1
|
Quinlivan KM, Howard IV, Southan F, Bayer RL, Torres KL, Serhan CN, Panigrahy D. Exploring the unique role of specialized pro-resolving mediators in cancer therapeutics. Prostaglandins Other Lipid Mediat 2024; 178:106944. [PMID: 39722403 DOI: 10.1016/j.prostaglandins.2024.106944] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Unresolved chronic inflammation, a hallmark of cancer, promotes tumor growth and metastasis in various cancer types. In contrast to blocking inflammation, stimulation of resolution of inflammation is an entirely novel approach to "resolve" inflammation. Resolution of inflammation mechanisms in cancer includes clearance of tumor debris, counter-regulation of pro-inflammatory eicosanoids and cytokines, and suppression of leukocyte infiltration. Conventional cytotoxic chemotherapy, radiation, anti-angiogenic therapy, and immune checkpoint inhibitors directly or indirectly can lead to the generation of pro-tumorigenic cellular debris. Over the past two decades, a potential paradigm shift has emerged in the inflammation field with the discovery of specialized pro-resolving mediators (SPMs), including resolvins, lipoxins, maresins, and protectins. SPMs are structurally distinct families of mediators grouped together by their pro-resolving "debris-clearing" functions. "Pro-resolving" therapies are in clinical development for various inflammation-driven diseases, including cancer. SPMs, as novel cancer therapeutics, have tremendous potential to enhance current cancer therapy. The mechanisms of SPMs as anti-cancer therapeutics are under active investigation by various laboratories worldwide. Here, we explore the current appreciation of the SPMs as innovative potential treatments designed to harness the unique anti-cancer activity of SPMs.
Collapse
Affiliation(s)
- Katherine M Quinlivan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Isabella V Howard
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Franciska Southan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Rachel L Bayer
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Kimberly L Torres
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| |
Collapse
|
2
|
Elhady SS, Goda MS, Mehanna ET, El-Sayed NM, Hazem RM, Elfaky MA, Almalki AJ, Mohamed MS, Abdelhameed RFA. Ziziphus spina-christi L. extract attenuates bleomycin-induced lung fibrosis in mice via regulating TGF-β1/SMAD pathway: LC-MS/MS Metabolic profiling, chemical composition, and histology studies. Biomed Pharmacother 2024; 176:116823. [PMID: 38834008 DOI: 10.1016/j.biopha.2024.116823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/06/2024] Open
Abstract
Ancient Egyptians (including Bedouins and Nubians) have long utilized Ziziphus spina-christi (L.), a traditional Arabian medicinal herb, to alleviate swellings and inflammatory disorders. It is also mentioned in Christian and Muslim traditions. Ziziphus spina-christi L. (Family: Rhamnaceae) is a plentiful source of polyphenols, revealing free radical scavenging, antioxidant, metal chelating, cytotoxic, and anti-inflammatory activities. Herein, different classes of the existing bioactive metabolites in Z. spina-christi L. were detected using liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the first time. The study also aimed to assess the anti-inflammatory and antifibrotic properties of Z. spina-christi L. extract against bleomycin-induced lung fibrosis in an experimental mouse model. 32 male Swiss Albino mice were assigned into 4 groups; the first and second were the normal control group and the bleomycin positive control (single 2.5 U/kg bleomycin intratracheal dose). The third and fourth groups received 100 and 200 mg/kg/day Z. spina-christi L. extract orally for 3 weeks, 2 weeks before bleomycin, and 1 week after. The bioactive metabolites in Z. spina-christi L. extract were identified as phenolic acids, catechins, flavonoids, chalcones, stilbenes, triterpenoid acids, saponins, and sterols. The contents of total phenolic compounds and flavonoids were found to be 196.62 mg GAE/gm and 33.29 mg QE/gm, respectively. In the experimental study, histopathological examination revealed that lung fibrosis was attenuated in both Z. spina-christi L.- treated groups. Z. spina-christi L. extract downregulated the expression of nuclear factor kappa B (NF-κB) p65 and decreased levels of the inflammatory markers tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and c-Jun N-terminal kinase (JNK) in lung tissue. Z. spina-christi L. also downregulated the expression of the fibrotic parameters collagen-1, alpha-smooth muscle actin (α-SMA), transforming growth factor-beta 1 (TGF-β1), matrix metalloproteinase-9 (MMP-9) and SMAD3, with upregulation of the antifibrotic SMAD7 in lung tissue. Overall, the present study suggests a potential protective effect of Z. spina-christi L. extract against bleomycin-induced lung fibrosis through regulation of the TGF-β1/SMAD pathway.
Collapse
Affiliation(s)
- Sameh S Elhady
- King Abdulaziz University Herbarium, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Marwa S Goda
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Reem M Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Mahmoud A Elfaky
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmad J Almalki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Malik Suliman Mohamed
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia; Department of Pharmaceutics, Faculty of Pharmacy, Khartoum University, Khartoum 11111, Sudan
| | - Reda F A Abdelhameed
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt.
| |
Collapse
|
3
|
Kim JS, Ma SF, Ma JZ, Huang Y, Bonham CA, Oldham JM, Adegunsoye A, Strek ME, Flaherty KR, Strickland E, Udofia I, Mooney JJ, Ghosh S, Maddipati K, Noth I. Associations of Plasma Omega-3 Fatty Acids With Progression and Survival in Pulmonary Fibrosis. Chest 2024; 165:621-631. [PMID: 37866772 PMCID: PMC10925547 DOI: 10.1016/j.chest.2023.09.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/22/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Preclinical experiments suggest protective effects of omega-3 fatty acids and their metabolites in lung injury and fibrosis. Whether higher intake of omega-3 fatty acids is associated with disease progression and survival in humans with pulmonary fibrosis is unknown. RESEARCH QUESTION What are the associations of plasma omega-3 fatty acid levels (a validated marker of omega-3 nutritional intake) with disease progression and transplant-free survival in pulmonary fibrosis? STUDY DESIGN AND METHODS Omega-3 fatty acid levels were measured from plasma samples of patients with clinically diagnosed pulmonary fibrosis from the Pulmonary Fibrosis Foundation Patient Registry (n = 150), University of Virginia (n = 58), and University of Chicago (n = 101) cohorts. The N-3 index (docosahexaenoic acid + eicosapentaenoic acid) was the primary exposure variable of interest. Linear-mixed effects models with random intercept and slope were used to examine associations of plasma omega-3 fatty acid levels with changes in FVC and diffusing capacity for carbon monoxide over a period of 12 months. Cox proportional hazards models were used to examine transplant-free survival. Stratified analyses by telomere length were performed in the University of Chicago cohort. RESULTS Most of the cohort were patients with idiopathic pulmonary fibrosis (88%) and male patients (74%). One-unit increment in log-transformed N-3 index plasma level was associated with a change in diffusing capacity for carbon monoxide of 1.43 mL/min/mm Hg per 12 months (95% CI, 0.46-2.41) and a hazard ratio for transplant-free survival of 0.44 (95% CI, 0.24-0.83). Cardiovascular disease history, smoking, and antifibrotic usage did not significantly modify associations. Omega-3 fatty acid levels were not significantly associated with changes in FVC. Higher eicosapentaenoic acid plasma levels were associated with longer transplant-free survival among University of Chicago participants with shorter telomere length (P value for interaction = .02). INTERPRETATION Further research is needed to investigate underlying biological mechanisms and whether omega-3 fatty acids are a potential disease-modifying therapy.
Collapse
Affiliation(s)
- John S Kim
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA; Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY.
| | - Shwu-Fan Ma
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - Jennie Z Ma
- Department of Public Health, University of Virginia School of Medicine, Charlottesville, VA
| | - Yong Huang
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - Catherine A Bonham
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - Justin M Oldham
- Department of Medicine, University of Michigan, Ann Arbor, MI
| | | | - Mary E Strek
- Department of Medicine, University of Chicago, Chicago, IL
| | | | - Emma Strickland
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | | | | | - Shrestha Ghosh
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Department of Immunology, Harvard Medical School, Boston, MA
| | | | - Imre Noth
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
4
|
Isshiki T, Naiel S, Vierhout M, Otsubo K, Ali P, Tsubouchi K, Yazdanshenas P, Kumaran V, Dvorkin-Gheva A, Kolb MRJ, Ask K. Therapeutic strategies to target connective tissue growth factor in fibrotic lung diseases. Pharmacol Ther 2024; 253:108578. [PMID: 38103794 DOI: 10.1016/j.pharmthera.2023.108578] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The treatment of interstitial lung diseases, including idiopathic pulmonary fibrosis (IPF), remains challenging as current available antifibrotic agents are not effective in halting disease progression. Connective tissue growth factor (CTGF), also known as cellular communication factor 2 (CCN2), is a member of the CCN family of proteins that regulates cell signaling through cell surface receptors such as integrins, the activity of cytokines/growth factors, and the turnover of extracellular matrix (ECM) proteins. Accumulating evidence indicates that CTGF plays a crucial role in promoting lung fibrosis through multiple processes, including inducing transdifferentiation of fibroblasts to myofibroblasts, epithelial-mesenchymal transition (EMT), and cooperating with other fibrotic mediators such as TGF-β. Increased expression of CTGF has been observed in fibrotic lungs and inhibiting CTGF signaling has been shown to suppress lung fibrosis in several animal models. Thus, the CTGF signaling pathway is emerging as a potential therapeutic target in IPF and other pulmonary fibrotic conditions. This review provides a comprehensive overview of the current evidence on the pathogenic role of CTGF in pulmonary fibrosis and discusses the current therapeutic agents targeting CTGF using a systematic review approach.
Collapse
Affiliation(s)
- Takuma Isshiki
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada; Department of Respiratory Medicine, Toho University School of Medicine, 6-11-1 Omori Nisi, Ota-ku, Tokyo 143-8541, Japan
| | - Safaa Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Megan Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Kohei Otsubo
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Pareesa Ali
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Kazuya Tsubouchi
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Parichehr Yazdanshenas
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Vaishnavi Kumaran
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Anna Dvorkin-Gheva
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Martin R J Kolb
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada.
| |
Collapse
|
5
|
Thatcher TH, Freeberg MAT, Myo YPA, Sime PJ. Is there a role for specialized pro-resolving mediators in pulmonary fibrosis? Pharmacol Ther 2023; 247:108460. [PMID: 37244406 PMCID: PMC10335230 DOI: 10.1016/j.pharmthera.2023.108460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Pulmonary fibrotic diseases are characterized by proliferation of lung fibroblasts and myofibroblasts and excessive deposition of extracellular matrix proteins. Depending on the specific form of lung fibrosis, there can be progressive scarring of the lung, leading in some cases to respiratory failure and/or death. Recent and ongoing research has demonstrated that resolution of inflammation is an active process regulated by families of small bioactive lipid mediators termed "specialized pro-resolving mediators." While there are many reports of beneficial effects of SPMs in animal and cell culture models of acute and chronic inflammatory and immune diseases, there have been fewer reports investigating SPMs and fibrosis, especially pulmonary fibrosis. Here, we will review evidence that resolution pathways are impaired in interstitial lung disease, and that SPMs and other similar bioactive lipid mediators can inhibit fibroblast proliferation, myofibroblast differentiation, and accumulation of excess extracellular matrix in cell culture and animal models of pulmonary fibrosis, and we will consider future therapeutic implications of SPMs in fibrosis.
Collapse
Affiliation(s)
- Thomas H Thatcher
- Division of Pulmonary Care and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Margaret A T Freeberg
- Division of Pulmonary Care and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yu Par Aung Myo
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
| | - Patricia J Sime
- Division of Pulmonary Care and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
6
|
Jaiswal A, Rehman R, Dutta J, Singh S, Ray A, Shridhar M, Jaisankar J, Bhatt M, Khandelwal D, Sahoo B, Ram A, Mabalirajan U. Cellular Distribution of Secreted Phospholipase A2 in Lungs of IPF Patients and Its Inhibition in Bleomycin-Induced Pulmonary Fibrosis in Mice. Cells 2023; 12:cells12071044. [PMID: 37048117 PMCID: PMC10092981 DOI: 10.3390/cells12071044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/13/2023] [Accepted: 01/29/2023] [Indexed: 04/03/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease with a very poor prognosis as it has a 2.5 to 5 years mean survival after proper diagnosis. Even nintedanib and pirfenidone cannot halt the progression, though they slow the progression of IPF. Hence, there is a need to understand the novel pathophysiology. Phospholipase A2 (PLA2) could be the ideal candidate to study in IPF, as they have a role in both inflammation and fibrosis. In the present study, we have shown the expression profile of various secretory Phospholipase A2 (PLA2) isoforms by analyzing publicly available transcriptome data of single cells from the lungs of healthy individuals and IPF patients. Among 11 members of sPLA2, PLA2G2A is found to be increased in the fibroblasts and mesothelial cells while PLA2G5 is found to be increased in the fibroblasts of IPF patients. We identified a subset of fibroblasts expressing high PLA2G2A with moderate expression of PLA2G5 and which are specific to IPF only; we named it as PLA2G2A+ IPF fibroblast. Pathway analysis revealed that these PLA2G2A+ IPF fibroblast have upregulation of both inflammatory and fibrosis-related pathways like the TGF-β signaling pathway, IL-17 signaling, the arachidonic acid metabolism pathway and ECM-receptor interaction. In addition to this, we found elevated levels of sPLA2-IIA in plasma samples of IPF patients in our cohort. PLA2G3, PLA2G10 and PLA2G12B are found in to be increased in certain epithelial cells of IPF patients. Thus, these findings indicate that these five isoforms have a disease-dominant role along with innate immune roles as these isoforms are found predominantly in structural cells of IPF patients. Further, we have targeted sPLA2 in mice model of bleomycin-induced lung fibrosis by pBPB, a known sPLA2 inhibitor. pBPB treatment attenuated lung fibrosis induced by bleomycin along with a reduction in TGF-β and deposition of extracellular matrix in lung. Thus, these findings indicate that these sPLA2 isoforms especially PLA2G2A may serve as a therapeutic target in lung fibrosis.
Collapse
|
7
|
Melo CPB, Saito P, Martinez RM, Staurengo-Ferrari L, Pinto IC, Rodrigues CCA, Badaro-Garcia S, Vignoli JA, Baracat MM, Bussmann AJC, Georgetti SR, Verri WA, Casagrande R. Aspirin-Triggered Resolvin D1 (AT-RvD1) Protects Mouse Skin against UVB-Induced Inflammation and Oxidative Stress. Molecules 2023; 28:molecules28052417. [PMID: 36903662 PMCID: PMC10005614 DOI: 10.3390/molecules28052417] [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: 12/31/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
Intense exposure to UVB radiation incites excessive production of reactive oxygen species (ROS) and inflammation. The resolution of inflammation is an active process orchestrated by a family of lipid molecules that includes AT-RvD1, a specialized proresolving lipid mediator (SPM). AT-RvD1 is derived from omega-3, which presents anti-inflammatory activity and reduces oxidative stress markers. The present work aims to investigate the protective effect of AT-RvD1 on UVB-induced inflammation and oxidative stress in hairless mice. Animals were first treated with 30, 100, and 300 pg/animal AT-RvD1 (i.v.) and then exposed to UVB (4.14 J/cm2). The results showed that 300 pg/animal of AT-RvD1 could restrict skin edema, neutrophil and mast cell infiltration, COX-2 mRNA expression, cytokine release, and MMP-9 activity and restore skin antioxidant capacity as per FRAP and ABTS assays and control O2•- production, lipoperoxidation, epidermal thickening, and sunburn cells development. AT-RvD1 could reverse the UVB-induced downregulation of Nrf2 and its downstream targets GSH, catalase, and NOQ-1. Our results suggest that by upregulating the Nrf2 pathway, AT-RvD1 promotes the expression of ARE genes, restoring the skin's natural antioxidant defense against UVB exposition to avoid oxidative stress, inflammation, and tissue damage.
Collapse
Affiliation(s)
- Cristina P. B. Melo
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
| | - Priscila Saito
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
| | - Renata M. Martinez
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
| | - Larissa Staurengo-Ferrari
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Rodovia Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011, Londrina 86057-970, PR, Brazil
- Department of Immunology, Harvard Medical School, Blavatnik Institute, Boston, MA 02115, USA
| | - Ingrid C. Pinto
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
| | - Camilla C. A. Rodrigues
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
| | - Stephanie Badaro-Garcia
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Rodovia Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011, Londrina 86057-970, PR, Brazil
- Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Josiane A. Vignoli
- Department of Biochemistry and Biotechnology, Centre of Exact Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Marcela M. Baracat
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
| | - Allan J. C. Bussmann
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Rodovia Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011, Londrina 86057-970, PR, Brazil
| | - Sandra R. Georgetti
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
| | - Waldiceu A. Verri
- Department of Pathology, Centre of Biological Sciences, Londrina State University, Rodovia Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011, Londrina 86057-970, PR, Brazil
- Correspondence: (W.A.V.); (R.C.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Centre of Health Science, Londrina State University, Londrina 86038-350, PR, Brazil
- Correspondence: (W.A.V.); (R.C.)
| |
Collapse
|
8
|
Guilherme RF, Silva JBN, Waclawiack I, Fraga-Junior VS, Nogueira TO, Pecli C, Araújo-Silva CA, Magalhães NS, Lemos FS, Bulant CA, Blanco PJ, Serra R, Svensjö E, Scharfstein J, Moraes JA, Canetti C, Benjamim CF. Pleiotropic antifibrotic actions of aspirin-triggered resolvin D1 in the lungs. Front Immunol 2023; 14:886601. [PMID: 36960058 PMCID: PMC10030054 DOI: 10.3389/fimmu.2023.886601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 02/02/2023] [Indexed: 03/09/2023] Open
Abstract
Introduction Pulmonary fibrosis is a destructive, progressive disease that dramatically reduces life quality of patients, ultimately leading to death. Therapeutic regimens for pulmonary fibrosis have shown limited benefits, hence justifying the efforts to evaluate the outcome of alternative treatments. Methods Using a mouse model of bleomycin (BLM)-induced lung fibrosis, in the current work we asked whether treatment with pro-resolution molecules, such as pro-resolving lipid mediators (SPMs) could ameliorate pulmonary fibrosis. To this end, we injected aspirin-triggered resolvin D1 (7S,8R,17R-trihydroxy-4Z,9E,11E,13Z,15E19Z-docosahexaenoic acid; ATRvD1; i.v.) 7 and 10 days after BLM (intratracheal) challenge and samples were two weeks later. Results and discussion Assessment of outcome in the lung tissues revealed that ATRvD1 partially restored lung architecture, reduced leukocyte infiltration, and inhibited formation of interstitial edema. In addition, lung tissues from BLM-induced mice treated with ATRvD1 displayed reduced levels of TNF-α, MCP-1, IL-1-β, and TGF-β. Of further interest, ATRvD1 decreased lung tissue expression of MMP-9, without affecting TIMP-1. Highlighting the beneficial effects of ATRvD1, we found reduced deposition of collagen and fibronectin in the lung tissues. Congruent with the anti-fibrotic effects that ATRvD1 exerted in lung tissues, α-SMA expression was decreased, suggesting that myofibroblast differentiation was inhibited by ATRvD1. Turning to culture systems, we next showed that ATRvD1 impaired TGF-β-induced fibroblast differentiation into myofibroblast. After showing that ATRvD1 hampered extracellular vesicles (EVs) release in the supernatants from TGF-β-stimulated cultures of mouse macrophages, we verified that ATRvD1 also inhibited the release of EVs in the bronco-alveolar lavage (BAL) fluid of BLM-induced mice. Motivated by studies showing that BLM-induced lung fibrosis is linked to angiogenesis, we asked whether ATRvD1 could blunt BLM-induced angiogenesis in the hamster cheek pouch model (HCP). Indeed, our intravital microscopy studies confirmed that ATRvD1 abrogates BLM-induced angiogenesis. Collectively, our findings suggest that treatment of pulmonary fibrosis patients with ATRvD1 deserves to be explored as a therapeutic option in the clinical setting.
Collapse
Affiliation(s)
- Rafael F. Guilherme
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Bruno N.F. Silva
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biotecnologia, Imunobiologia e Estudos em Saúde, Universidade Federal do Tocantins, Palmas, TO, Brazil
| | - Ingrid Waclawiack
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanderlei S. Fraga-Junior
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thaís O. Nogueira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cyntia Pecli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlla A. Araújo-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathalia S. Magalhães
- Laboratório de Pesquisa em Infecção Hospitalar, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Felipe S. Lemos
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Carlos A. Bulant
- Pladema Institute, National Scientific and Technical Research Council (CONICET), Tandil, Buenos Aires, Argentina
| | - Pablo J. Blanco
- Departamento de Métodos Matemático e Computacional, Laboratório Nacional para Computação Científica, Rio de Janeiro, Brazil
| | - Rafaela Serra
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Erik Svensjö
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Júlio Scharfstein
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João A. Moraes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudio Canetti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia F. Benjamim
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Claudia F. Benjamim,
| |
Collapse
|
9
|
Resolvin D1 Prevents Epithelial-to-Mesenchymal Transition and Reduces Collagen Deposition by Stimulating Autophagy in Intestinal Fibrosis. Dig Dis Sci 2022; 67:4749-4759. [PMID: 35013877 DOI: 10.1007/s10620-021-07356-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Intestinal fibrosis is the most common complication of inflammatory bowel disease; nevertheless, specific therapies are still unavailable. Resolvin D1 (RvD1), a typical endogenous ω-3 fatty acid-derived lipid mediator, has attracted wide attention due to its remarkable anti-fibrosis effects. However, the efficacy and mechanisms of RvD1 in intestinal fibrosis remain unclear. AIM To investigate the protective effect of RvD1 in a dextran sulfate sodium (DSS)-induced intestinal fibrosis model and explore the molecular mechanisms underlying its anti-fibrotic effect. METHODS A DSS-induced intestinal fibrosis model and intestinal epithelial-to-mesenchymal transition (EMT) model were used to observe the efficacy of RvD1, and fibroblasts were stimulated with conditioned medium with or without TGF-β1 to investigate the probable mechanisms of RvD1 in intestinal fibrosis disease. RESULTS Intestinal fibrosis was effectively alleviated by RvD1 in a DSS-induced model, both preventively and therapeutically, and autophagy inhibition-induced EMT in intestinal epithelial cells was significantly suppressed in vivo and in vitro. Furthermore, RvD1 reduced epithelial cell EMT paracrine signaling, which promoted the differentiation of local fibroblasts into myofibroblasts. CONCLUSIONS Our results suggested that RvD1 reduces autophagy-induced EMT in intestinal epithelial cells and ameliorates intestinal fibrosis by disrupting epithelial-fibroblast crosstalk.
Collapse
|
10
|
Yang M, Song XQ, Han M, Liu H. The role of Resolvin D1 in liver diseases. Prostaglandins Other Lipid Mediat 2022; 160:106634. [PMID: 35292355 DOI: 10.1016/j.prostaglandins.2022.106634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023]
Abstract
The liver is a parenchymatous organ closely related to immunity, detoxification and metabolism of the three major nutrients. The inflammatory response is a protective mechanism of the body to eliminate harmful stimuli. However, continuous inflammatory stimulation leads to occurrence of many liver diseases and brings great social burden. Resolvin D1, a member of the specialized pro-resolving lipid mediators family, exerts anti-inflammatory, anti-oxidant stress, anti-fibrosis, anti-apoptotic, and anti-tumor effects by binding to ALX/FPR2 or GPR32. RvD1 plays an important role and has great therapeutic potential in liver diseases, which has been validated in multiple models of preclinical disease. This review will provide a detailed summary of the role of RvD1 in different liver diseases, including acute liver injury, liver ischemia/reperfusion injury, non-alcoholic fatty liver disease, liver fibrosis, and liver cancer, so as to help people have a more comprehensive understanding of RvD1 and promote its further research.
Collapse
Affiliation(s)
- Mei Yang
- Department of Gastroenterology, Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xian-Qi Song
- Department of Gastroenterology, Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mei Han
- Department of Gastroenterology, Second Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Hui Liu
- Department of Gastroenterology, Second Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
11
|
Molaei E, Molaei A, Hayes AW, Karimi G. Resolvin D1, therapeutic target in acute respiratory distress syndrome. Eur J Pharmacol 2021; 911:174527. [PMID: 34582846 PMCID: PMC8464084 DOI: 10.1016/j.ejphar.2021.174527] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 12/25/2022]
Abstract
Acute lung injury (ALI), or its more severe form, acute respiratory distress syndrome (ARDS), is a disease with high mortality and is a serious challenge facing the World Health Organization because there is no specific treatment. The excessive and prolonged immune response is the hallmark of this disorder, so modulating and regulating inflammation plays an important role in its prevention and treatment. Resolvin D1 (RvD1) as a specialized pro-resolving mediator has the potential to suppress the expression of inflammatory cytokines and to facilitate the production of antioxidant proteins by stimulating lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2). These changes limit the invasion of immune cells into the lung tissue, inhibit coagulation, and enhance cell protection against oxidative stress (OS). In particular, this biomolecule reduces the generation of reactive oxygen species (ROS) by blocking the activation of inflammatory transcription factors, especially nuclear factor-κB (NF-κB), and accelerating the synthesis of antioxidant compounds such as heme oxygenase 1 (HO-1) and superoxide dismutase (SOD). Therefore, the destruction and dysfunction of important cell components such as cytoplasmic membrane, mitochondria, Na+/k + adenosine triphosphatase (ATPase) and proteins involved in the phagocytic activity of scavenger macrophages are attenuated. Numerous studies on the effect of RvD1 over inflammation using animal models revealed that Rvs have both anti-inflammatory and pro-resolving capabilities and therefore, might have potential therapeutic value in treating ALI. Here, we review the current knowledge on the classification, biosynthesis, receptors, mechanisms of action, and role of Rvs in ALI/ARDS.
Collapse
Affiliation(s)
- Emad Molaei
- Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Molaei
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Scott TE, Qin CX, Drummond GR, Hobbs AJ, Kemp-Harper BK. Innovative Anti-Inflammatory and Pro-resolving Strategies for Pulmonary Hypertension: High Blood Pressure Research Council of Australia Award 2019. Hypertension 2021; 78:1168-1184. [PMID: 34565184 DOI: 10.1161/hypertensionaha.120.14525] [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: 11/16/2022]
Abstract
Pulmonary hypertension is a rare, ostensibly incurable, and etiologically diverse disease with an unacceptably high 5-year mortality rate (≈50%), worse than many cancers. Irrespective of pathogenic origin, dysregulated immune processes underlie pulmonary hypertension pathobiology, particularly pertaining to pulmonary vascular remodeling. As such, a variety of proinflammatory pathways have been mooted as novel therapeutic targets. One such pathway involves the family of innate immune regulators known as inflammasomes. In addition, a new and emerging concept is differentiating between anti-inflammatory approaches versus those that promote pro-resolving pathways. This review will briefly introduce inflammasomes and examine recent literature concerning their role in pulmonary hypertension. Moreover, it will explore the difference between inflammation-suppressing and pro-resolution approaches and how this links to inflammasomes. Finally, we will investigate new avenues for targeting inflammation in pulmonary hypertension via more targeted anti-inflammatory or inflammation resolving strategies.
Collapse
Affiliation(s)
- Tara E Scott
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute (T.E.S., B.K.K.-H.), Monash University, Parkville, VIC, Australia
- Monash University, Clayton, VIC, Australia and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences (T.E.S., C.X.Q.), Monash University, Parkville, VIC, Australia
| | - Cheng Xue Qin
- Monash University, Clayton, VIC, Australia and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences (T.E.S., C.X.Q.), Monash University, Parkville, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (C.X.Q.)
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia (G.R.D.)
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (A.J.H.)
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute (T.E.S., B.K.K.-H.), Monash University, Parkville, VIC, Australia
| |
Collapse
|
13
|
Silva JBNF, Calcia TBB, Silva CP, Guilherme RF, Almeida-Souza F, Lemos FS, Calabrese KS, Caruso-Neves C, Neves JS, Benjamim CF. ATRvD1 Attenuates Renal Tubulointerstitial Injury Induced by Albumin Overload in Sepsis-Surviving Mice. Int J Mol Sci 2021; 22:ijms222111634. [PMID: 34769064 PMCID: PMC8583751 DOI: 10.3390/ijms222111634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/15/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Novel strategies for the prevention and treatment of sepsis-associated acute kidney injury and its long-term outcomes have been required and remain a challenge in critical care medicine. Therapeutic strategies using lipid mediators, such as aspirin-triggered resolvin D1 (ATRvD1), can contribute to the resolution of acute and chronic inflammation. In this study, we examined the potential effect of ATRvD1 on long-term kidney dysfunction after severe sepsis. Fifteen days after cecal ligation and puncture (CLP), sepsis-surviving BALB/c mice were subjected to a tubulointerstitial injury through intraperitoneal injections of bovine serum albumin (BSA) for 7 days, called the subclinical acute kidney injury (subAKI) animal model. ATRvD1 treatment was performed right before BSA injections. On day 22 after CLP, the urinary protein/creatinine ratio (UPC), histologic parameters, fibrosis, cellular infiltration, apoptosis, inflammatory markers levels, and mRNA expression were determined. ATRvD1 treatment mitigated tubulointerstitial injury by reducing proteinuria excretion, the UPC ratio, the glomerular cell number, and extracellular matrix deposition. Pro-fibrotic markers, such as transforming growth factor β (TGFβ), type 3 collagen, and metalloproteinase (MMP)-3 and -9 were reduced after ATRvD1 administration. Post-septic mice treated with ATRvD1 were protected from the recruitment of IBA1+ cells. The interleukin-1β (IL-1β) levels were increased in the subAKI animal model, being attenuated by ATRvD1. Tumor necrosis factor-α (TNF-α), IL-10, and IL-4 mRNA expression were increased in the kidney of BSA-challenged post-septic mice, and it was also reduced after ATRvD1. These results suggest that ATRvD1 protects the kidney against a second insult such as BSA-induced tubulointerstitial injury and fibrosis by suppressing inflammatory and pro-fibrotic mediators in renal dysfunction after sepsis.
Collapse
Affiliation(s)
- José Bruno N. F. Silva
- Institute of Microbiology Paulo de Góes (IMPG), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.B.N.F.S.); (R.F.G.)
| | - Thayanne B. B. Calcia
- Institute of Biophysics Carlos Chagas Filho (IBCCF), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (T.B.B.C.); (C.C.-N.)
| | - Cyntia P. Silva
- Institute of Biomedical Sciences (ICB), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (C.P.S.); (F.S.L.); (J.S.N.)
| | - Rafael F. Guilherme
- Institute of Microbiology Paulo de Góes (IMPG), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.B.N.F.S.); (R.F.G.)
| | - Fernando Almeida-Souza
- Laboratory of Immunomodulation and Protozoology, Oswaldo Cruz Institute (IOC), Fiocruz, Rio de Janeiro 21040-900, Brazil; (F.A.-S.); (K.S.C.)
- Postgraduate in Animal Science, State University of Maranhão, São Luís 65055-310, Brazil
| | - Felipe S. Lemos
- Institute of Biomedical Sciences (ICB), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (C.P.S.); (F.S.L.); (J.S.N.)
| | - Kátia S. Calabrese
- Laboratory of Immunomodulation and Protozoology, Oswaldo Cruz Institute (IOC), Fiocruz, Rio de Janeiro 21040-900, Brazil; (F.A.-S.); (K.S.C.)
| | - Celso Caruso-Neves
- Institute of Biophysics Carlos Chagas Filho (IBCCF), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (T.B.B.C.); (C.C.-N.)
| | - Josiane S. Neves
- Institute of Biomedical Sciences (ICB), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (C.P.S.); (F.S.L.); (J.S.N.)
| | - Claudia F. Benjamim
- Institute of Biophysics Carlos Chagas Filho (IBCCF), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (T.B.B.C.); (C.C.-N.)
- Correspondence: or ; Tel.: +55-21-3938-6709
| |
Collapse
|
14
|
Liu GJ, Tao T, Zhang XS, Lu Y, Wu LY, Gao YY, Wang H, Dai HB, Zhou Y, Zhuang Z, Hang CH, Li W. Resolvin D1 Attenuates Innate Immune Reactions in Experimental Subarachnoid Hemorrhage Rat Model. Mol Neurobiol 2021; 58:1963-1977. [PMID: 33411245 DOI: 10.1007/s12035-020-02237-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 11/25/2020] [Indexed: 12/22/2022]
Abstract
Excessive inflammation is a major cause contributing to early brain injury (EBI) and is associated with negative or catastrophic outcomes of subarachnoid hemorrhage (SAH). Resolvin D1 (RvD1) exerts strong anti-inflammatory and pro-resolving effects on either acute or chronic inflammation of various origin. Henceforth, we hypothesized that RvD1 potentially attenuates excessive inflammation in EBI following SAH. Therefore, we generated a filament perforation SAH model and administered 3 different doses (0.3, 0.6, and 1.2 nmol) of RvD1 after experimental SAH. Neurological scores, brain edema, and blood-brain barrier integrity were evaluated; besides, neutrophil infiltration, neuronal deaths, and microglial pro-inflammatory polarization were observed using histopathology or immunofluorescence staining, western blots, and qPCR. After confirming the effectiveness of RvD1 in SAH, we administered the FPR2-specific antagonist Trp-Arg-Trp-Trp-Trp-Trp-NH2 (WRW4) 30 min before SAH establishment to observe whether this compound could abolish the anti-inflammatory effect of RvD1. Altogether, our results showed that RvD1 exerted a strong anti-inflammatory effect and markedly reduced neutrophil infiltration and microglial pro-inflammatory activation, leading to remarkable improvements in neurological function and brain tissue restoration. After addition of WRW4, the anti-inflammatory effects of RvD1 were abolished. These results indicated that RvD1 could exert a good anti-inflammatory effect and alleviate EBI, which suggested that RvD1 might be a novel therapeutic alternative for SAH-induced injury.
Collapse
Affiliation(s)
- Guang-Jie Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Tao Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xiang-Sheng Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ling-Yun Wu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yong-Yue Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Han Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Southern Medical University (Guangzhou), Nanjing, China
| | - Hai-Bin Dai
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yan Zhou
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
15
|
Perakakis N, Stefanakis K, Mantzoros CS. The role of omics in the pathophysiology, diagnosis and treatment of non-alcoholic fatty liver disease. Metabolism 2020; 111S:154320. [PMID: 32712221 PMCID: PMC7377759 DOI: 10.1016/j.metabol.2020.154320] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifaceted metabolic disorder, whose spectrum covers clinical, histological and pathophysiological developments ranging from simple steatosis to non-alcoholic steatohepatitis (NASH) and liver fibrosis, potentially evolving into cirrhosis, hepatocellular carcinoma and liver failure. Liver biopsy remains the gold standard for diagnosing NAFLD, while there are no specific treatments. An ever-increasing number of high-throughput Omics investigations on the molecular pathobiology of NAFLD at the cellular, tissue and system levels produce comprehensive biochemical patient snapshots. In the clinical setting, these applications are considerably enhancing our efforts towards obtaining a holistic insight on NAFLD pathophysiology. Omics are also generating non-invasive diagnostic modalities for the distinct stages of NAFLD, that remain though to be validated in multiple, large, heterogenous and independent cohorts, both cross-sectionally as well as prospectively. Finally, they aid in developing novel therapies. By tracing the flow of information from genomics to epigenomics, transcriptomics, proteomics, metabolomics, lipidomics and glycomics, the chief contributions of these techniques in understanding, diagnosing and treating NAFLD are summarized herein.
Collapse
Affiliation(s)
- Nikolaos Perakakis
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA..
| | - Konstantinos Stefanakis
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
16
|
Avanoǧlu Güler A, Rossi FW, Bellando-Randone S, Prevete N, Tufan A, Manetti M, de Paulis A, Matucci-Cerinic M. The Role of Endogenous Eicosapentaenoic Acid and Docosahexaenoic Acid-Derived Resolvins in Systemic Sclerosis. Front Immunol 2020; 11:1249. [PMID: 32636845 PMCID: PMC7318896 DOI: 10.3389/fimmu.2020.01249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/18/2020] [Indexed: 12/19/2022] Open
Abstract
Resolvins, the member of specialized pro-resolving mediators, are produced from omega-3 polyunsaturated fatty acids as a response to an acute inflammatory process in that termination and resolution of inflammation. In the acute inflammation, these lipid mediators limit polymorphonuclear cells infiltration, proinflammatory cytokine production; promote efferocytosis, and regulate several cell types being important roles in innate and adaptive immunity. Any dysregulation or defect of the resolution phase result in prolonged, persistent inflammation and eventually fibrosis. Resolvins are implicated in the development of various chronic autoimmune diseases. Systemic sclerosis (SSc) is a very complicated, chronic autoimmune disorder proceeding with vasculopathy, inflammation, and fibrosis. Dysregulation of innate and adaptive immunity is another important contributing factor in the pathogenesis of SSc. In this review, we will focus on the different roles of this new family of lipid mediators, characterized by the ability to prevent the spread of inflammation and its chronicity in various ways and how they can control the development of fibrotic diseases like SSc.
Collapse
Affiliation(s)
- Aslıhan Avanoǧlu Güler
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
- Department of Internal Medicine, Division of Rheumatology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Francesca Wanda Rossi
- Department of Internal Medicine, Clinical Immunology and Rheumatology, University of Naples Federico II, Naples, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
| | - Nella Prevete
- Department of Internal Medicine, Clinical Immunology and Rheumatology, University of Naples Federico II, Naples, Italy
| | - Abdurrahman Tufan
- Department of Internal Medicine, Division of Rheumatology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
| | - Amato de Paulis
- Department of Internal Medicine, Clinical Immunology and Rheumatology, University of Naples Federico II, Naples, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
| |
Collapse
|
17
|
Agudelo CW, Samaha G, Garcia-Arcos I. Alveolar lipids in pulmonary disease. A review. Lipids Health Dis 2020; 19:122. [PMID: 32493486 PMCID: PMC7268969 DOI: 10.1186/s12944-020-01278-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Lung lipid metabolism participates both in infant and adult pulmonary disease. The lung is composed by multiple cell types with specialized functions and coordinately acting to meet specific physiologic requirements. The alveoli are the niche of the most active lipid metabolic cell in the lung, the type 2 cell (T2C). T2C synthesize surfactant lipids that are an absolute requirement for respiration, including dipalmitoylphosphatidylcholine. After its synthesis and secretion into the alveoli, surfactant is recycled by the T2C or degraded by the alveolar macrophages (AM). Surfactant biosynthesis and recycling is tightly regulated, and dysregulation of this pathway occurs in many pulmonary disease processes. Alveolar lipids can participate in the development of pulmonary disease from their extracellular location in the lumen of the alveoli, and from their intracellular location in T2C or AM. External insults like smoke and pollution can disturb surfactant homeostasis and result in either surfactant insufficiency or accumulation. But disruption of surfactant homeostasis is also observed in many chronic adult diseases, including chronic obstructive pulmonary disease (COPD), and others. Sustained damage to the T2C is one of the postulated causes of idiopathic pulmonary fibrosis (IPF), and surfactant homeostasis is disrupted during fibrotic conditions. Similarly, surfactant homeostasis is impacted during acute respiratory distress syndrome (ARDS) and infections. Bioactive lipids like eicosanoids and sphingolipids also participate in chronic lung disease and in respiratory infections. We review the most recent knowledge on alveolar lipids and their essential metabolic and signaling functions during homeostasis and during some of the most commonly observed pulmonary diseases.
Collapse
Affiliation(s)
- Christina W Agudelo
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Ghassan Samaha
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Itsaso Garcia-Arcos
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 11203, USA.
| |
Collapse
|
18
|
Park GT, Kwon YW, Lee TW, Kwon SG, Ko HC, Kim MB, Kim JH. Formyl Peptide Receptor 2 Activation Ameliorates Dermal Fibrosis and Inflammation in Bleomycin-Induced Scleroderma. Front Immunol 2019; 10:2095. [PMID: 31552041 PMCID: PMC6733889 DOI: 10.3389/fimmu.2019.02095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/20/2019] [Indexed: 02/02/2023] Open
Abstract
Systemic sclerosis is a profibrotic autoimmune disease mediated by the dysregulation of extracellular matrix synthesis. Formyl peptide receptor 2 (Fpr2) is a G protein-coupled receptor that modulates inflammation and host defense by regulating the activation of inflammatory cells, such as macrophages. However, the role of Fpr2 in the development and therapy of scleroderma is still unclear. The present study was conducted to investigate the effects of Fpr2 activation in the treatment of scleroderma fibrosis. We found that intradermal administration of WKYMVm, an Fpr2-specific agonist, alleviated bleomycin-induced scleroderma fibrosis in mice and decreased dermal thickness in scleroderma skin. WKYMVm-treated scleroderma skin tissues displayed reduced numbers of myofibroblasts expressing α-smooth muscle actin, Vimentin, and phosphorylated SMAD3. WKYMVm treatment attenuated macrophage infiltration in scleroderma skin and reduced the number of M2 macrophages. The therapeutic effects of WKYMVm in scleroderma-associated fibrosis and inflammation were completely abrogated in Fpr2 knockout mice. Moreover, WKYMVm treatment reduced the serum levels of inflammatory cytokines, such as tumor necrosis factor-α, and interferon-γ, in the scleroderma model of wild-type mice but not in Fpr2 knockout mice. These results suggest that WKYMVm-induced activation of Fpr2 leads to alleviation of fibrosis by stimulating immune resolution in systemic sclerosis.
Collapse
Affiliation(s)
- Gyu Tae Park
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Yang Woo Kwon
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Tae Wook Lee
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Seong Gyu Kwon
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Hyun-Chang Ko
- Department of Dermatology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Moon Bum Kim
- Department of Dermatology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea.,Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan-si, South Korea
| |
Collapse
|
19
|
Abstract
The laboratory mouse Mus musculus has long been used as a model organism to test hypotheses and treatments related to understanding the mechanisms of disease in humans; however, for these experiments to be relevant, it is important to know the complex ways in which mice are similar to humans and, crucially, the ways in which they differ. In this chapter, an in-depth analysis of these similarities and differences is provided to allow researchers to use mouse models of human disease and primary cells derived from these animal models under the most appropriate and meaningful conditions. Although there are considerable differences between mice and humans, particularly regarding genetics, physiology, and immunology, a more thorough understanding of these differences and their effects on the function of the whole organism will provide deeper insights into relevant disease mechanisms and potential drug targets for further clinical investigation. Using specific examples of mouse models of human lung disease, i.e., asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis, this chapter explores the most salient features of mouse models of human disease and provides a full assessment of the advantages and limitations of these models, focusing on the relevance of disease induction and their ability to replicate critical features of human disease pathophysiology and response to treatment. The chapter concludes with a discussion on the future of using mice in medical research with regard to ethical and technological considerations.
Collapse
|
20
|
Rago F, Melo EM, Kraemer L, Galvão I, Cassali GD, Santos RAS, Russo RC, Teixeira MM. Effect of preventive or therapeutic treatment with angiotensin 1–7 in a model of bleomycin‐induced lung fibrosis in mice. J Leukoc Biol 2019; 106:677-686. [DOI: 10.1002/jlb.ma1218-490rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/19/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Flávia Rago
- Laboratório de ImunofarmacologiaDepartamento de Bioquímica e ImunologiaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Eliza Mathias Melo
- Laboratório de ImunofarmacologiaDepartamento de Bioquímica e ImunologiaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Lucas Kraemer
- Laboratorio de Imunologia e Mecânica PulmonarDepartamento de Fisiologia e BiofísicaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Izabela Galvão
- Laboratório de ImunofarmacologiaDepartamento de Bioquímica e ImunologiaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Geovanni D. Cassali
- Departamento de Patologia GeralUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Robson A. S. Santos
- Departamento de Fisiologia e BiofísicaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Remo C. Russo
- Laboratorio de Imunologia e Mecânica PulmonarDepartamento de Fisiologia e BiofísicaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Mauro Martins Teixeira
- Laboratório de ImunofarmacologiaDepartamento de Bioquímica e ImunologiaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
- Centro de Desenvolvimento de FármacosInstituto de Ciências BiológicasUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| |
Collapse
|
21
|
Shichino S, Ueha S, Hashimoto S, Otsuji M, Abe J, Tsukui T, Deshimaru S, Nakajima T, Kosugi-Kanaya M, Shand FH, Inagaki Y, Shimano H, Matsushima K. Transcriptome network analysis identifies protective role of the LXR/SREBP-1c axis in murine pulmonary fibrosis. JCI Insight 2019; 4:122163. [PMID: 30626759 DOI: 10.1172/jci.insight.122163] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022] Open
Abstract
Pulmonary fibrosis (PF) is an intractable disorder with a poor prognosis. Although lung fibroblasts play a central role in PF, the key regulatory molecules involved in this process remain unknown. To address this issue, we performed a time-course transcriptome analysis on lung fibroblasts of bleomycin- and silica-treated murine lungs. We found gene modules whose expression kinetics were associated with the progression of PF and human idiopathic PF (IPF). Upstream analysis of a transcriptome network helped in identifying 55 hub transcription factors that were highly connected with PF-associated gene modules. Of these hubs, the expression of Srebf1 decreased in line with progression of PF and human IPF, suggesting its suppressive role in fibroblast activation. Consistently, adoptive transfer and genetic modification studies revealed that the hub transcription factor SREBP-1c suppressed PF-associated gene expression changes in lung fibroblasts and PF pathology in vivo. Moreover, therapeutic pharmacological activation of LXR, an SREBP-1c activator, suppressed the Srebf1-dependent activation of fibroblasts and progression of PF. Thus, SREBP-1c acts as a protective hub of lung fibroblast activation in PF. Collectively, the findings of the current study may prove to be valuable in the development of effective therapeutic strategies for PF.
Collapse
Affiliation(s)
- Shigeyuki Shichino
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shinichi Hashimoto
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan.,Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Mikiya Otsuji
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Jun Abe
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Tatsuya Tsukui
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shungo Deshimaru
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Takuya Nakajima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Mizuha Kosugi-Kanaya
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Francis Hw Shand
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Chiba, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
22
|
Krishnamoorthy N, Abdulnour REE, Walker KH, Engstrom BD, Levy BD. Specialized Proresolving Mediators in Innate and Adaptive Immune Responses in Airway Diseases. Physiol Rev 2018; 98:1335-1370. [PMID: 29717929 DOI: 10.1152/physrev.00026.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Airborne pathogens and environmental stimuli evoke immune responses in the lung. It is critical to health that these responses be controlled to prevent tissue damage and the compromise of organ function. Resolution of inflammation is a dynamic process that is coordinated by biochemical and cellular mechanisms. Recently, specialized proresolving mediators (SPMs) have been identified in resolution exudates. These molecules orchestrate anti-inflammatory and proresolving actions that are cell type specific. In this review, we highlight SPM biosynthesis, the influence of SPMs on the innate and adaptive immune responses in the lung, as well as recent insights from SPMs on inflammatory disease pathophysiology. Uncovering these mediators and cellular mechanisms for resolution is providing new windows into physiology and disease pathogenesis.
Collapse
Affiliation(s)
- Nandini Krishnamoorthy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Raja-Elie E Abdulnour
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Katherine H Walker
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Braden D Engstrom
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts ; and Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
23
|
Musso G, Cassader M, Paschetta E, Gambino R. Bioactive Lipid Species and Metabolic Pathways in Progression and Resolution of Nonalcoholic Steatohepatitis. Gastroenterology 2018; 155:282-302.e8. [PMID: 29906416 DOI: 10.1053/j.gastro.2018.06.031] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 02/06/2023]
Abstract
The prevalence of nonalcoholic steatohepatitis (NASH) is increasing worldwide, yet there are no effective treatments. A decade has passed since the initial lipidomics analyses of liver tissues from patients with nonalcoholic fatty liver disease. We have learned that liver cells from patients with NASH have an abnormal lipid composition and that the accumulation of lipids leads to organelle dysfunction, cell injury and death, and chronic inflammation, called lipotoxicity. We review the lipid species and metabolic pathways that contribute to the pathogenesis of NASH and potential therapeutic targets, including enzymes involved in fatty acid and triglyceride synthesis, bioactive sphingolipids and polyunsaturated-derived eicosanoids, and specialized pro-resolving lipid mediators. We discuss the concept that NASH is a disease that can resolve and the roles of lipid molecules in the resolution of inflammation and regression of fibrosis.
Collapse
Affiliation(s)
| | - Maurizio Cassader
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | | | - Roberto Gambino
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| |
Collapse
|
24
|
Targeting formyl peptide receptors to facilitate the resolution of inflammation. Eur J Pharmacol 2018; 833:339-348. [PMID: 29935171 DOI: 10.1016/j.ejphar.2018.06.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/08/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022]
Abstract
The formyl peptide receptors (FPRs) are G protein coupled receptors that recognize a broad range of structurally distinct pathogen and danger-associated molecular patterns and mediate host defense to infection and tissue injury. It became evident that the cellular distribution and biological functions of FPRs extend beyond myeloid cells and governing their activation and trafficking. In recent years, significant progress has been made to position FPRs at check points that control the resolution of inflammation, tissue repair and return to homeostasis. Accumulating data indicate a role for FPRs in an ever-increasing range of human diseases, including atherosclerosis, chronic obstructive pulmonary disease, asthma, autoimmune diseases and cancer, in which dysregulated or defective resolution are increasingly recognized as critical component of the pathogenesis. This review summarizes recent advances on how FPRs recognize distinct ligands and integrate opposing cues to govern various responses and will discuss how this knowledge could be harnessed for developing novel therapeutic strategies to counter inflammation that underlies many human diseases.
Collapse
|
25
|
Zhang Z, Hu X, Qi X, Di G, Zhang Y, Wang Q, Zhou Q. Resolvin D1 promotes corneal epithelial wound healing and restoration of mechanical sensation in diabetic mice. Mol Vis 2018; 24:274-285. [PMID: 29643724 PMCID: PMC5881880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/30/2018] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To investigate the effect and mechanism of proresolving lipid mediator resolvin D1 (RvD1) on the corneal epithelium and the restoration of mechanical sensation in diabetic mice. METHODS Type 1 diabetes was induced in mice with intraperitoneal streptozocin injections. The healthy and diabetic mice underwent removal of the central corneal epithelium, and then 100 ng/ml RvD1 or its formyl peptide receptor 2 (FPR2) antagonist WRW4 was used to treat the diabetic mice. Regeneration of the corneal epithelium and nerves was observed with sodium fluorescein staining and whole-mount anti-β3-tubulin fluorescence staining. The inflammatory response level was measured with hematoxylin and eosin staining (inflammatory cell infiltration), enzyme-linked immunosorbent assay (tumor necrosis factor alpha and interleukin-1 beta content), myeloperoxidase activity, and fluorescence staining (macrophage content). The reactive oxygen species (ROS) and glutathione (GSH) levels were examined with incubation with fluorescent probes, and oxidative stress-related protein expression levels were evaluated with fluorescence staining and western blotting. RESULTS Topical application of RvD1 promoted regeneration of the corneal epithelium in diabetic mice, accompanied by the reactivation of signaling and inflammation resolution related to regeneration of the epithelium. Furthermore, RvD1 directly attenuated the accumulation of ROS and nicotinamide adenine dinucleotide phosphate oxidase 2/4 expression, while RvD1 enhanced GSH synthesis and reactivated the Nrf2-ARE signaling pathway that was impaired in the corneal epithelium in the diabetic mice. More interestingly, topical application of RvD1 promoted regeneration of corneal nerves and completely restored impaired mechanical sensitivity of the cornea in diabetic mice. In addition, the promotion of corneal epithelial wound healing by RvD1 in diabetic mice was abolished by its FPR2 antagonist WRW4. CONCLUSIONS Topical application of RvD1 promotes corneal epithelial wound healing and the restoration of mechanical sensation in diabetic mice, which may be related to the lipid mediator's regulation of inflammation resolution, the reactivation of regenerative signaling in the epithelium, and the attenuation of oxidative stress.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Xiaoli Hu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Xia Qi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Guohu Di
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Yangyang Zhang
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Qian Wang
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| |
Collapse
|
26
|
Specialized Proresolving Mediators: Enhancing Nonalcoholic Steatohepatitis and Fibrosis Resolution. Trends Pharmacol Sci 2018; 39:387-401. [PMID: 29499974 DOI: 10.1016/j.tips.2018.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/06/2018] [Accepted: 01/09/2018] [Indexed: 12/13/2022]
|
27
|
Understanding novel mechanisms of microbial pathogenesis in chronic lung disease: implications for new therapeutic targets. Clin Sci (Lond) 2018; 132:375-379. [PMID: 29439118 DOI: 10.1042/cs20171261] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/31/2017] [Accepted: 01/13/2018] [Indexed: 12/20/2022]
Abstract
Airway infections are considered as one of the vital factors driving the pathophysiology of chronic lung disease with significant influences on disease trajectory. Opportunistic lung microbes in diseased conditions induce excessive exacerbations and contribute to airflow limitation. Though there has been considerable amount of information that ascertains their links with airway inflammation, the intricate interaction in clinical conditions are poorly understood and requires further deciphering. Current therapeutic interventions for such pathologies are few and lack the ability to modulate underlying dysfunctional immunity as well as suppress the excessive infectious conditions. Thus, in this Commentary we provide a focused outlook on the mechanisms involved in microbial infestation in lung diseases and provides important information on new therapeutic interventions including the potential role of Resolvins and their derivatives as alternative therapeutic agents in combating such multifaceted pathological mechanisms.
Collapse
|
28
|
ResolvinD 1 stimulates epithelial wound repair and inhibits TGF-β-induced EMT whilst reducing fibroproliferation and collagen production. J Transl Med 2018; 98:130-140. [PMID: 29083412 PMCID: PMC5754464 DOI: 10.1038/labinvest.2017.114] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 08/09/2017] [Accepted: 08/21/2017] [Indexed: 01/18/2023] Open
Abstract
Acute and chronic inflammatory lung diseases are often associated with epithelial cell injury/loss and fibroproliferative responses. ResolvinD1 (RvD1) is biosynthesized during the resolution phase of inflammatory response and exerts potent anti-inflammatory and promotes resolution of inflammatory lung diseases. The aim of this study was to investigate whether RvD1 exerts protective effects on alveolar epithelial cell function/differentiation and protects against fibroproliferative stimuli. Primary human alveolar type II cells were used to model the effects of RvD1 in vitro upon wound repair, proliferation, apoptosis, transdifferentiation, and epithelial-mesenchymal transition (EMT). Effects of RvD1 upon primary human lung fibroblast proliferation, collagen production, and myofibroblast differentiation were also examined. RvD1 promoted alveolar type II (ATII) cell wound repair and proliferation. RvD1 protected ATII cells against sFas-ligand/TNF-α-induced apoptosis and inhibition on cell proliferation and viability. RvD1 promoted ATII cells transdifferentiation. Moreover, we demonstrate that RvD1 inhibited EMT in response to TGF-β. Furthermore RvD1 inhibited human lung fibroblast proliferation, collagen production, and myofibroblast differentiation induced by both TGF-β and bronchoalveolar lavage fluid from acute respiratory distress syndrome (ARDS) patients. The effects of RvD1 were PI3-kinase dependent and mediated via the resolvin receptor. RvD1 seems to promote alveolar epithelial repair by stimulating ATII cells wound repair, proliferation, reducing apoptosis, and inhibiting TGF-β-induced EMT. While RvD1 reduced fibroproliferation, collagen production, and myofibroblast differentiation. Together, these results suggest a potential new therapeutic strategy for preventing and treating chronic diseases (such as idiopathic pulmonary fibrosis) as well as the fibroproliferative phase of ARDS by targeting RvD1 actions that emphasizes natural resolution signaling pathways.
Collapse
|
29
|
Fredman G, Tabas I. Boosting Inflammation Resolution in Atherosclerosis: The Next Frontier for Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1211-1221. [PMID: 28527709 DOI: 10.1016/j.ajpath.2017.01.018] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 01/30/2017] [Indexed: 02/08/2023]
Abstract
Defective inflammation resolution is the underlying cause of prevalent chronic inflammatory diseases, such as arthritis, asthma, cancer, and neurodegenerative and cardiovascular diseases. Inflammation resolution is governed by several endogenous factors, including fatty acid-derived specialized proresolving mediators and proteins, such as annexin A1. Specifically, specialized proresolving mediators comprise a family of mediators that include arachidonic acid-derived lipoxins, omega-3 fatty acid eicosapentaenoic acid-derived resolvins, docosahexaenoic acid-derived resolvins, protectins, and maresins. Emerging evidence indicates that imbalances between specialized proresolving mediators and proinflammatory mediators are associated with several prevalent human diseases, including atherosclerosis. Mechanisms that drive this imbalance remain largely unknown and will be discussed in this review. Furthermore, the concept of dysregulated inflammation resolution in atherosclerosis has been known for several decades. Recently, there has been an explosion of new work with regard to the therapeutic application of proresolving ligands in experimental atherosclerosis. Therefore, this review will highlight recent advances in our understanding of how inflammation resolution may become defective in atherosclerosis and the potential for proresolving therapeutics in atherosclerosis. Last, we offer insight for future implications of the field.
Collapse
Affiliation(s)
- Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, Albany Medical College, Albany, New York.
| | - Ira Tabas
- Departments of Medicine, Pathology and Cell Biology, and Physiology, Columbia University Medical Center, New York, New York
| |
Collapse
|
30
|
Guo J, Lin Q, Shao Y, Rong L, Zhang D. BMP‑7 suppresses excessive scar formation by activating the BMP‑7/Smad1/5/8 signaling pathway. Mol Med Rep 2017. [PMID: 28627680 PMCID: PMC5561965 DOI: 10.3892/mmr.2017.6779] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Scarring is the inevitable consequence of wound repair, which may cause significant physical and mental pain to patients when excessive. Bone morphogenetic protein-7 (BMP-7) has been proved to inhibit TGF-β-induced fibrosis in various tissues including dermal papilla cells. However, the effect of BMP-7 on hypertrophic scarring, a common proliferative disorder of dermal fibroblasts, has not been determined. To overcome this problem, the present study established a mouse model of thermal injury to investigate the inhibitory effects of BMP-7 on scar formation. The histological analysis of scar tissues was performed by H&E and Masson's trichrome staining. Western blot assay was used to determine the level changes of related proteins and TUNEL assay was performed to assess the apoptosis of scar tissues. The results demonstrated that BMP-7 promoted wound healing and inhibited scar formation when compared with untreated mice. Collagen deposition and the expression of fibrotic proteins were suppressed in the scar tissues of mice treated with BMP-7. In addition, BMP-7 induced fibroblast apoptosis in scar tissues. Furthermore, activation of the BMP-7/Smad1/5/8 signaling pathway may have been involved in the inhibitory effects of BMP-7 on scar formation. In conclusion, the results of the present study indicate that BMP-7 may inhibit excessive scar formation via activation of the BMP-7/Smad1/5/8 signaling pathway. The results present a potential alternative therapeutic strategy for the treatment of hypertrophic scarring.
Collapse
Affiliation(s)
- Jingdong Guo
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Quan Lin
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ying Shao
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Li Rong
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Duo Zhang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
31
|
Brennan EP, Cacace A, Godson C. Specialized pro-resolving mediators in renal fibrosis. Mol Aspects Med 2017; 58:102-113. [PMID: 28479307 DOI: 10.1016/j.mam.2017.05.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/28/2017] [Accepted: 05/01/2017] [Indexed: 12/31/2022]
Abstract
Inflammation and its timely resolution play a critical role in effective host defence and wound healing. Unresolved inflammatory responses underlie the pathology of many prevalent diseases resulting in tissue fibrosis and eventual organ failure as typified by kidney, lung and liver fibrosis. The role of autocrine and paracrine mediators including cytokines, prostaglandins and leukotrienes in initiating and sustaining inflammation is well established. More recently a physiological role for specialized pro-resolving lipid mediators [SPMs] in modulating inflammatory responses and promoting the resolution of inflammation has been appreciated. As will be discussed in this review, SPMs not only attenuate the development of fibrosis through promoting the resolution of inflammation but may also directly suppress fibrotic responses. These findings suggest novel therapeutic paradigms to treat intractable life-limiting diseases such as renal fibrosis.
Collapse
Affiliation(s)
- Eoin P Brennan
- UCD Diabetes Complications Research Centre, UCD Conway Institute & UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Antonino Cacace
- UCD Diabetes Complications Research Centre, UCD Conway Institute & UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, UCD Conway Institute & UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
32
|
Specialized pro-resolving mediators in cardiovascular diseases. Mol Aspects Med 2017; 58:65-71. [PMID: 28257820 DOI: 10.1016/j.mam.2017.02.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/26/2017] [Indexed: 12/31/2022]
Abstract
The resolution of inflammation is a highly regulated process enacted by endogenous mediators including specialized pro-resolving lipid mediators (SPMs): the lipoxins, resolvins, protectins and maresins. SPMs activate specific cellular receptors to temper the production of pro-inflammatory mediators, diminish the recruitment of neutrophils, and promote the clearance of dead cells by macrophages. These mediators also enhance host-defense and couple resolution of inflammation to subsequent phases of tissue repair. Given that unresolved inflammation plays a causal role in the development of cardiovascular diseases, an understanding of these endogenous pro-resolving processes is critical for determining why cardiovascular inflammation does not resolve. Here, we discuss the receptor-dependent actions of resolvins and related pro-resolving mediators and highlight their emerging roles in the cardiovascular system. We propose that stimulating resolution could be a novel approach for treating chronic cardiovascular inflammation without promoting immunosuppression.
Collapse
|
33
|
Fredman G, Hellmann J, Proto JD, Kuriakose G, Colas RA, Dorweiler B, Connolly ES, Solomon R, Jones DM, Heyer EJ, Spite M, Tabas I. An imbalance between specialized pro-resolving lipid mediators and pro-inflammatory leukotrienes promotes instability of atherosclerotic plaques. Nat Commun 2016; 7:12859. [PMID: 27659679 PMCID: PMC5036151 DOI: 10.1038/ncomms12859] [Citation(s) in RCA: 331] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022] Open
Abstract
Chronic unresolved inflammation plays a causal role in the development of advanced atherosclerosis, but the mechanisms that prevent resolution in atherosclerosis remain unclear. Here, we use targeted mass spectrometry to identify specialized pro-resolving lipid mediators (SPM) in histologically-defined stable and vulnerable regions of human carotid atherosclerotic plaques. The levels of SPMs, particularly resolvin D1 (RvD1), and the ratio of SPMs to pro-inflammatory leukotriene B4 (LTB4), are significantly decreased in the vulnerable regions. SPMs are also decreased in advanced plaques of fat-fed Ldlr−/− mice. Administration of RvD1 to these mice during plaque progression restores the RvD1:LTB4 ratio to that of less advanced lesions and promotes plaque stability, including decreased lesional oxidative stress and necrosis, improved lesional efferocytosis, and thicker fibrous caps. These findings provide molecular support for the concept that defective inflammation resolution contributes to the formation of clinically dangerous plaques and offer a mechanistic rationale for SPM therapy to promote plaque stability. Atherosclerosis progression is linked to inflammatory processes in the blood vessel wall. Here, the authors show that, with the progression of atherosclerosis, the resolution of inflammation is impaired as the result of an imbalance between specialized pro-resolving lipid mediators and leukotrienes.
Collapse
Affiliation(s)
- Gabrielle Fredman
- Department of Anesthesiology, Perioperative and Pain Medicine, Departments of Medicine, Pathology &Cell Biology, and Physiology, Columbia University Medical Center, 630 West 168th Street, New York, New York 10032, USA.,The Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| | - Jason Hellmann
- Department of Anesthesiology, Perioperative and Pain Medicine, The Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jonathan D Proto
- Department of Anesthesiology, Perioperative and Pain Medicine, Departments of Medicine, Pathology &Cell Biology, and Physiology, Columbia University Medical Center, 630 West 168th Street, New York, New York 10032, USA
| | - George Kuriakose
- Department of Anesthesiology, Perioperative and Pain Medicine, Departments of Medicine, Pathology &Cell Biology, and Physiology, Columbia University Medical Center, 630 West 168th Street, New York, New York 10032, USA
| | - Romain A Colas
- Department of Anesthesiology, Perioperative and Pain Medicine, The Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bernhard Dorweiler
- Division of Vascular Surgery, Department of Cardiothoracic and Vascular Surgery, University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, Mainz D-55131, Germany
| | - E Sander Connolly
- Department of Neurosurgery, Columbia University Medical Center, New York, New York 10032, USA
| | - Robert Solomon
- Department of Neurosurgery, Columbia University Medical Center, New York, New York 10032, USA
| | - David M Jones
- The Department of Pathology, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| | - Eric J Heyer
- Department of Anesthesiology, Columbia University Medical Center, New York, New York 10032, USA
| | - Matthew Spite
- Department of Anesthesiology, Perioperative and Pain Medicine, The Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ira Tabas
- Department of Anesthesiology, Perioperative and Pain Medicine, Departments of Medicine, Pathology &Cell Biology, and Physiology, Columbia University Medical Center, 630 West 168th Street, New York, New York 10032, USA
| |
Collapse
|
34
|
Yatomi M, Hisada T, Ishizuka T, Koga Y, Ono A, Kamide Y, Seki K, Aoki-Saito H, Tsurumaki H, Sunaga N, Kaira K, Dobashi K, Yamada M, Okajima F. 17(R)-resolvin D1 ameliorates bleomycin-induced pulmonary fibrosis in mice. Physiol Rep 2015; 3:3/12/e12628. [PMID: 26660549 PMCID: PMC4760456 DOI: 10.14814/phy2.12628] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a destructive inflammatory disease with limited therapeutic options. Inflammation plays an integral role in the development of pulmonary fibrosis. Unresolved inflammatory responses can lead to substantial tissue injury, chronic inflammation, and fibrosis. The resolvins are a family of endogenous ω‐3 fatty acid derived‐lipid mediators of inflammation resolution. Resolvin D1 (RvD1) displays potent anti‐inflammatory, pro‐resolving activity, without causing immunosuppression. Its epimer, 17(R)‐resolvin D1 (17(R)‐RvD1), exhibits equivalent functionality to RvD1. In addition, 17(R)‐RvD1 is resistant to rapid inactivation by eicosanoid oxidoreductases. In the present study, we tested the hypothesis that 17(R)‐RvD1 can provide a therapeutic benefit in IPF by reducing inflammation and pulmonary fibrosis, while leaving the normal immune response intact. Mice were exposed to bleomycin (BLM) via micro‐osmotic pump to induce pulmonary fibrosis, and were then treated with 17(R)‐RvD1 or vehicle by intraperitoneal injection. Administration of 17(R)‐RvD1 from the start of BLM treatment attenuated neutrophil alveolar infiltration, lung collagen content, and Interleukin‐1β (IL‐1β), transforming growth factor‐β1 (TGF‐β1), connective tissue growth factor (CTGF), and type I collagen mRNA expression, along with subsequent reduction in histologically detectable fibrosis. The 17(R)‐RvD1‐induced infiltration of inflammatory cells was inhibited by an antagonist of lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2). The administration of 17(R)‐RvD1 at the later fibrotic stage also improved the lung failure. These results suggest that 17(R)‐RvD1 attenuates pulmonary fibrosis by promoting the resolution of neutrophilic inflammation and also provides pulmonary restoration. These data highlight the therapeutic potential of 17(R)‐RvD1 in the management of this intractable disease.
Collapse
Affiliation(s)
- Masakiyo Yatomi
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Takeshi Hisada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tamotsu Ishizuka
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun Fukui, Japan
| | - Yasuhiko Koga
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihiro Ono
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yosuke Kamide
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kaori Seki
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Haruka Aoki-Saito
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hiroaki Tsurumaki
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Noriaki Sunaga
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kyoichi Kaira
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kunio Dobashi
- Graduate School of Health Sciences, Gunma University, Maebashi, Japan
| | - Masanobu Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Fumikazu Okajima
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|