States
| | - Akira Funahashi
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Marcio Luis Acencio
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ahmed Hemedan
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael Aichem
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany
| | - Karsten Klein
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany
| | - Tobias Czauderna
- Faculty of Applied Computer Sciences & Biosciences, University of Applied Sciences Mittweida, Mittweida, Germany
| | - Felicia Burtscher
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Takahiro G. Yamada
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Yusuke Hiki
- Center for Biosciences and Informatics, Graduate School of Fundamental Science and Technology, Keio University, Kanagawa, Japan
| | - Noriko F. Hiroi
- Faculty of Creative Engineering, Kanagawa Institute of Technology, Kanagawa, Japan
- Keio University School of Medicine, Tokyo, Japan
| | - Finterly Hu
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, Netherlands
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, Netherlands
| | - Nhung Pham
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, Netherlands
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, Netherlands
| | - Egon L. Willighagen
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, Netherlands
| | - Alberto Valdeolivas
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | - Aurelien Dugourd
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | - Francesco Messina
- Department of Epidemiology, Preclinical Research and Advanced Diagnostic, National Institute for Infectious Diseases’ Lazzaro Spallanzani’ - IRCCS, Rome, Italy
| | - Marina Esteban-Medina
- Computational Medicine Platform, Andalusian Public Foundation Progress and Health-FPS, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Sevilla, Spain
| | - Maria Peña-Chilet
- Computational Medicine Platform, Andalusian Public Foundation Progress and Health-FPS, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Sevilla, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Seville, Spain
| | - Kinza Rian
- Computational Medicine Platform, Andalusian Public Foundation Progress and Health-FPS, Sevilla, Spain
| | - Sylvain Soliman
- Lifeware Group, Inria, Saclay-île de France, Palaiseau, France
| | - Sara Sadat Aghamiri
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Bhanwar Lal Puniya
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Aurélien Naldi
- Lifeware Group, Inria, Saclay-île de France, Palaiseau, France
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Vidisha Singh
- Université Paris-Saclay, Laboratoire Européen de Recherche pour la Polyarthrite rhumatoïde - Genhotel, Univ Evry, Evry, France
| | | | - Viviam Bermudez
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Eirini Tsirvouli
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Arnau Montagud
- Barcelona Supercomputing Center (BSC.), Barcelona, Spain
| | - Vincent Noël
- Institut Curie, P.S.L. Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | | | | | | | - Benjamin M. Gyori
- Harvard Medical School, Laboratory of Systems Pharmacology, Boston, MA, United States
| | - John A. Bachman
- Harvard Medical School, Laboratory of Systems Pharmacology, Boston, MA, United States
| | - Augustin Luna
- Computational Biology Branch, National Library of Medicine, Bethesda, MD, United States
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Janet Piñero
- Medbioinformatics Solutions SL, Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Dept. of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Laura I. Furlong
- Medbioinformatics Solutions SL, Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Dept. of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Irina Balaur
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Adrien Rougny
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Aomi, Tokyo, Japan
- Com. Bio Big Data Open Innovation Lab. (CBBD-OIL), AIST, Aomi, Tokyo, Japan
| | - Yohan Jarosz
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Rupert W. Overall
- Institute for Biology, Humboldt University of Berlin, Berlin, Germany
| | - Robert Phair
- Integrative Bioinformatics, Inc., Mountain View, CA, United States
| | - Livia Perfetto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Lisa Matthews
- Department of Biochemistry & Molecular Pharmacology, NYU. Langone Medical Center, New York, NY, United States
| | | | | | - Luis Cristobal Monraz Gomez
- Institut Curie, P.S.L. Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | | | - Jean Marie Ravel
- Institut Curie, P.S.L. Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Bijay Jassal
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Venkata Satagopam
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Frankfurt Institute for Advanced Studies, Johann Wolfgang Goethe-Universität Frankfurt, Frankfurt am Main, Germany
| | - Guanming Wu
- Oregon Health Sciences University, Portland, OR, United States
| | - Martin Golebiewski
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
| | - Piotr Gawron
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Laurence Calzone
- Institut Curie, P.S.L. Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | | | - Chris T. Evelo
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, Netherlands
| | - Peter D’Eustachio
- Department of Biochemistry & Molecular Pharmacology, NYU. Langone Medical Center, New York, NY, United States
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany
- Faculty of Information Technology, Monash University, Clayton, Victoria, VIC, Australia
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | - Joaquin Dopazo
- Computational Medicine Platform, Andalusian Public Foundation Progress and Health-FPS, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Sevilla, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Seville, Spain
- FPS/ELIXIR-es, Hospital Virgen del Rocío, Sevilla, Spain
| | - Martin Kuiper
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC.), Barcelona, Spain
- I.C.R.E.A., Pg. Lluís Companys 23, Barcelona, Spain
| | - Olaf Wolkenhauer
- Department of Systems Biology & Bioinformatics, University of Rostock, Rostock, Germany
- Leibniz Institute for Food Systems Biology, at the Technical University Munich, Munich, Germany
| | | | - Emmanuel Barillot
- Institut Curie, P.S.L. Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | | | - Rudi Balling
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | |
Collapse
|
13
|
Natami M, Hosseini SM, Khaleel RA, Addulrahman TS, Zarei M, Asadi S, Gholami S, Mehrvar A. The role of specialized pro-resolving mediators (SPMs) in inflammatory arthritis: A therapeutic strategy. Prostaglandins Other Lipid Mediat 2024; 170:106798. [PMID: 37977352 DOI: 10.1016/j.prostaglandins.2023.106798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/28/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Rheumatoid arthritis (RA) is classified as a persistent inflammatory autoimmune disorder leading to the subsequent erosion of articular cartilage and bone tissue originating from the synovium. The fundamental objective of therapeutic interventions in RA has been the suppression of inflammation. Nevertheless, conventional medicines that lack target specificity may exhibit unpredictable effects on cell metabolism. In recent times, there has been evidence suggesting that specialized pro-resolving mediators (SPMs), which are lipid metabolites, have a role in facilitating the resolution of inflammation and the reestablishment of tissue homeostasis. SPMs are synthesized by immune cells through the enzymatic conversion of omega-3 fatty acids. In the context of RA, there is a possibility of dysregulation in the production of these SPMs. In this review, we delve into the present comprehension of the endogenous functions of SPMs in RA as lipids that exhibit pro-resolutive, protective, and immunoresolvent properties.
Collapse
Affiliation(s)
- Mohammad Natami
- Department of Urology, Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Seyed Mehdi Hosseini
- Department of Oral and Maxillofacial surgery, School of Dentistry, Azad University of Medical Science, Shiraz, Iran
| | | | | | - Mehdi Zarei
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Sahar Asadi
- Department of Community and Family Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sepideh Gholami
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Amir Mehrvar
- Taleghani Hospital Clinical Research Development Unit, Shahid Beheshti University of Medical Science, Tehran, Iran.
| |
Collapse
|
14
|
Gonzalez AL, Dungan MM, Smart CD, Madhur MS, Doran AC. Inflammation Resolution in the Cardiovascular System: Arterial Hypertension, Atherosclerosis, and Ischemic Heart Disease. Antioxid Redox Signal 2024; 40:292-316. [PMID: 37125445 PMCID: PMC11071112 DOI: 10.1089/ars.2023.0284] [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: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
Significance: Chronic inflammation has emerged as a major underlying cause of many prevalent conditions in the Western world, including cardiovascular diseases. Although targeting inflammation has emerged as a promising avenue by which to treat cardiovascular disease, it is also associated with increased risk of infection. Recent Advances: Though previously assumed to be passive, resolution has now been identified as an active process, mediated by unique immunoresolving mediators and mechanisms designed to terminate acute inflammation and promote tissue repair. Recent work has determined that failures of resolution contribute to chronic inflammation and the progression of human disease. Specifically, failure to produce pro-resolving mediators and the impaired clearance of dead cells from inflamed tissue have been identified as major mechanisms by which resolution fails in disease. Critical Issues: Drawing from a rapidly expanding body of experimental and clinical studies, we review here what is known about the role of inflammation resolution in arterial hypertension, atherosclerosis, myocardial infarction, and ischemic heart disease. For each, we discuss the involvement of specialized pro-resolving mediators and pro-reparative cell types, including T regulatory cells, myeloid-derived suppressor cells, and macrophages. Future Directions: Pro-resolving therapies offer the promise of limiting chronic inflammation without impairing host defense. Therefore, it is imperative to better understand the mechanisms underlying resolution to identify therapeutic targets. Antioxid. Redox Signal. 40, 292-316.
Collapse
Affiliation(s)
- Azuah L. Gonzalez
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew M. Dungan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - C. Duncan Smart
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Amanda C. Doran
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Torres-Rêgo M, Nogueira PCDN, Santos SPDD, Daniele-Silva A, Cavalcanti FF, Oliveira CIFBD, Rocha HAO, Fernandes-Pedrosa MDF, Silveira ER, Araújo RM. Isolation of indole alkaloids and a new norneolignan of hydroethanol extract from the stem barks of Aspidosperma nitidum Benth: Preclinical evaluation of safety and anti-inflammatory and healing properties. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117076. [PMID: 37619858 DOI: 10.1016/j.jep.2023.117076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aspidosperma nitidum Benth (Apocynaceae) is a tree found in Brazil especially in the Amazonia region, known as "carapanaúba", being used by indigenous and cabloco population in folk medicine in the treatment of malaria, leprosy, rheumatism, cancer, diabetes and inflammatory disorders. However, there are no scientific reports, up to now, to evidence its popular use as anti-inflammatory and healing agent. AIM OF THE STUDY This study aimed to isolate indole alkaloids, as well as investigate the safety, anti-inflammatory and healing properties of hydroethanol extract from the stem barks of Aspidosperma nitidum Benth (An). MATERIAL AND METHODS The compounds were isolated using diverse chromatographic methodologies and the structures were determined by extensive spectroscopic analyses. The safety was evaluated in vitro through 3-methyl-[4-5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay using murine fibroblast (3T3) and monkey kidney (Vero E6) cell lines and by the hemolytic assay, as well as, in vivo, through acute toxicity model, which the mice received a single dose of 2000 mg/kg of An, by intra-gastric (i.g.) route, and behavioral, hematological and biochemical parameters were evaluated. The anti-edematogenic effect was monitored through carrageenan-induced paw edema model, in which the rodents were treated with 50, 100 and 200 mg/kg of An by i. g., the percentage of edema (0-4 h), myeloperoxidase (MPO) and pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) levels were quantified. The anti-inflammatory activity was demonstrated through the zymosan-air-pouch model, in which the animals were treated with 50, 100 and 200 mg/kg of An by i. g, and the leukocytes number, MPO, total protein and cytokines levels were determined. In addition, the healing potential was evaluated through a skin wound model, in which the mice received 50, 100 and 200 mg/mL of An in wound area, and the wound skins were photographed and the area calculated. RESULTS In total, five compounds were isolated in the An, being a new 8,9-dinorneolignan glucoside and four known indole alkaloids. The MTT and hemolytic assays, in all concentrations of the extract, demonstrated not be cytotoxic. Acute toxicity model also evidenced no sign of toxicity or significant changes on the behavior, biochemical and hematological parameters after use of the extract. In the edematogenic model, the An reduced significantly the percentage of edema, as well as, the MPO and pro-inflammatory cytokines levels. The same form, An revealed to be efficient in decreasing the leukocytes migration (mainly polymorphonuclears), total proteins, MPO and cytokines concentrations in the zymosan-air-pouch assay. Moreover, the An revealed a healing effect, reducing the area of the skin wound. CONCLUSION Ours results evidence in the first time, the anti-inflammatory and healing property of An, justifying its use in traditional medicine. Moreover, include cytotoxicity in vitro and acute toxicity in vivo tests, which indicate the safety of use of the extract.
Collapse
Affiliation(s)
- Manoela Torres-Rêgo
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil; Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Patrícia Coelho do Nascimento Nogueira
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza, Humberto Monte Street, S/N, Campus Pici, Pici, Fortaleza, 60021-970, Brazil.
| | - Sarah Pollyana Dias Dos Santos
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Alessandra Daniele-Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Felipe França Cavalcanti
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil; Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | | | - Hugo Alexandre Oliveira Rocha
- Laboratory of Biotechnology of Natural Biopolymers, Department of Biochemistry, Bioscience Center, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, 59072-970, Natal, Brazil.
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Farias Street, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Edilberto Rocha Silveira
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza, Humberto Monte Street, S/N, Campus Pici, Pici, Fortaleza, 60021-970, Brazil.
| | - Renata Mendonça Araújo
- Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Senador Salgado Filho Avenue, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| |
Collapse
|
16
|
Zamora A, Nougué M, Verdu L, Balzan E, Draia-Nicolau T, Benuzzi E, Pujol F, Baillif V, Lacazette E, Morfoisse F, Galitzky J, Bouloumié A, Dubourdeau M, Chaput B, Fazilleau N, Malloizel-Delaunay J, Bura-Rivière A, Prats AC, Garmy-Susini B. 15-Lipoxygenase promotes resolution of inflammation in lymphedema by controlling T reg cell function through IFN-β. Nat Commun 2024; 15:221. [PMID: 38177096 PMCID: PMC10766617 DOI: 10.1038/s41467-023-43554-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/14/2023] [Indexed: 01/06/2024] Open
Abstract
Lymphedema (LD) is characterized by the accumulation of interstitial fluid, lipids and inflammatory cell infiltrate in the limb. Here, we find that LD tissues from women who developed LD after breast cancer exhibit an inflamed gene expression profile. Lipidomic analysis reveals decrease in specialized pro-resolving mediators (SPM) generated by the 15-lipoxygenase (15-LO) in LD. In mice, the loss of SPM is associated with an increase in apoptotic regulatory T (Treg) cell number. In addition, the selective depletion of 15-LO in the lymphatic endothelium induces an aggravation of LD that can be rescued by Treg cell adoptive transfer or ALOX15-expressing lentivector injections. Mechanistically, exogenous injections of the pro-resolving cytokine IFN-β restores both 15-LO expression and Treg cell number in a mouse model of LD. These results provide evidence that lymphatic 15-LO may represent a therapeutic target for LD by serving as a mediator of Treg cell populations to resolve inflammation.
Collapse
Affiliation(s)
- A Zamora
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - M Nougué
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - L Verdu
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - E Balzan
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - T Draia-Nicolau
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - E Benuzzi
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - F Pujol
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | | | - E Lacazette
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - F Morfoisse
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - J Galitzky
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - A Bouloumié
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | | | - B Chaput
- Service de Chirurgie Plastique et des Brûlés, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - N Fazilleau
- Infinity, Toulouse Institute for Infectious and Inflammatory Diseases, Inserm UMR1291, CNRS UMR5051, University of Toulouse, 31024, Toulouse, France
| | - J Malloizel-Delaunay
- Service de Médecine Vasculaire, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - A Bura-Rivière
- Service de Médecine Vasculaire, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - A C Prats
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France
| | - B Garmy-Susini
- I2MC, Université de Toulouse, Inserm UMR 1297, UT3, Toulouse, France.
| |
Collapse
|
17
|
Arciola CR, Ravaioli S, Mirzaei R, Dolzani P, Montanaro L, Daglia M, Campoccia D. Biofilms in Periprosthetic Orthopedic Infections Seen through the Eyes of Neutrophils: How Can We Help Neutrophils? Int J Mol Sci 2023; 24:16669. [PMID: 38068991 PMCID: PMC10706149 DOI: 10.3390/ijms242316669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Despite advancements in our knowledge of neutrophil responses to planktonic bacteria during acute inflammation, much remains to be elucidated on how neutrophils deal with bacterial biofilms in implant infections. Further complexity transpires from the emerging findings on the role that biomaterials play in conditioning bacterial adhesion, the variety of biofilm matrices, and the insidious measures that biofilm bacteria devise against neutrophils. Thus, grasping the entirety of neutrophil-biofilm interactions occurring in periprosthetic tissues is a difficult goal. The bactericidal weapons of neutrophils consist of the following: ready-to-use antibacterial proteins and enzymes stored in granules; NADPH oxidase-derived reactive oxygen species (ROS); and net-like structures of DNA, histones, and granule proteins, which neutrophils extrude to extracellularly trap pathogens (the so-called NETs: an allusive acronym for "neutrophil extracellular traps"). Neutrophils are bactericidal (and therefore defensive) cells endowed with a rich offensive armamentarium through which, if frustrated in their attempts to engulf and phagocytose biofilms, they can trigger the destruction of periprosthetic bone. This study speculates on how neutrophils interact with biofilms in the dramatic scenario of implant infections, also considering the implications of this interaction in view of the design of new therapeutic strategies and functionalized biomaterials, to help neutrophils in their arduous task of managing biofilms.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Paolo Dolzani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Lucio Montanaro
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy;
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| |
Collapse
|
18
|
Abstract
Although historically pre-eclampsia, preterm birth, abruption, fetal growth restriction and stillbirth have been viewed as clinically distinct entities, a growing body of literature has demonstrated that the placenta and its development is the root cause of many cases of these conditions. This has led to the term 'the great obstetrical syndromes' being coined to reflect this common origin. Although these conditions mostly manifest in the second half of pregnancy, a failure to complete deep placentation (the transition from histiotrophic placentation to haemochorial placenta at 10-18 weeks of gestation via a second wave of extravillous trophoblast invasion), is understood to be key to the pathogenesis of the great obstetrical syndromes. While the reasons that the placenta fails to achieve deep placentation remain active areas of investigation, maternal inflammation and thrombosis have been clearly implicated. From a clinical standpoint these mechanisms provide a biological explanation of how low-dose aspirin, which affects the COX-1 receptor (thrombosis) and the COX-2 receptor (inflammation), prevents not just pre-eclampsia but all the components of the great obstetrical syndromes if initiated early in pregnancy. The optimal dose of low-dose aspirin that is maximally effective in pregnancy remains a question open for further research. Additionally, other candidate medications have been identified that may also prevent pre-eclampsia, and further study of them may offer therapeutic options beyond low-dose aspirin. Interestingly, three of the eight identified compounds (hydroxychloroquine, metformin and pravastatin) are known to decrease inflammation.
Collapse
Affiliation(s)
- Matthew K Hoffman
- Departments of Obstetrics and Gynecology, Christiana Care Health Services, Newark, Delaware, USA
| |
Collapse
|
19
|
Ali O, Szabó A. Review of Eukaryote Cellular Membrane Lipid Composition, with Special Attention to the Fatty Acids. Int J Mol Sci 2023; 24:15693. [PMID: 37958678 PMCID: PMC10649022 DOI: 10.3390/ijms242115693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Biological membranes, primarily composed of lipids, envelop each living cell. The intricate composition and organization of membrane lipids, including the variety of fatty acids they encompass, serve a dynamic role in sustaining cellular structural integrity and functionality. Typically, modifications in lipid composition coincide with consequential alterations in universally significant signaling pathways. Exploring the various fatty acids, which serve as the foundational building blocks of membrane lipids, provides crucial insights into the underlying mechanisms governing a myriad of cellular processes, such as membrane fluidity, protein trafficking, signal transduction, intercellular communication, and the etiology of certain metabolic disorders. Furthermore, comprehending how alterations in the lipid composition, especially concerning the fatty acid profile, either contribute to or prevent the onset of pathological conditions stands as a compelling area of research. Hence, this review aims to meticulously introduce the intricacies of membrane lipids and their constituent fatty acids in a healthy organism, thereby illuminating their remarkable diversity and profound influence on cellular function. Furthermore, this review aspires to highlight some potential therapeutic targets for various pathological conditions that may be ameliorated through dietary fatty acid supplements. The initial section of this review expounds on the eukaryotic biomembranes and their complex lipids. Subsequent sections provide insights into the synthesis, membrane incorporation, and distribution of fatty acids across various fractions of membrane lipids. The last section highlights the functional significance of membrane-associated fatty acids and their innate capacity to shape the various cellular physiological responses.
Collapse
Affiliation(s)
- Omeralfaroug Ali
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
| | - András Szabó
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
- HUN-REN-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary
| |
Collapse
|
20
|
Nikaein N, Tuerxun K, Cedersund G, Eklund D, Kruse R, Särndahl E, Nånberg E, Thonig A, Repsilber D, Persson A, Nyman E. Mathematical models disentangle the role of IL-10 feedbacks in human monocytes upon proinflammatory activation. J Biol Chem 2023; 299:105205. [PMID: 37660912 PMCID: PMC10556785 DOI: 10.1016/j.jbc.2023.105205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023] Open
Abstract
Inflammation is one of the vital mechanisms through which the immune system responds to harmful stimuli. During inflammation, proinflammatory and anti-inflammatory cytokines interplay to orchestrate fine-tuned and dynamic immune responses. The cytokine interplay governs switches in the inflammatory response and dictates the propagation and development of the inflammatory response. Molecular pathways underlying the interplay are complex, and time-resolved monitoring of mediators and cytokines is necessary as a basis to study them in detail. Our understanding can be advanced by mathematical models that enable to analyze the system of interactions and their dynamical interplay in detail. We, therefore, used a mathematical modeling approach to study the interplay between prominent proinflammatory and anti-inflammatory cytokines with a focus on tumor necrosis factor and interleukin 10 (IL-10) in lipopolysaccharide-primed primary human monocytes. Relevant time-resolved data were generated by experimentally adding or blocking IL-10 at different time points. The model was successfully trained and could predict independent validation data and was further used to perform simulations to disentangle the role of IL-10 feedbacks during an acute inflammatory event. We used the insight to obtain a reduced predictive model including only the necessary IL-10-mediated feedbacks. Finally, the validated reduced model was used to predict early IL-10-tumor necrosis factor switches in the inflammatory response. Overall, we gained detailed insights into fine-tuning of inflammatory responses in human monocytes and present a model for further use in studying the complex and dynamic process of cytokine-regulated acute inflammation.
Collapse
Affiliation(s)
- Niloofar Nikaein
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden.
| | - Kedeye Tuerxun
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Gunnar Cedersund
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden; Department of Biomedical Engineering, Linköping University, Linköping, Sweden; Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Daniel Eklund
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Robert Kruse
- Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Department of Clinical Research Laboratory, Örebro University, Örebro, Sweden
| | - Eva Särndahl
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Eewa Nånberg
- Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden; Faculty of Medicine and Health, School of Health Sciences, Örebro University, Örebro, Sweden
| | - Antje Thonig
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Dirk Repsilber
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Alexander Persson
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Elin Nyman
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden.
| |
Collapse
|
21
|
Johnson PA, Ackerman JE, Kurowska-Stolarska M, Coles M, Buckley CD, Dakin SG. Three-dimensional, in-vitro approaches for modelling soft-tissue joint diseases. THE LANCET. RHEUMATOLOGY 2023; 5:e553-e563. [PMID: 38251499 DOI: 10.1016/s2665-9913(23)00190-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 01/23/2024]
Abstract
Diseases affecting the soft tissues of the joint represent a considerable global health burden, causing pain and disability and increasing the likelihood of developing metabolic comorbidities. Current approaches to investigating the cellular basis of joint diseases, including osteoarthritis, rheumatoid arthritis, tendinopathy, and arthrofibrosis, involve well phenotyped human tissues, animal disease models, and in-vitro tissue culture models. Inherent challenges in preclinical drug discovery have driven the development of state-of-the-art, in-vitro human tissue models to rapidly advance therapeutic target discovery. The clinical potential of such models has been substantiated through successful recapitulation of the pathobiology of cancers, generating accurate predictions of patient responses to therapeutics and providing a basis for equivalent musculoskeletal models. In this Review, we discuss the requirement to develop physiologically relevant three-dimensional (3D) culture systems that could advance understanding of the cellular and molecular basis of diseases that affect the soft tissues of the joint. We discuss the practicalities and challenges associated with modelling the complex extracellular matrix of joint tissues-including cartilage, synovium, tendon, and ligament-highlighting the importance of considering the joint as a whole organ to encompass crosstalk across tissues and between diverse cell types. The design of bespoke in-vitro models for soft-tissue joint diseases has the potential to inform functional studies of the cellular and molecular mechanisms underlying disease onset, progression, and resolution. Use of these models could inform precision therapeutic targeting and advance the field towards personalised medicine for patients with common musculoskeletal diseases.
Collapse
Affiliation(s)
- Peter A Johnson
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Jessica E Ackerman
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | | | - Mark Coles
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Christopher D Buckley
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Stephanie G Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
22
|
Chi J, Cheng J, Wang S, Li C, Chen M. Promising Anti-Inflammatory Tools: Biomedical Efficacy of Lipoxins and Their Synthetic Pathways. Int J Mol Sci 2023; 24:13282. [PMID: 37686088 PMCID: PMC10487465 DOI: 10.3390/ijms241713282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023] Open
Abstract
Lipoxins (LXs) have attracted widespread attention as a class of anti-inflammatory lipid mediators that are produced endogenously by the organism. LXs are arachidonic acid (ARA) derivatives that include four different structures: lipoxin A4 (LXA4), lipoxin B4 (LXB4), and the aspirin-induced differential isomers 15-epi-LXA4 and 15-epi-LXB4. Because of their unique biological activity of reducing inflammation in the body, LXs have great potential for neuroprotection, anti-inflammatory treatment of COVID-19, and other related diseases. The synthesis of LXs in vivo is achieved through the action of lipoxygenase (LO). As a kind of important enzyme, LO plays a major role in the physiological processes of living organisms in mammals and functions in some bacteria and fungi. This suggests new options for the synthesis of LXs in vitro. Meanwhile, there are other chemical and biochemical methods to synthesize LXs. In this review, the recent progress on physiological activity and synthetic pathways of LXs is summarized, and new insights into the synthesis of LXs in vitro are provided.
Collapse
Affiliation(s)
| | | | | | | | - Ming Chen
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
23
|
Tsuji T, Tseng YH. Adipose tissue-derived lipokines in metabolism. Curr Opin Genet Dev 2023; 81:102089. [PMID: 37473635 PMCID: PMC10528474 DOI: 10.1016/j.gde.2023.102089] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023]
Abstract
Adipose tissue is a crucial regulator of metabolism with functions that include energy storage and dissipation as well as the secretion of bioactive molecules. As the largest endocrine organ in the body, the adipose tissue produces diverse bioactive molecules, including peptides, metabolites, and extracellular vesicles, which communicate with and modulate the function of other organs. In recent years, lipid metabolites, also known as lipokines, have emerged as key signaling molecules that actively participate in multiple metabolic processes. This review highlights the latest advances in adipose tissue-derived lipokines and their underlying cellular and molecular functions. Furthermore, we offer our perspective on the future directions for adipose-derived bioactive lipids and potential therapeutic implications for obesity and its associated complications.
Collapse
Affiliation(s)
- Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
24
|
Bhattacharya P, Dhawan UK, Hussain MT, Singh P, Bhagat KK, Singhal A, Austin-Williams S, Sengupta S, Subramanian M. Efferocytes release extracellular vesicles to resolve inflammation and tissue injury via prosaposin-GPR37 signaling. Cell Rep 2023; 42:112808. [PMID: 37436891 DOI: 10.1016/j.celrep.2023.112808] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/16/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Macrophages release soluble mediators following efferocytic clearance of apoptotic cells to facilitate intercellular communication and promote the resolution of inflammation. However, whether inflammation resolution is modulated by extracellular vesicles (EVs) and vesicular mediators released by efferocytes is not known. We report that efferocyte-derived EVs express prosaposin, which binds to macrophage GPR37 to increase expression of the efferocytosis receptor Tim4 via an ERK-AP1-dependent signaling axis, leading to increased macrophage efferocytosis efficiency and accelerated resolution of inflammation. Neutralization and knockdown of prosaposin or blocking GRP37 abrogates the pro-resolution effects of efferocyte-derived EVs in vivo. Administration of efferocyte-derived EVs in a murine model of atherosclerosis is associated with an increase in lesional macrophage efferocytosis efficiency and a decrease in plaque necrosis and lesional inflammation. Thus, we establish a critical role for efferocyte-derived vesicular mediators in increasing macrophage efferocytosis efficiency and accelerating the resolution of inflammation and tissue injury.
Collapse
Affiliation(s)
- Purbasha Bhattacharya
- CSIR - Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Umesh Kumar Dhawan
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mohammed Tayab Hussain
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Praveen Singh
- CSIR - Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Karran Kiran Bhagat
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aarushi Singhal
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shani Austin-Williams
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shantanu Sengupta
- CSIR - Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manikandan Subramanian
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
25
|
Sun Q, Wang J, Jing Y, Liu J, Jin J, Wang S, Zhang J, Liu K, Chen X, Zhou H, Dong C. Influences of resolvin D1 and D2 on the risk of type 2 diabetes mellitus: a Chinese community-based cohort study. Front Immunol 2023; 14:1143456. [PMID: 37334388 PMCID: PMC10272361 DOI: 10.3389/fimmu.2023.1143456] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Background Although cellular and animal studies have reported that resolvin D1 (RvD1) and resolvin D2 (RvD2) are mechanisms involved in the development of type 2 diabetes mellitus (T2DM), the impact of RvD1 and RvD2 on the risk of T2DM at a population level remains unclear. Methods We included 2755 non-diabetic adults from a community-based cohort in China and followed them for seven years. Cox proportional hazards model was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the association of RvD1 and RvD2 with T2DM probability. Time-dependent receiver operator characteristics (ROC) curve was used to evaluate the predictive performance of RvD1 and RvD2 for the risk of T2DM based on the Chinese CDC T2DM prediction model (CDRS). Results A total of 172 incident T2DM cases were identified. Multivariate-adjusted HRs (95% CI) for T2DM across quartiles of RvD1 levels (Q1, Q2, Q3 and Q4) were 1.00, 1.64 (1.03-2.63), 1.80 (1.13-2.86) and 1.61 (1.01-2.57), respectively. Additionally, body mass index (BMI) showed a significant effect modification in the association of RvD1 with incident T2DM (P interaction = 0.026). After multivariate adjustment, the HR (95% CI) for T2DM in the fourth compared with the first quartile of RvD2 was 1.94 (95% CI: 1.24-3.03). Time-dependent ROC analysis showed that the area under time-dependent ROC curves of the "CDRS+RvD1+RvD2" model for the 3-, 5- and 7-year risk of T2DM were 0.842, 0.835 and 0.828, respectively. Conclusions Higher RvD1 and RvD2 levels are associated with a higher risk of T2DM at the population level.
Collapse
Affiliation(s)
- Qian Sun
- Department of Epidemiology and Statistics, School of Public Health, Medical College of Soochow University, Soochow, Jiangsu, China
| | - Junrong Wang
- Department of Epidemiology and Statistics, School of Public Health, Medical College of Soochow University, Soochow, Jiangsu, China
| | - Yang Jing
- Division of Non-communicable Diseases, Suzhou Industrial Park Centers for Disease Control and Prevention, Soochow, China
| | - Jingchao Liu
- Division of Non-communicable Diseases, Suzhou Wuzhong Centers for Disease Control and Prevention, Soochow, China
| | - Jianrong Jin
- Division of Non-communicable Diseases, Suzhou Wuzhong Centers for Disease Control and Prevention, Soochow, China
| | - Sudan Wang
- Department of Epidemiology and Statistics, School of Public Health, Medical College of Soochow University, Soochow, Jiangsu, China
| | - Jin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Medical College of Soochow University, Soochow, Jiangsu, China
| | - Kaili Liu
- Department of Epidemiology and Statistics, School of Public Health, Medical College of Soochow University, Soochow, Jiangsu, China
| | - Xiaofang Chen
- Division of Non-communicable Diseases, Suzhou Industrial Park Centers for Disease Control and Prevention, Soochow, China
| | - Hui Zhou
- Division of Non-communicable Diseases, Suzhou Industrial Park Centers for Disease Control and Prevention, Soochow, China
| | - Chen Dong
- Department of Epidemiology and Statistics, School of Public Health, Medical College of Soochow University, Soochow, Jiangsu, China
| |
Collapse
|
26
|
Sousa AB, Barbosa JN. The Use of Specialized Pro-Resolving Mediators in Biomaterial-Based Immunomodulation. J Funct Biomater 2023; 14:jfb14040223. [PMID: 37103313 PMCID: PMC10145769 DOI: 10.3390/jfb14040223] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/28/2023] Open
Abstract
The implantation of a biomaterial will lead to the immediate onset of an acute inflammatory response, which is of key importance in shaping the quality of the repair process. However, the return to homeostasis is critical to prevent a chronic inflammatory response that may impair the healing process. The resolution of the inflammatory response is now recognized as an active and highly regulated process, being described as specialized immunoresolvents that have a fundamental role in the termination of the acute inflammatory response. These mediators collectively coined as specialized pro-resolving mediators (SPMs) are a family of endogenous molecules that include lipoxins (Lx), resolvins (Rv), protectins (PD), maresins (Mar), Cysteinyl-SPMs (Cys-SPMs) and n-3 docosapentaenoic acid-derived SPMs (n-3 DPA-derived SPMs). SPMs have important anti-inflammatory and pro-resolutive actions such as decreasing the recruitment of polymorphonuclear leukocytes (PMNs), inducing the recruitment of anti-inflammatory macrophages, and increasing macrophage clearance of apoptotic cells through a process known as efferocytosis. Over the last years, the trend in biomaterials research has shifted towards the engineering of materials that are able to modulate the inflammatory response and thus stimulate appropriate immune responses, the so-called immunomodulatory biomaterials. These materials should be able to modulate the host immune response with the aim of creating a pro-regenerative microenvironment. In this review, we explore the potential of using of SPMs in the development of new immunomodulatory biomaterials and we propose insights for future research in this field.
Collapse
Affiliation(s)
- Ana Beatriz Sousa
- i3S-Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Judite N Barbosa
- i3S-Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
27
|
A cross-talk between sestrins, chronic inflammation and cellular senescence governs the development of age-associated sarcopenia and obesity. Ageing Res Rev 2023; 86:101852. [PMID: 36642190 DOI: 10.1016/j.arr.2023.101852] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
The rapid increase in both the lifespan and proportion of older adults is accompanied by the unprecedented rise in age-associated chronic diseases, including sarcopenia and obesity. Aging is also manifested by increased susceptibility to multiple endogenous and exogenous stresses enabling such chronic conditions to develop. Among the main physiological regulators of cellular adaption to various stress stimuli, such as DNA damage, hypoxia, and oxidative stress, are sestrins (Sesns), a family of three evolutionarily conserved proteins, Sesn1, 2, and 3. Age-associated sarcopenia and obesity are characterized by two key processes: (i) accumulation of senescent cells in the skeletal muscle and adipose tissue and (ii) creation of a systemic, chronic, low-grade inflammation (SCLGI). Presumably, failed SCLGI resolution governs the development of these chronic conditions. Noteworthy, Sesns activate senolytics, which are agents that selectively eliminate senescent cells, as well as specialized pro-resolving mediators, which are factors that physiologically provide inflammation resolution. Sesns reveal clear beneficial effects in pre-clinical models of sarcopenia and obesity. Based on these observations, we propose a novel treatment strategy for age-associated sarcopenia and obesity, complementary to the conventional therapeutic modalities: Sesn activation, SCLGI resolution, and senescent cell elimination.
Collapse
|
28
|
Hoch M, Smita S, Cesnulevicius K, Schultz M, Lescheid D, Wolkenhauer O, Gupta S. Network analyses reveal new insights into the effect of multicomponent Tr14 compared to single-component diclofenac in an acute inflammation model. J Inflamm (Lond) 2023; 20:12. [PMID: 36973809 PMCID: PMC10044762 DOI: 10.1186/s12950-023-00335-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Modifying the acute inflammatory response has wide clinical benefits. Current options include non-steroidal anti-inflammatory drugs (NSAIDs) and therapies that may resolve inflammation. Acute inflammation involves multiple cell types and various processes. We, therefore, investigated whether an immunomodulatory drug that acts simultaneously at multiple sites shows greater potential to resolve acute inflammation more effectively and with fewer side effects than a common anti-inflammatory drug developed as a small molecule for a single target. In this work, we used time-series gene expression profiles from a wound healing mouse model to compare the effects of Traumeel (Tr14), a multicomponent natural product, to diclofenac, a single component NSAID on inflammation resolution. RESULTS We advance previous studies by mapping the data onto the "Atlas of Inflammation Resolution", followed by in silico simulations and network analysis. We found that Tr14 acts primarily on the late phase of acute inflammation (during resolution) compared to diclofenac, which suppresses acute inflammation immediately after injury. CONCLUSIONS Our results provide new insights how network pharmacology of multicomponent drugs may support inflammation resolution in inflammatory conditions.
Collapse
Affiliation(s)
- Matti Hoch
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany
| | - Suchi Smita
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany
| | | | | | | | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Freising, 85354, Germany
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch, 7602, South Africa
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, 18055, Germany.
| |
Collapse
|
29
|
Dobrev D, Heijman J, Hiram R, Li N, Nattel S. Inflammatory signalling in atrial cardiomyocytes: a novel unifying principle in atrial fibrillation pathophysiology. Nat Rev Cardiol 2023; 20:145-167. [PMID: 36109633 PMCID: PMC9477170 DOI: 10.1038/s41569-022-00759-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 02/08/2023]
Abstract
Inflammation has been implicated in atrial fibrillation (AF), a very common and clinically significant cardiac rhythm disturbance, but its precise role remains poorly understood. Work performed over the past 5 years suggests that atrial cardiomyocytes have inflammatory signalling machinery - in particular, components of the NLRP3 (NACHT-, LRR- and pyrin domain-containing 3) inflammasome - that is activated in animal models and patients with AF. Furthermore, work in animal models suggests that NLRP3 inflammasome activation in atrial cardiomyocytes might be a sufficient and necessary condition for AF occurrence. In this Review, we evaluate the evidence for the role and pathophysiological significance of cardiomyocyte NLRP3 signalling in AF. We first summarize the evidence for a role of inflammation in AF and review the biochemical properties of the NLRP3 inflammasome, as defined primarily in studies of classic inflammation. We then briefly consider the broader evidence for a role of inflammatory signalling in heart disease, particularly conditions that predispose individuals to develop AF. We provide a detailed discussion of the available information about atrial cardiomyocyte NLRP3 inflammasome signalling in AF and related conditions and evaluate the possibility that similar signalling might be important in non-myocyte cardiac cells. We then review the evidence on the role of active resolution of inflammation and its potential importance in suppressing AF-related inflammatory signalling. Finally, we consider the therapeutic potential and broader implications of this new knowledge and highlight crucial questions to be addressed in future research.
Collapse
Affiliation(s)
- Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Roddy Hiram
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Na Li
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Stanley Nattel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany.
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada.
- IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
30
|
Abstract
While neutrophils are the main effectors of protective innate immune responses, they are also key players in inflammatory pathologies. Sickle cell disease (SCD) is a genetic blood disorder in which red blood cells (RBCs) are constantly destroyed in the circulation which generates a highly inflammatory environment that culminates in vascular occlusions. Vaso-occlusion is the hallmark of SCD and a predictor of disease severity. Neutrophils initiate and propagate SCD-related vaso-occlusion through adhesive interactions with the activated and dysfunctional endothelium, sickle RBCs, and platelets, leading to acute and chronic complications that progress to irreversible organ damage and ultimately death. The use of SCD humanized mouse models, in combination with in vivo imaging techniques, has emerged as a fundamental tool to understand the dynamics of neutrophils under complex inflammatory contexts and their contribution to vascular injury in SCD. In this review, we discuss the various mechanisms by which circulating neutrophils sense and respond to the wide range of stimuli present in the blood of SCD patients and mice. We argue that the central role of neutrophils in SCD can be rationalized to develop targets for the management of clinical complications in SCD patients.
Collapse
Affiliation(s)
- Lidiane S Torres
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
31
|
Yasmeen N, Selvaraj H, Lakhawat SS, Datta M, Sharma PK, Jain A, Khanna R, Srinivasan J, Kumar V. Possibility of averting cytokine storm in SARS-COV 2 patients using specialized pro-resolving lipid mediators. Biochem Pharmacol 2023; 209:115437. [PMID: 36731803 PMCID: PMC9884647 DOI: 10.1016/j.bcp.2023.115437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023]
Abstract
Fatal "cytokine storms (CS)" observed in critically ill COVID-19 patients are consequences of dysregulated host immune system and over-exuberant inflammatory response. Acute respiratory distress syndrome (ARDS), multi-system organ failure, and eventual death are distinctive symptoms, attributed to higher morbidity and mortality rates among these patients. Consequent efforts to save critical COVID-19 patients via the usage of several novel therapeutic options are put in force. Strategically, drugs being used in such patients are dexamethasone, remdesivir, hydroxychloroquine, etc. along with the approved vaccines. Moreover, it is certain that activation of the resolution process is important for the prevention of chronic diseases. Until recently Inflammation resolution was considered a passive process, rather it's an active biochemical process that can be achieved by the use of specialized pro-resolving mediators (SPMs). These endogenous mediators are an array of atypical lipid metabolites that include Resolvins, lipoxins, maresins, protectins, considered as immunoresolvents, but their role in COVID-19 is ambiguous. Recent evidence from studies such as the randomized clinical trial, in which omega 3 fatty acid was used as supplement to resolve inflammation in COVID-19, suggests that direct supplementation of SPMs or the use of synthetic SPM mimetics (which are still being explored) could enhance the process of resolution by regulating the aberrant inflammatory process and can be useful in pain relief and tissue remodeling. Here we discussed the biosynthesis of SPMs, & their mechanistic pathways contributing to inflammation resolution along with sequence of events leading to CS in COVID-19, with a focus on therapeutic potential of SPMs.
Collapse
Affiliation(s)
- Nusrath Yasmeen
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Harikrishnan Selvaraj
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Sudarshan S Lakhawat
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Manali Datta
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Pushpender K Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Ajay Jain
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Rakhi Khanna
- Rajasthan State Regional Forensic Science Laboratory, Kota, Rajasthan, India
| | | | - Vikram Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India.
| |
Collapse
|
32
|
Hoch M, Rauthe J, Cesnulevicius K, Schultz M, Lescheid D, Wolkenhauer O, Chiurchiù V, Gupta S. Cell-Type-Specific Gene Regulatory Networks of Pro-Inflammatory and Pro-Resolving Lipid Mediator Biosynthesis in the Immune System. Int J Mol Sci 2023; 24:ijms24054342. [PMID: 36901771 PMCID: PMC10001763 DOI: 10.3390/ijms24054342] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Lipid mediators are important regulators in inflammatory responses, and their biosynthetic pathways are targeted by commonly used anti-inflammatory drugs. Switching from pro-inflammatory lipid mediators (PIMs) to specialized pro-resolving (SPMs) is a critical step toward acute inflammation resolution and preventing chronic inflammation. Although the biosynthetic pathways and enzymes for PIMs and SPMs have now been largely identified, the actual transcriptional profiles underlying the immune cell type-specific transcriptional profiles of these mediators are still unknown. Using the Atlas of Inflammation Resolution, we created a large network of gene regulatory interactions linked to the biosynthesis of SPMs and PIMs. By mapping single-cell sequencing data, we identified cell type-specific gene regulatory networks of the lipid mediator biosynthesis. Using machine learning approaches combined with network features, we identified cell clusters of similar transcriptional regulation and demonstrated how specific immune cell activation affects PIM and SPM profiles. We found substantial differences in regulatory networks in related cells, accounting for network-based preprocessing in functional single-cell analyses. Our results not only provide further insight into the gene regulation of lipid mediators in the immune response but also shed light on the contribution of selected cell types in their biosynthesis.
Collapse
Affiliation(s)
- Matti Hoch
- Department of Systems Biology and Bioinformatics, University of Rostock, 18055 Rostock, Germany
| | - Jannik Rauthe
- Department of Systems Biology and Bioinformatics, University of Rostock, 18055 Rostock, Germany
| | | | | | | | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18055 Rostock, Germany
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch 7602, South Africa
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, 18055 Rostock, Germany
- Correspondence:
| |
Collapse
|
33
|
Millar B, de Gaetano M. Posing the rationale for synthetic lipoxin mimetics as an adjuvant treatment to gold standard atherosclerosis therapies. Front Pharmacol 2023; 14:1125858. [PMID: 36865918 PMCID: PMC9971729 DOI: 10.3389/fphar.2023.1125858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Atherosclerosis is a progressive, multifactorial inflammatory, and dyslipidaemic disease, responsible for the majority of cardiovascular diseases globally. The chronic inflammation is the main driver of the initiation and progression of such disease, as a result of an imbalanced lipid metabolism and an ineffective immune response to attenuate the inflammatory component. The importance of inflammation resolution is being increasingly recognised in atherosclerosis and cardiovascular disease. It has a complex mechanism consisting of multiple stages, including restoring an effective removal of apoptotic bodies (efferocytosis) and their degradation (effero-metabolism), a macrophage phenotype switching towards resolving phenotypes, and the promotion of tissue healing and regeneration. The low-grade inflammation associated with atherosclerosis development is a driving force in disease exacerbation, and hence inflammation resolution is a key area of research. In this review, we explore the complex disease pathogenesis and its many contributing factors to gain a greater understanding of the disease and identify the current and potential therapeutic targets. First-line treatments and their efficacy will also be discussed in detail, to highlight the emerging field of resolution pharmacology. Despite the great efforts made by current gold-standard treatments, such as lipid-lowering and glucose-lowering drugs, they remain ineffective at tackling residual inflammatory risk and residual cholesterol risk. Resolution pharmacology represents a new era of atherosclerosis therapy, as endogenous ligands associated with inflammation resolution are exploited for their pharmacological benefits in a more potent and longer-acting manner. Novel FPR2-agonists, such as synthetic lipoxin analogues, provide an exciting new approach to enhance the pro-resolving response of the immune system and subsequently end the pro-inflammatory response to allow for an anti-inflammatory and pro-resolving environment for tissue healing, regeneration, and return to homeostasis.
Collapse
Affiliation(s)
| | - Monica de Gaetano
- Diabetes Complications Research Centre, Conway Institute & School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Talamonti E, Jacobsson A, Chiurchiù V. Impairment of Endogenous Synthesis of Omega-3 DHA Exacerbates T-Cell Inflammatory Responses. Int J Mol Sci 2023; 24:ijms24043717. [PMID: 36835128 PMCID: PMC9966148 DOI: 10.3390/ijms24043717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Omega-3 (ω-3) polyunsaturated fatty acids, including docosahexaenoic acid (DHA), are involved in numerous biological processes and have a range of health benefits. DHA is obtained through the action of elongases (ELOVLs) and desaturases, among which Elovl2 is the key enzyme involved in its synthesis, and can be further metabolized into several mediators that regulate the resolution of inflammation. Our group has recently reported that ELOVL2 deficient mice (Elovl2-/-) not only display reduced DHA levels in several tissues, but they also have higher pro-inflammatory responses in the brain, including the activation of innate immune cells such as macrophages. However, whether impaired synthesis of DHA affects cells of adaptive immunity, i.e., T lymphocytes, is unexplored. Here we show that Elovl2-/- mice have significantly higher lymphocytes in peripheral blood and that both CD8+ and CD4+ T cell subsets produce greater amounts of pro-inflammatory cytokines in both blood and spleen compared to wild type mice, with a higher percentage of cytotoxic CD8+ T cells (CTLs) as well as IFN-γ-producing Th1 and IL-17-producing Th17 CD4+ cells. Furthermore, we also found that DHA deficiency impacts the cross-talk between dendritic cells (DC) and T cells, inasmuch as mature DCs of Elovl2-/- mice bear higher expression of activation markers (CD80, CD86 and MHC-II) and enhance the polarization of Th1 and Th17 cells. Reintroducing DHA back into the diets of Elovl2-/- mice reversed the exacerbated immune responses observed in T cells. Hence, impairment of endogenous synthesis of DHA exacerbates T cell inflammatory responses, accounting for an important role of DHA in regulating adaptive immunity and in potentially counteracting T-cell-mediated chronic inflammation or autoimmunity.
Collapse
Affiliation(s)
- Emanuela Talamonti
- Department of Biochemistry and Biophysics, Stockholm University, 114 Stockholm, Sweden
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 114 Stockholm, Sweden
| | - Anders Jacobsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 114 Stockholm, Sweden
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
- Correspondence: or
| |
Collapse
|
35
|
Abstract
Inflammation and its timely resolution are critical to ensure effective host defense and appropriate tissue repair after injury and or infection. Chronic, unresolved inflammation typifies many prevalent pathologies. The key mediators that initiate and drive the inflammatory response are well defined and targeted by conventional anti-inflammatory therapeutics. More recently, there is a growing appreciation that specific mediators, including arachidonate-derived lipoxins, are generated in self-limiting inflammatory responses to promote the resolution of inflammation and endogenous repair mechanisms without compromising host defense. We discuss the proresolving biological actions of lipoxins and recent efforts to harness their therapeutic potential through the development of novel, potent lipoxin mimetics generated via efficient, modular stereoselective synthetic pathways. We consider the evidence that lipoxin mimetics may have applications in limiting inflammation and reversing fibrosis and the underlying mechanisms.
Collapse
Affiliation(s)
- Catherine Godson
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland;
- The Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Patrick Guiry
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland;
- The Conway Institute, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
36
|
de Gaetano M. Development of synthetic lipoxin-A4 mimetics (sLXms): New avenues in the treatment of cardio-metabolic diseases. Semin Immunol 2023; 65:101699. [PMID: 36428172 DOI: 10.1016/j.smim.2022.101699] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
Resolution of inflammation is a complex, dynamic process consisting of several distinct processes, including inhibition of endothelial activation and leukocyte trafficking; promotion of inflammatory cell apoptosis and subsequent non-phlogistic scavenging and degradation; augmentation of pathogen phagocytosis; modulation of stromal cell phenotype coupled to the promotion of tissue regeneration and repair. Among these tightly regulated processes, the clearance and degradation of apoptotic cells without eliciting an inflammatory response is a crucial allostatic mechanism vital to developmental processes, host defence, and the effective resolution of inflammation. These efferocytic and subsequent effero-metabolism processes can be carried out by professional and non-professional phagocytes. Defective removal or inadequate processing of apoptotic cells leads to persistent unresolved inflammation, which may promote insidious pathologies including scarring, fibrosis, and eventual organ failure. In this manuscript, the well-established role of endothelial activation and leukocyte extravasation, as classical vascular targets of the 'inflammation pharmacology', will be briefly reviewed. The main focus of this work is to bring attention to a less explored aspect of the 'resolution pharmacology', aimed at tackling defective efferocytosis and inefficient effero-metabolism, as key targeted mechanisms to prevent or pre-empt vascular complications in cardio-metabolic diseases. Despite the use of gold standard lipid-lowering drugs or glucose-lowering drugs, none of them are able to tackle the so called residual inflammatory risk and/or the metabolic memory. In this review, the development of synthetic mimetics of endogenous mediators of inflammation is highlighted. Such molecules finely tune key components across the whole inflammatory process, amongst various other novel therapeutic paradigms that have emerged over the past decade, including anti-inflammatory therapy. More specifically, FPR2-agonists in general, and Lipoxin analogues in particular, greatly enhance the reprogramming and cross-talk between classical and non-classical innate immune cells, thus inducing both termination of the pro-inflammatory state as well as promoting the subsequent resolving phase, which represent pivotal mechanisms in inflammatory cardio-metabolic diseases.
Collapse
Affiliation(s)
- Monica de Gaetano
- Diabetes Complications Research Centre, Conway Institute & School of Biomolecular & Biomedical Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
37
|
Madrid M, Bojalil R, Brianza-Padilla M, Zapoteco-Nava J, Márquez-Velasco R, Rivera-González R. The molecular profile of the inflammatory process differs among various neurodevelopmental disorders with or without cognitive component: A hypothesis of persistent systemic dysfunction and hyper-resolution. Front Pediatr 2023; 11:1132175. [PMID: 37152315 PMCID: PMC10157392 DOI: 10.3389/fped.2023.1132175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Challenges of diverse origin in childhood can alter the growth and development of the central nervous system, affecting structures and functions. As a consequence of the damage suffered during the perinatal period, long periods of dysfunctionality may occur, such as regulatory disorders, which may result in remaining in a process of low-grade inflammation. We previously found that perinatal risks and neurological signs are associated with long-term changes in circulating concentrations of molecules of the inflammatory process, findings that are consistent with the postulate that long periods of dysfunction may condition long-lasting low-grade inflammation or parainflammation. The aim of this study was to assess whether different expressions of neurological disorders show variations in their inflammatory molecule profiles or whether there is a common pattern. Methods We included screening for (a) caregiver-perceived risk detection of regulatory disturbances, using the DeGangi instrument; (b) dysautonomia or asymmetries, through neurodevelopmental assessments; (c) cognitive developmental disturbances (using the Bailey instrument). We assessed protein molecules on a multiplex system, and lipid molecules by ELISA. Results We found a similar, although not identical, pattern of cytokine profiles with the presence of risk of regulatory disturbances, dysautonomia and asymmetries; but an opposite inflammatory profile was associated with cognitive impairment. Discussion Our results suggest that there are diverse, probably limited, molecular footprints associated with impaired function, and that these footprints may depend on the response requirements necessary to adjust to the altered internal environment. Here we propose a theoretical model that suggests possible scenarios for inflammatory outcomes associated with chronic challenges.
Collapse
Affiliation(s)
- Miriam Madrid
- Department of Health Care, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Rafael Bojalil
- Department of Health Care, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
- Correspondence: Rafael Bojalil
| | | | - Jasbet Zapoteco-Nava
- Neurodevelopmental Research Center, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Ricardo Márquez-Velasco
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | |
Collapse
|
38
|
Warnecke A, Staecker H, Rohde E, Gimona M, Giesemann A, Szczepek AJ, Di Stadio A, Hochmair I, Lenarz T. Extracellular Vesicles in Inner Ear Therapies-Pathophysiological, Manufacturing, and Clinical Considerations. J Clin Med 2022; 11:jcm11247455. [PMID: 36556073 PMCID: PMC9788356 DOI: 10.3390/jcm11247455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
(1) Background: Sensorineural hearing loss is a common and debilitating condition. To date, comprehensive pharmacologic interventions are not available. The complex and diverse molecular pathology that underlies hearing loss may limit our ability to intervene with small molecules. The current review foccusses on the potential for the use of extracellular vesicles in neurotology. (2) Methods: Narrative literature review. (3) Results: Extracellular vesicles provide an opportunity to modulate a wide range of pathologic and physiologic pathways and can be manufactured under GMP conditions allowing for their application in the human inner ear. The role of inflammation in hearing loss with a focus on cochlear implantation is shown. How extracellular vesicles may provide a therapeutic option for complex inflammatory disorders of the inner ear is discussed. Additionally, manufacturing and regulatory issues that need to be addressed to develop EVs as advanced therapy medicinal product for use in the inner ear are outlined. (4) Conclusion: Given the complexities of inner ear injury, novel therapeutics such as extracellular vesicles could provide a means to modulate inflammation, stress pathways and apoptosis in the inner ear.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence of the German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”) “Hearing4all”, 30625 Hannover, Germany
- Correspondence:
| | - Hinrich Staecker
- Department of Otolaryngology Head and Neck Surgery, University of Kansas School of Medicine, Rainbow Blvd., Kansas City, KS 66160, USA
| | - Eva Rohde
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV-TT), 5020 Salzburg, Austria
- Department of Transfusion Medicine, University Hospital, Salzburger Landeskliniken GesmbH (SALK) Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mario Gimona
- GMP Unit, Spinal Cord Injury & Tissue Regeneration Centre Salzburg (SCI-TReCS), Paracelsus Medical University, 5020 Salzburg, Austria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies (EV-TT), 5020 Salzburg, Austria
- Research Program “Nanovesicular Therapies”, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Anja Giesemann
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Agnieszka J. Szczepek
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Faculty of Medicine and Health Sciences, University of Zielona Gora, 65-046 Zielona Gora, Poland
| | - Arianna Di Stadio
- Department GF Ingrassia, University of Catania, 95124 Catania, Italy
| | | | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence of the German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”) “Hearing4all”, 30625 Hannover, Germany
| |
Collapse
|
39
|
Rizo-Téllez SA, Sekheri M, Filep JG. Myeloperoxidase: Regulation of Neutrophil Function and Target for Therapy. Antioxidants (Basel) 2022; 11:antiox11112302. [PMID: 36421487 PMCID: PMC9687284 DOI: 10.3390/antiox11112302] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Neutrophils, the most abundant white blood cells in humans, are critical for host defense against invading pathogens. Equipped with an array of antimicrobial molecules, neutrophils can eradicate bacteria and clear debris. Among the microbicide proteins is the heme protein myeloperoxidase (MPO), stored in the azurophilic granules, and catalyzes the formation of the chlorinating oxidant HOCl and other oxidants (HOSCN and HOBr). MPO is generally associated with killing trapped bacteria and inflicting collateral tissue damage to the host. However, the characterization of non-enzymatic functions of MPO suggests additional roles for this protein. Indeed, evolving evidence indicates that MPO can directly modulate the function and fate of neutrophils, thereby shaping immunity. These actions include MPO orchestration of neutrophil trafficking, activation, phagocytosis, lifespan, formation of extracellular traps, and MPO-triggered autoimmunity. This review scrutinizes the multifaceted roles of MPO in immunity, focusing on neutrophil-mediated host defense, tissue damage, repair, and autoimmunity. We also discuss novel therapeutic approaches to target MPO activity, expression, or MPO signaling for the treatment of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Salma A. Rizo-Téllez
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada
| | - Meriem Sekheri
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada
| | - János G. Filep
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada
- Correspondence: ; Tel.: +1-514-252-3400 (ext. 4662)
| |
Collapse
|
40
|
Hoch M, Ehlers L, Bannert K, Stanke C, Brauer D, Caton V, Lamprecht G, Wolkenhauer O, Jaster R, Wolfien M. In silico investigation of molecular networks linking gastrointestinal diseases, malnutrition, and sarcopenia. Front Nutr 2022; 9:989453. [DOI: 10.3389/fnut.2022.989453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
Malnutrition (MN) is a common primary or secondary complication in gastrointestinal diseases. The patient’s nutritional status also influences muscle mass and function, which can be impaired up to the degree of sarcopenia. The molecular interactions in diseases leading to sarcopenia are complex and multifaceted, affecting muscle physiology, the intestine (nutrition), and the liver at different levels. Although extensive knowledge of individual molecular factors is available, their regulatory interplay is not yet fully understood. A comprehensive overall picture of pathological mechanisms and resulting phenotypes is lacking. In silico approaches that convert existing knowledge into computationally readable formats can help unravel mechanisms, underlying such complex molecular processes. From public literature, we manually compiled experimental evidence for molecular interactions involved in the development of sarcopenia into a knowledge base, referred to as the Sarcopenia Map. We integrated two diseases, namely liver cirrhosis (LC), and intestinal dysfunction, by considering their effects on nutrition and blood secretome. We demonstrate the performance of our model by successfully simulating the impact of changing dietary frequency, glycogen storage capacity, and disease severity on the carbohydrate and muscle systems. We present the Sarcopenia Map as a publicly available, open-source, and interactive online resource, that links gastrointestinal diseases, MN, and sarcopenia. The map provides tools that allow users to explore the information on the map and perform in silico simulations.
Collapse
|
41
|
Potential Role of Phytochemical Extract from Saffron in Development of Functional Foods and Protection of Brain-Related Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6480590. [PMID: 36193081 PMCID: PMC9526642 DOI: 10.1155/2022/6480590] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022]
Abstract
The present review is designed to measure the effects of saffron extract in functional foods and its pharmacological properties against various disorders. Saffron is a traditional medicinal plant used as a food additive. The stigma of saffron has bioactive compounds such as safranal, crocin, crocetin, picrocrocin, kaempferol, and flavonoid. These bioactive compounds can be extracted using conventional (maceration, solvent extraction, soxhlet extraction, and vapor or hydrodistillation) and novel techniques (emulsion liquid membrane extraction, ultrasound-assisted extraction, enzyme-associated extraction, pulsed electric field extraction, microwave-assisted extraction, and supercritical fluid extraction). Saffron is used as a functional ingredient, natural colorant, shelf-life enhancer, and fortifying agent in developing different food products. The demand for saffron has been increasing in the pharma industry due to its protection against cardiovascular and Alzheimer disease and its antioxidant, anti-inflammatory, antitumor, and antidepressant properties. Conclusively, the phytochemical compounds of saffron improve the nutrition value of products and protect humans against various disorders.
Collapse
|
42
|
Roohbakhsh A, Etemad L, Karimi G. Resolvin D1: A key endogenous inhibitor of neuroinflammation. Biofactors 2022; 48:1005-1026. [PMID: 36176016 DOI: 10.1002/biof.1891] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/08/2022] [Indexed: 12/14/2022]
Abstract
After the initiation of inflammation, a series of processes start to resolve the inflammation. A group of endogenous lipid mediators, namely specialized pro-resolving lipid mediators is at the top list of inflammation resolution. Resolvin D1 (RvD1), is one of the lipid mediators with significant anti-inflammatory properties. It is produced from docosahexaenoic acid (omega-3 polyunsaturated fatty acid) in the body. In this article, we aimed to review the most recent findings concerning the pharmacological effects of RvD1 in the central nervous system with a focus on major neurological diseases and dysfunctions. A literature review of the past studies demonstrated that RvD1 plasma level changes during mania, depression, and Parkinson's disease. Furthermore, RVD1 and its epimer, aspirin-triggered RvD1 (AT-RvD1), have significant therapeutic effects on experimental models of ischemic and traumatic brain injuries, memory dysfunction, pain, depression, amyotrophic lateral sclerosis, and Alzheimer's and Parkinson's diseases. Interestingly, the beneficial effects of RvD1 and AT-RvD1 were mostly induced at nanomolar and micromolar concentrations implying the significant potency of these lipid mediators in treating diseases with inflammation.
Collapse
Affiliation(s)
- Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
43
|
Salamone S, Waltl L, Pompignan A, Grassi G, Chianese G, Koeberle A, Pollastro F. Phytochemical Characterization of Cannabis sativa L. Chemotype V Reveals Three New Dihydrophenanthrenoids That Favorably Reprogram Lipid Mediator Biosynthesis in Macrophages. PLANTS 2022; 11:plants11162130. [PMID: 36015434 PMCID: PMC9414986 DOI: 10.3390/plants11162130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022]
Abstract
The growing general interest surrounding Cannabis sativa L. has led to a renewal in breeding and resulted in an impressive variability of chemotypical characteristics that required the division of cannabis into different recognized chemotypes. The chemotype V has been overlooked in terms of phytochemical composition due to the almost total absence of cannabinoids, on which biomedical attention is focused. Systematic approaches addressing diverse chemotypes are, however, needed to discriminate and define phytochemical aspects beyond cannabinoids. Such thoroughly characterized chemotypes guarantee blinding in controlled studies by mimicking the sensory properties of hemp and may help to unravel the “entourage effect”. Capitalizing on the ability of cannabis to synthesize a large number of non-cannabinoid phenolic compounds, we here investigated, for the first time, the composition of the Ermo chemotype V and identified new compounds: two dihydrophenanthrenes and the methoxy-dihydrodenbinobin. All three compounds suppress pro-inflammatory leukotriene biosynthesis in activated macrophage subtypes by targeting 5-lipoxygenase, but substantially differ in their capacity to elevate the levels of specialized pro-resolving lipid mediators and their precursors in M2 macrophages. We conclude that the discovered compounds likely contribute to the anti-inflammatory properties of Cannabis sativa L. chemotype V and might promote inflammation resolution by promoting a lipid mediator class switch.
Collapse
Affiliation(s)
- Stefano Salamone
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy
- PlantaChem Srls, 28100 Novara, Italy
| | - Lorenz Waltl
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Anna Pompignan
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | | | - Giuseppina Chianese
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (A.K.); (F.P.); Tel.: +43-512-507-57903 (A.K.); +39-321-375-744 (F.P.)
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy
- PlantaChem Srls, 28100 Novara, Italy
- Correspondence: (A.K.); (F.P.); Tel.: +43-512-507-57903 (A.K.); +39-321-375-744 (F.P.)
| |
Collapse
|
44
|
Interferon-β regulates proresolving lipids to promote the resolution of acute airway inflammation. Proc Natl Acad Sci U S A 2022; 119:e2201146119. [PMID: 35878041 PMCID: PMC9351544 DOI: 10.1073/pnas.2201146119] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Acute respiratory distress syndrome is characterized by aberrant inflammatory responses, including polymorphonuclear neutrophil granulocyte dysfunction and hyperactive Toll-like receptor signaling. Timely resolution of bacterial infections depends on efficient removal of neutrophils from the inflamed tissue. Here we show that the antiviral cytokine interferon-β is essential for the resolution of neutrophil-driven airway inflammation by countering Toll-like receptor 9–mediated suppression of phagocytosis, neutrophil apoptosis, and uptake by macrophages. We also report that the beneficial effects of interferon-β are, in part, mediated by production of proresolving lipid mediators, such as 15-epi-lipoxin A4 and resolvin D1, which act through the lipoxin receptor ALX/FPR2. These findings uncover an interferon-β–initiated ALX/FPR2-centered resolution program as a potential target for facilitating the resolution of airway inflammation. Aberrant immune responses, including hyperresponsiveness to Toll-like receptor (TLR) ligands, underlie acute respiratory distress syndrome (ARDS). Type I interferons confer antiviral activities and could also regulate the inflammatory response, whereas little is known about their actions to resolve aberrant inflammation. Here we report that interferon-β (IFN-β) exerts partially overlapping, but also cooperative actions with aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 to counter TLR9-generated cues to regulate neutrophil apoptosis and phagocytosis in human neutrophils. In mice, TLR9 activation impairs bacterial clearance, prolongs Escherichia coli–evoked lung injury, and suppresses production of IFN-β and the proresolving lipid mediators 15-epi-LXA4 and resolvin D1 (RvD1) in the lung. Neutralization of endogenous IFN-β delays pulmonary clearance of E. coli and aggravates mucosal injury. Conversely, treatment of mice with IFN-β accelerates clearance of bacteria, restores neutrophil phagocytosis, promotes neutrophil apoptosis and efferocytosis, and accelerates resolution of airway inflammation with concomitant increases in 15-epi-LXA4 and RvD1 production in the lungs. Pharmacological blockade of the lipoxin receptor ALX/FPR2 partially prevents IFN-β–mediated resolution. These findings point to a pivotal role of IFN-β in orchestrating timely resolution of neutrophil and TLR9 activation–driven airway inflammation and uncover an IFN-β–initiated resolution program, activation of an ALX/FPR2-centered, proresolving lipids-mediated circuit, for ARDS.
Collapse
|
45
|
Soták M, Rajan MR, Clark M, Harms M, Rani A, Kraft JD, Tandio D, Shen T, Borkowski K, Fiehn O, Newman JW, Quiding-Järbrink M, Biörserud C, Apelgren P, Staalesen T, Hagberg CE, Boucher J, Wallenius V, Lange S, Börgeson E. Lipoxins reduce obesity-induced adipose tissue inflammation in 3D-cultured human adipocytes and explant cultures. iScience 2022; 25:104602. [PMID: 35789845 PMCID: PMC9249816 DOI: 10.1016/j.isci.2022.104602] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/16/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue inflammation drives obesity-related cardiometabolic diseases. Enhancing endogenous resolution mechanisms through administration of lipoxin A4, a specialized pro-resolving lipid mediator, was shown to reduce adipose inflammation and subsequently protects against obesity-induced systemic disease in mice. Here, we demonstrate that lipoxins reduce inflammation in 3D-cultured human adipocytes and adipose tissue explants from obese patients. Approximately 50% of patients responded particularly well to lipoxins by reducing inflammatory cytokines and promoting an anti-inflammatory M2 macrophage phenotype. Responding patients were characterized by elevated systemic levels of C-reactive protein, which causes inflammation in cultured human adipocytes. Responders appeared more prone to producing anti-inflammatory oxylipins and displayed elevated prostaglandin D2 levels, which has been interlinked with transcription of lipoxin-generating enzymes. Using explant cultures, this study provides the first proof-of-concept evidence supporting the therapeutic potential of lipoxins in reducing human adipose tissue inflammation. Our data further indicate that lipoxin treatment may require a tailored personalized-medicine approach.
Collapse
Affiliation(s)
- Matúš Soták
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Region Vaestra Goetaland, Sahlgrenska University Hospital, Department of Clinical Physiology, 41345 Gothenburg, Sweden
| | - Meenu Rohini Rajan
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Region Vaestra Goetaland, Sahlgrenska University Hospital, Department of Clinical Physiology, 41345 Gothenburg, Sweden
| | - Madison Clark
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Matthew Harms
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alankrita Rani
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Region Vaestra Goetaland, Sahlgrenska University Hospital, Department of Clinical Physiology, 41345 Gothenburg, Sweden
| | - Jamie D. Kraft
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - David Tandio
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Tong Shen
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - Kamil Borkowski
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - John W. Newman
- NIH West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
- USDA, ARS, Western Human Nutrition Research Center, Davis, USA
| | - Marianne Quiding-Järbrink
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Christina Biörserud
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Apelgren
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Vaestra Goetaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden
| | - Trude Staalesen
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Vaestra Goetaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden
| | - Carolina E. Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jeremie Boucher
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ville Wallenius
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stephan Lange
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Division of Cardiology, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Emma Börgeson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Region Vaestra Goetaland, Sahlgrenska University Hospital, Department of Clinical Physiology, 41345 Gothenburg, Sweden
| |
Collapse
|
46
|
Liotti F, Marotta M, Melillo RM, Prevete N. The Impact of Resolution of Inflammation on Tumor Microenvironment: Exploring New Ways to Control Cancer Progression. Cancers (Basel) 2022; 14:3333. [PMID: 35884394 PMCID: PMC9316558 DOI: 10.3390/cancers14143333] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/23/2022] Open
Abstract
Non-resolving inflammation is an enabling feature of cancer. A novel super-family of lipid mediators termed Specialized Pro-resolving Mediators (SPMs) have a role as bioactive molecules mediating the resolution of inflammation in cancer biology. SPMs are derived from ω-3 and ω-6 polyunsaturated fatty acids through the activity of lipoxygenases. SPMs have been described to directly modulate cancer progression by interfering with the epithelial to mesenchymal transition and invasion of cancer cells. SPMs have also been demonstrated to act on several components of the tumor microenvironment (TME). Consistently with their natural immunomodulatory and anti-inflammatory properties, SPMs are able to reprogram macrophages to favor phagocytosis of cell debris, which are an important source of pro-inflammatory and pro-angiogenic signals; sustain a direct cytotoxic immune response against cancer cells; stimulate neutrophils anti-tumor activities; and inhibit the development of regulatory T and B cells, thus indirectly leading to enhanced anti-tumor immunity. Furthermore, the resolution pathways exert crucial anti-angiogenic functions in lung, liver, and gastrointestinal cancers, and inhibit cancer-associated fibroblast differentiation and functions in hepatocellular carcinoma and pancreatic cancer. The present review will be focused on the potential protective effects of resolution pathways against cancer, exerted by modulating different components of the TME.
Collapse
Affiliation(s)
- Federica Liotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.L.); (M.M.)
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, 80131 Naples, Italy
| | - Maria Marotta
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.L.); (M.M.)
| | - Rosa Marina Melillo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.L.); (M.M.)
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, 80131 Naples, Italy
| | - Nella Prevete
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, 80131 Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
47
|
Ge J, Liu Z, Zhong Z, Wang L, Zhuo X, Li J, Jiang X, Ye XY, Xie T, Bai R. Natural terpenoids with anti-inflammatory activities: Potential leads for anti-inflammatory drug discovery. Bioorg Chem 2022; 124:105817. [DOI: 10.1016/j.bioorg.2022.105817] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/17/2022] [Accepted: 04/15/2022] [Indexed: 12/19/2022]
|
48
|
Lipidomics of Bioactive Lipids in Alzheimer's and Parkinson's Diseases: Where Are We? Int J Mol Sci 2022; 23:ijms23116235. [PMID: 35682914 PMCID: PMC9181703 DOI: 10.3390/ijms23116235] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 12/16/2022] Open
Abstract
Lipids are not only constituents of cellular membranes, but they are also key signaling mediators, thus acting as “bioactive lipids”. Among the prominent roles exerted by bioactive lipids are immune regulation, inflammation, and maintenance of homeostasis. Accumulated evidence indicates the existence of a bidirectional relationship between the immune and nervous systems, and lipids can interact particularly with the aggregation and propagation of many pathogenic proteins that are well-renowned hallmarks of several neurodegenerative disorders, including Alzheimer’s (AD) and Parkinson’s (PD) diseases. In this review, we summarize the current knowledge about the presence and quantification of the main classes of endogenous bioactive lipids, namely glycerophospholipids/sphingolipids, classical eicosanoids, pro-resolving lipid mediators, and endocannabinoids, in AD and PD patients, as well as their most-used animal models, by means of lipidomic analyses, advocating for these lipid mediators as powerful biomarkers of pathology, diagnosis, and progression, as well as predictors of response or activity to different current therapies for these neurodegenerative diseases.
Collapse
|
49
|
Hiram R. Resolution-promoting autacoids demonstrate promising cardioprotective effects against heart diseases. Mol Biol Rep 2022; 49:5179-5197. [PMID: 35142983 PMCID: PMC9262808 DOI: 10.1007/s11033-022-07230-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
Abstract
Chronic heart diseases have in common an unresolved inflammatory status. In atherosclerosis, myocarditis, myocardial infarction, or atrial fibrillation, mounting evidence suggests that unresolved inflammation contributes to the chronicity, aggravation, and morbidity of the disease. Following cardiac injury or infection, acute inflammation is a normal and required process to repair damaged tissues or eliminate pathogens and promote restoration of normal functions and structures. However, if acute inflammation is not followed by resolution, a chronic and deleterious inflammatory status may occur, characterized by the persistence of inflammatory biomarkers, promoting aggravation of myocardial pathogenesis, abnormal structural remodeling, development of cardiac fibrosis, and loss of function. Although traditional antiinflammatory strategies, including the use of COX-inhibitors, to inhibit the production of inflammation promotors failed to promote homeostasis, mounting evidence suggests that activation of specific endogenous autacoids may promote resolution and perpetuate cardioprotective effects. The recent discovery of the active mechanism of resolution suggests that proresolving signals and cellular processes may help to terminate inflammation and combat the development of its chronic profile in cardiac diseases. This review discussed (I) the preclinical and clinical evidence of inflammation-resolution in cardiac disorders including atrial fibrillation; (II) how and why many traditional antiinflammatory treatments failed to prevent or cure cardiac inflammation and fibrosis; and (III) whether new therapeutic strategies may interact with the resolution machinery to have cardioprotective effects. RvD D-series resolving, RvE E-series resolving, LXA4 lipoxin A4, MaR1 maresin-1.
Collapse
Affiliation(s)
- Roddy Hiram
- Department of Medicine, Faculty of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Research Center, 5000 Belanger, St. Montreal, QC, H1T 1C8, Canada.
| |
Collapse
|
50
|
Zhang K, Jordan PM, Pace S, Hofstetter RK, Werner M, Chen X, Werz O. Modulation of Inflammation-Related Lipid Mediator Pathways by Celastrol During Human Macrophage Polarization. J Inflamm Res 2022; 15:3285-3304. [PMID: 35676971 PMCID: PMC9169975 DOI: 10.2147/jir.s356964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/25/2022] [Indexed: 11/23/2022] Open
Abstract
Background and Purpose Celastrol (CS) is a major active ingredient of the Chinese/Asian herb Tripterygium wilfordii that is frequently used as phytomedicine to treat inflammation and autoimmune diseases. We showed before that short-term exposure to CS (1 µM) favorably impacts the biosynthesis of inflammation-related lipid mediators (LM) in human polarized macrophages by modulating the activities of different lipoxygenases (LOXs). However, whether CS regulates the expression of LOXs and other related LM-biosynthetic enzymes during macrophage polarization is unknown. Here, we investigated how CS affects LM-biosynthetic enzyme expression on the protein level and studied concomitant LM signature profiles during polarization of human monocyte-derived macrophages (MDM) towards M1- and M2-like phenotypes. Methods and Results We used LM metabololipidomics to study the long-term effects of CS on LM profile signatures after manipulation of human monocyte-derived macrophages (MDM) during polarization. Exposure of MDM to low concentrations of CS (ie, 0.2 µM) during polarization to an inflammatory M1 phenotype potently suppressed the formation of pro-inflammatory cyclooxygenase (COX)- and 5-LOX-derived LM, especially prostaglandin (PG)E2. Notably, gene and enzyme expression of COX-2 and microsomal PGE2 synthase (mPGES)-1 as well as M1 markers were strongly decreased by CS during M1-MDM polarization, along with impaired activation of nuclear factor-κB and p38 mitogen-activated protein kinase. During IL-4-induced M2 polarization, CS decreased the capacity of the resulting M2-MDM to generate pro-inflammatory COX and 5-LOX products as well but it also reduced the formation of 12/15-LOX products and specialized pro-resolving mediators, without affecting the levels of liberated fatty acid substrates. Conclusion Depending on the timing and concentration, CS not only favorably affects LOX activities in macrophages but also the expression of LM-biosynthetic enzymes during macrophage polarization connected to changes of inflammation-related LM which might be of relevance for potential application of CS to treat inflammatory disorders.
Collapse
Affiliation(s)
- Kehong Zhang
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, D-07743, Germany
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518000, People’s Republic of China
| | - Paul Mike Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, D-07743, Germany
| | - Simona Pace
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, D-07743, Germany
| | - Robert K Hofstetter
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, D-07743, Germany
| | - Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, D-07743, Germany
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, 518000, People’s Republic of China
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, D-07743, Germany
- Correspondence: Oliver Werz, Email
| |
Collapse
|