1
|
Ping P, Yang T, Ning C, Zhao Q, Zhao Y, Yang T, Gao Z, Fu S. Chlorogenic acid attenuates cardiac hypertrophy via up-regulating Sphingosine-1-phosphate receptor1 to inhibit endoplasmic reticulum stress. ESC Heart Fail 2024; 11:1580-1593. [PMID: 38369950 PMCID: PMC11098655 DOI: 10.1002/ehf2.14707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 10/06/2023] [Accepted: 01/23/2024] [Indexed: 02/20/2024] Open
Abstract
AIMS Cardiac hypertrophy, an adaptive response of the heart to stress overload, is closely associated with heart failure and sudden cardiac death. This study aimed to investigate the therapeutic effects of chlorogenic acid (CGA) on cardiac hypertrophy and elucidate the underlying mechanisms. METHODS AND RESULTS To simulate cardiac hypertrophy, myocardial cells were exposed to isoproterenol (ISO, 10 μM). A rat model of ISO-induced cardiac hypertrophy was also established. The expression levels of cardiac hypertrophy markers, endoplasmic reticulum stress (ERS) markers, and apoptosis markers were measured using quantitative reverse transcription PCR and western blotting. The apoptosis level, size of myocardial cells, and heart tissue pathological changes were determined by terminal deoxynucleotidyl transferase dUTP nick-end labelling staining, immunofluorescence staining, haematoxylin and eosin staining, and Masson's staining. We found that CGA treatment decreased the size of ISO-treated H9c2 cells. Moreover, CGA inhibited ISO-induced up-regulation of cardiac hypertrophy markers (atrial natriuretic peptide, brain natriuretic peptide, and β-myosin heavy chain), ERS markers (C/EBP homologous protein, glucose regulatory protein 78, and protein kinase R-like endoplasmic reticulum kinase), and apoptosis markers (bax and cleaved caspase-12/9/3) but increased the expression of anti-apoptosis marker bcl-2 in a dose-dependent way (0, 10, 50, and 100 μM). Knockdown of sphingosine-1-phosphate receptor 1 (S1pr1) reversed the protective effect of CGA on cardiac hypertrophy, ERS, and apoptosis in vitro (P < 0.05). CGA also restored ISO-induced inhibition on the AMP-activated protein kinase (AMPK)/sirtuin 1 (SIRT1) signalling in H9c2 cells, while S1pr1 knockdown abolished these CGA-induced effects (P < 0.05). CGA (90 mg/kg/day, for six consecutive days) protected rats against cardiac hypertrophy in vivo (P < 0.05). CONCLUSIONS CGA treatment attenuated ISO-induced ERS and cardiac hypertrophy by activating the AMPK/SIRT1 pathway via modulation of S1pr1.
Collapse
Affiliation(s)
- Ping Ping
- General Station for Drug and Instrument Supervision and ControlJoint Logistic Support Force of Chinese People's Liberation ArmyBeijingChina
| | - Ting Yang
- Central LaboratoryHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Chaoxue Ning
- Central LaboratoryHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Qingkai Zhao
- Department of Health and MedicineHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Yali Zhao
- Central LaboratoryHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Tao Yang
- Department of OncologyHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Zhitao Gao
- School of Laboratory MedicineXinxiang Medical UniversityXinxiangChina
| | - Shihui Fu
- Department of CardiologyHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
- Department of Geriatric CardiologyChinese People's Liberation Army General HospitalBeijingChina
| |
Collapse
|
2
|
Stastna N, Hrabovska L, Homolka P, Homola L, Svoboda M, Brat K, Fila L. The long-term effect of elexacaftor/tezacaftor/ivacaftor on cardiorespiratory fitness in adolescent patients with cystic fibrosis: a pilot observational study. BMC Pulm Med 2024; 24:260. [PMID: 38807122 PMCID: PMC11134686 DOI: 10.1186/s12890-024-03069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Physical activity is a crucial demand on cystic fibrosis treatment management. The highest value of oxygen uptake (VO2peak) is an appropriate tool to evaluate the physical activity in these patients. However, there are several other valuable CPET parameters describing exercise tolerance (Wpeak, VO2VT1, VO2VT2, VO2/HRpeak, etc.), and helping to better understand the effect of specific treatment (VE, VT, VD/VT etc.). Limited data showed ambiguous results of this improvement after CFTR modulator treatment. Elexacaftor/tezacaftor/ivacaftor medication improves pulmonary function and quality of life, whereas its effect on CPET has yet to be sufficiently demonstrated. METHODS We performed a single group prospective observational study of 10 adolescent patients with cystic fibrosis who completed two CPET measurements between January 2019 and February 2023. During this period, elexacaftor/tezacaftor/ivacaftor treatment was initiated in all of them. The first CPET at the baseline was followed by controlled CPET at least one year after medication commencement. We focused on interpreting the data on their influence by the novel therapy. We hypothesized improvements in cardiorespiratory fitness following treatment. We applied the Wilcoxon signed-rank test. The data were adjusted for age at the time of CPET to eliminate bias of aging in adolescent patients. RESULTS We observed significant improvement in peak workload, VO2 peak, VO2VT1, VO2VT2, VE/VCO2 slope, VE, VT, RQ, VO2/HR peak and RR peak. The mean change in VO2 peak was 5.7 mL/kg/min, or 15.9% of the reference value (SD ± 16.6; p= 0.014). VO2VT1 improved by 15% of the reference value (SD ± 0.1; p= 0.014), VO2VT2 improved by 0.5 (SD ± 0.4; p= 0.01). There were no differences in other parameters. CONCLUSION Exercise tolerance improved after elexacaftor/tezacaftor/ivacaftor treatment initiation. We suggest that the CFTR modulator alone is not enough for recovering physical decondition, but should be supplemented with physical activity and respiratory physiotherapy. Further studies are needed to examine the effect of CFTR modulators and physical therapy on cardiopulmonary exercise tolerance.
Collapse
Affiliation(s)
- Nela Stastna
- Department of Pulmonology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.
- Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Lenka Hrabovska
- Department of Sports Medicine and Rehabilitation, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Homolka
- Department of Sports Medicine and Rehabilitation, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lukas Homola
- Department of Paediatric Infectious Diseases, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michal Svoboda
- Institute of Biostatistics and Analyses Ltd. and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kristian Brat
- Department of Pulmonology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Libor Fila
- Department of Pulmonology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
3
|
Gaastra B, Zhang J, Tapper W, Bulters D, Galea I. Sphingosine-1-phosphate Signalling in Aneurysmal Subarachnoid Haemorrhage: Basic Science to Clinical Translation. Transl Stroke Res 2024; 15:352-363. [PMID: 36749550 PMCID: PMC10891271 DOI: 10.1007/s12975-023-01133-9] [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: 01/01/2023] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/08/2023]
Abstract
Sphingosine-1-phosphate (S1P) is generated intracellularly and, when transported to the extracellular compartment, predominantly signals through S1P receptors. The S1P signalling pathway has been implicated in the pathophysiology of neurological injury following aneurysmal subarachnoid haemorrhage (aSAH). In this review, we bring together all the available data regarding the role of S1P in neurological injury following aSAH. There is agreement in the literature that S1P increases in the cerebrospinal fluid following aSAH and leads to cerebral artery vasospasm. On the other hand, the role of S1P in the parenchyma is less clear cut, with different studies arguing for beneficial and deleterious effects. A parsimonious interpretation of this apparently conflicting data is presented. We discuss the potential of S1P receptor modulators, in clinical use for multiple sclerosis, to be repurposed for aSAH. Finally, we highlight the gaps in our knowledge of S1P signalling in humans, the clinical challenges of targeting the S1P pathway after aSAH and other research priorities.
Collapse
Affiliation(s)
- Ben Gaastra
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK.
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton, Southampton, SO16 6YD, UK.
| | - John Zhang
- Center of Neuroscience Research, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Will Tapper
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| | - Diederik Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton, Southampton, SO16 6YD, UK
| | - Ian Galea
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
4
|
Kaushik S, Bhargava P, Sharma J, Arava S, Nag TC, Arya DS, Bhatia J. Sesamol attenuates bleomycin-induced pulmonary toxicity and fibrosis in experimental animals. J Biochem Mol Toxicol 2023; 37:e23472. [PMID: 37462223 DOI: 10.1002/jbt.23472] [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: 01/08/2022] [Revised: 06/29/2023] [Accepted: 07/08/2023] [Indexed: 11/10/2023]
Abstract
Sesamol, a lignan obtained from roasted seeds of Sesamum indicum, has high antioxidant and anti-inflammatory activity. In this study, we have investigated the effect of sesamol on Bleomycin (BLM) induced pulmonary toxicity as well as fibrosis in Wistar rats. Lung toxicity was induced by administration of BLM, 0.015 U/g ip, twice weekly for 28 days whereas lung fibrosis was induced by BLM, 0.015 U/g ip, every 5th day for 49 days. Sesamol administration was started 7 days before first dose of BLM in both the models. It was observed that sesamol 50 mg/kg most effectively attenuated pulmonary toxicity by reducing oxidative stress, inflammation and apoptosis. This dose was further evaluated for its anti-fibrotic effect. It was observed that there was a significant reduction in fibrosis. Lung collagen content was markedly reduced. Furthermore, expression of pro-fibrotic proteins, TGF-β/SMAD and α-SMA, was reduced and that of anti-fibrotic protein, AMPK, was markedly increased. Even though the combination of sesamol with pirfenidone exhibited no additional protection than either drug alone, it is evident from our study that our test drug, sesamol is comparable in efficacy to pirfenidone. Thus, sesamol has promising therapeutic potential in treatment of pulmonary toxicity and fibrosis.
Collapse
Affiliation(s)
- Swati Kaushik
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Poorva Bhargava
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jatin Sharma
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sudheer Arava
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas C Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir S Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
5
|
Wu Y, Chen L, Zhong F, Zhou K, Lu C, Cheng X, Wang S. Cognitive impairment in patients with heart failure: molecular mechanism and therapy. Heart Fail Rev 2023:10.1007/s10741-022-10289-9. [PMID: 36593370 DOI: 10.1007/s10741-022-10289-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 01/04/2023]
Abstract
Heart failure (HF) is associated with multiple organ dysfunction and many comorbidities. Its incidence is high among the elderly and is a major health burden worldwide. Cognitive impairment (CI) is highly prevalent in older patients with HF, which is an abnormality in one or more of the items of cognition, attention, memory, language, psychomotor function, and visual spatial acuity. Studies have shown that the incidence of CI in HF patients is between 13 and 54%, and patients with both conditions have poor self-care ability and prognosis, as well as increased mortality rates. However, the mechanisms of CI development in HF patients are still unclear. In this review, we describe the epidemiology and risk factors as well as measures of improving CI in HF patients. We update the latest pathophysiological mechanisms related to the neurocognitive changes in HF patients, expounding on the mechanisms associated with the development of CI in HF patients.
Collapse
Affiliation(s)
- Yanan Wu
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Liwen Chen
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Feng Zhong
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Kaiyi Zhou
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Chao Lu
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xiao Cheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sheng Wang
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China.
| |
Collapse
|
6
|
Uhl FE, Vanherle L, Meissner A. Cystic fibrosis transmembrane regulator correction attenuates heart failure-induced lung inflammation. Front Immunol 2022; 13:928300. [PMID: 35967318 PMCID: PMC9365932 DOI: 10.3389/fimmu.2022.928300] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Heart failure (HF) affects 64 million people worldwide. Despite advancements in prevention and therapy, quality of life remains poor for many HF patients due to associated target organ damage. Pulmonary manifestations of HF are well-established. However, difficulties in the treatment of HF patients with chronic lung phenotypes remain as the underlying patho-mechanistic links are still incompletely understood. Here, we aim to investigate the cystic fibrosis transmembrane regulator (CFTR) involvement in lung inflammation during HF, a concept that may provide new mechanism-based therapies for HF patients with pulmonary complications. In a mouse model of HF, pharmacological CFTR corrector therapy (Lumacaftor (Lum)) was applied systemically or lung-specifically for 2 weeks, and the lungs were analyzed using histology, flow cytometry, western blotting, and qPCR. Experimental HF associated with an apparent lung phenotype characterized by vascular inflammation and remodeling, pronounced tissue inflammation as evidenced by infiltration of pro-inflammatory monocytes, and a reduction of pulmonary CFTR+ cells. Moreover, the elevation of a classically-activated phenotype of non-alveolar macrophages coincided with a cell-specific reduction of CFTR expression. Pharmacological correction of CFTR with Lum mitigated the HF-induced downregulation of pulmonary CFTR expression and increased the proportion of CFTR+ cells in the lung. Lum treatment diminished the HF-associated elevation of classically-activated non-alveolar macrophages, while promoting an alternatively-activated macrophage phenotype within the lungs. Collectively, our data suggest that downregulation of CFTR in the HF lung extends to non-alveolar macrophages with consequences for tissue inflammation and vascular structure. Pharmacological CFTR correction possesses the capacity to alleviate HF-associated lung inflammation.
Collapse
Affiliation(s)
- Franziska E. Uhl
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Lotte Vanherle
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Anja Meissner
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
- *Correspondence: Anja Meissner,
| |
Collapse
|
7
|
Bernacchioni C, Squecco R, Gamberi T, Ghini V, Schumacher F, Mannelli M, Garella R, Idrizaj E, Cencetti F, Puliti E, Bruni P, Turano P, Fiaschi T, Donati C. S1P Signalling Axis Is Necessary for Adiponectin-Directed Regulation of Electrophysiological Properties and Oxidative Metabolism in C2C12 Myotubes. Cells 2022; 11:713. [PMID: 35203362 PMCID: PMC8869893 DOI: 10.3390/cells11040713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Adiponectin (Adn), released by adipocytes and other cell types such as skeletal muscle, has insulin-sensitizing and anti-inflammatory properties. Sphingosine 1-phosphate (S1P) is reported to act as effector of diverse biological actions of Adn in different tissues. S1P is a bioactive sphingolipid synthesized by the phosphorylation of sphingosine catalyzed by sphingosine kinase (SK) 1 and 2. Consolidated findings support the key role of S1P in the biology of skeletal muscle. METHODS AND RESULTS Here we provide experimental evidence that S1P signalling is modulated by globular Adn treatment being able to increase the phosphorylation of SK1/2 as well as the mRNA expression levels of S1P4 in C2C12 myotubes. These findings were confirmed by LC-MS/MS that showed an increase of S1P levels after Adn treatment. Notably, the involvement of S1P axis in Adn action was highlighted since, when SK1 and 2 were inhibited by PF543 and ABC294640 inhibitors, respectively, not only the electrophysiological changes but also the increase of oxygen consumption and of aminoacid levels induced by the hormone, were significantly inhibited. CONCLUSION Altogether, these findings show that S1P biosynthesis is necessary for the electrophysiological properties and oxidative metabolism of Adn in skeletal muscle cells.
Collapse
Affiliation(s)
- Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Roberta Squecco
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Veronica Ghini
- Magnetic Resonance Center (CERM), University of Florence, 50019 Florence, Italy; (V.G.); (P.T.)
| | - Fabian Schumacher
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany;
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Rachele Garella
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Eglantina Idrizaj
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Elisa Puliti
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Paola Turano
- Magnetic Resonance Center (CERM), University of Florence, 50019 Florence, Italy; (V.G.); (P.T.)
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| |
Collapse
|
8
|
Westhölter D, Schumacher F, Wülfinghoff N, Sutharsan S, Strassburg S, Kleuser B, Horn PA, Reuter S, Gulbins E, Taube C, Welsner M. CFTR modulator therapy alters plasma sphingolipid profiles in people with cystic fibrosis. J Cyst Fibros 2022; 21:713-720. [DOI: 10.1016/j.jcf.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/27/2022] [Accepted: 02/06/2022] [Indexed: 12/17/2022]
|
9
|
Uhl FE, Vanherle L, Matthes F, Meissner A. Therapeutic CFTR Correction Normalizes Systemic and Lung-Specific S1P Level Alterations Associated with Heart Failure. Int J Mol Sci 2022; 23:866. [PMID: 35055052 PMCID: PMC8777932 DOI: 10.3390/ijms23020866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
Heart failure (HF) is among the main causes of death worldwide. Alterations of sphingosine-1-phosphate (S1P) signaling have been linked to HF as well as to target organ damage that is often associated with HF. S1P's availability is controlled by the cystic fibrosis transmembrane regulator (CFTR), which acts as a critical bottleneck for intracellular S1P degradation. HF induces CFTR downregulation in cells, tissues and organs, including the lung. Whether CFTR alterations during HF also affect systemic and tissue-specific S1P concentrations has not been investigated. Here, we set out to study the relationship between S1P and CFTR expression in the HF lung. Mice with HF, induced by myocardial infarction, were treated with the CFTR corrector compound C18 starting ten weeks post-myocardial infarction for two consecutive weeks. CFTR expression, S1P concentrations, and immune cell frequencies were determined in vehicle- and C18-treated HF mice and sham controls using Western blotting, flow cytometry, mass spectrometry, and qPCR. HF led to decreased pulmonary CFTR expression, which was accompanied by elevated S1P concentrations and a pro-inflammatory state in the lungs. Systemically, HF associated with higher S1P plasma levels compared to sham-operated controls and presented with higher S1P receptor 1-positive immune cells in the spleen. CFTR correction with C18 attenuated the HF-associated alterations in pulmonary CFTR expression and, hence, led to lower pulmonary S1P levels, which was accompanied by reduced lung inflammation. Collectively, these data suggest an important role for the CFTR-S1P axis in HF-mediated systemic and pulmonary inflammation.
Collapse
Affiliation(s)
- Franziska E. Uhl
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (F.E.U.); (L.V.); (F.M.)
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Lotte Vanherle
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (F.E.U.); (L.V.); (F.M.)
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Frank Matthes
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (F.E.U.); (L.V.); (F.M.)
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Anja Meissner
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (F.E.U.); (L.V.); (F.M.)
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
10
|
Caterini JE, Ratjen F, Barker AR, Williams CA, Rendall K, Schneiderman JE, Wells GD. Exercise intolerance in cystic fibrosis-the role of CFTR modulator therapies. J Cyst Fibros 2021; 21:282-292. [PMID: 34955387 DOI: 10.1016/j.jcf.2021.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/18/2021] [Accepted: 11/20/2021] [Indexed: 12/11/2022]
Abstract
Exercise intolerance is common in people with CF (pwCF), but not universal among all individuals. While associated with disease prognosis, exercise intolerance is not simply a reflection of the degree of lung disease. In people with severe CF, respiratory limitations may contribute more significantly to impaired exercise capacity than in those with mild-moderate CF. At all levels of disease severity, there are peripheral factors e.g., abnormal macro- and micro-vascular function that impair blood flow and reduce oxygen extraction, and mitochondrial defects that diminish metabolic efficiency. We discuss advances in understanding the central and peripheral mechanisms underlying exercise intolerance in pwCF. Exploring both the central and peripheral factors that contribute to exercise intolerance in CF can help inform the development of new therapeutic targets, as well as help define prognostic criteria.
Collapse
Affiliation(s)
- Jessica E Caterini
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada; Queen's Medical School, Kingston, ON K7L 3N6, Canada
| | - Felix Ratjen
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada; Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; Division of Respiratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Alan R Barker
- Children's Health and Exercise Research Centre, Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, UK
| | - Craig A Williams
- Children's Health and Exercise Research Centre, Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, UK
| | - Kate Rendall
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada
| | - Jane E Schneiderman
- Division of Respiratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Greg D Wells
- Translational Medicine Program, SickKids Research Institute, Toronto, ON M5G 0A4, Canada.
| |
Collapse
|
11
|
Zulueta A, Dei Cas M, Luciano F, Mingione A, Pivari F, Righi I, Morlacchi L, Rosso L, Signorelli P, Ghidoni R, Paroni R, Caretti A. Spns2 Transporter Contributes to the Accumulation of S1P in Cystic Fibrosis Human Bronchial Epithelial Cells. Biomedicines 2021; 9:biomedicines9091121. [PMID: 34572307 PMCID: PMC8467635 DOI: 10.3390/biomedicines9091121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/03/2022] Open
Abstract
The role of S1P in Cystic Fibrosis (CF) has been investigated since 2001, when it was first described that the CFTR channel regulates the inward transport of S1P. From then on, various studies have associated F508del CFTR, the most frequent mutation in CF patients, with altered S1P expression in tissue and plasma. We found that human bronchial epithelial immortalized and primary cells from CF patients express more S1P than the control cells, as evidenced by mass spectrometry analysis. S1P accumulation relies on two- to four-fold transcriptional up-regulation of SphK1 and simultaneous halving of SGPL1 in CF vs. control cells. The reduction of SGPL1 transcription protects S1P from irreversible degradation, but the excessive accumulation is partially prevented by the action of the two phosphatases that are up-regulated compared to control cells. For the first time in CF, we describe that Spns2, a non-ATP dependent transporter that normally extrudes S1P out of the cells, shows deficient transcriptional and protein expression, thus impairing S1P accrual dissipation. The in vitro data on CF human bronchial epithelia correlates with the impaired expression of Spns2 observed in CF human lung biopsies compared to healthy control.
Collapse
Affiliation(s)
- Aida Zulueta
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (A.Z.); (F.L.); (A.M.); (F.P.); (P.S.); (R.G.)
| | - Michele Dei Cas
- Clinical Biochemistry and Mass Spectrometry Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.D.C.); (R.P.)
| | - Francesco Luciano
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (A.Z.); (F.L.); (A.M.); (F.P.); (P.S.); (R.G.)
| | - Alessandra Mingione
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (A.Z.); (F.L.); (A.M.); (F.P.); (P.S.); (R.G.)
| | - Francesca Pivari
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (A.Z.); (F.L.); (A.M.); (F.P.); (P.S.); (R.G.)
| | - Ilaria Righi
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (I.R.); (L.R.)
| | - Letizia Morlacchi
- Respiratory Unit and Cystic Fibrosis Center, Internal Medicine Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (I.R.); (L.R.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Paola Signorelli
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (A.Z.); (F.L.); (A.M.); (F.P.); (P.S.); (R.G.)
| | - Riccardo Ghidoni
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (A.Z.); (F.L.); (A.M.); (F.P.); (P.S.); (R.G.)
| | - Rita Paroni
- Clinical Biochemistry and Mass Spectrometry Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (M.D.C.); (R.P.)
| | - Anna Caretti
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, 20142 Milan, Italy; (A.Z.); (F.L.); (A.M.); (F.P.); (P.S.); (R.G.)
- Correspondence: ; Tel.: +39-02-50323264
| |
Collapse
|
12
|
Abstract
The limb-girdle muscular dystrophies (LGMD) are a collection of genetic diseases united in their phenotypical expression of pelvic and shoulder area weakness and wasting. More than 30 subtypes have been identified, five dominant and 26 recessive. The increase in the characterization of new genotypes in the family of LGMDs further adds to the heterogeneity of the disease. Meanwhile, better understanding of the phenotype led to the reconsideration of the disease definition, which resulted in eight old subtypes to be no longer recognized officially as LGMD and five new diseases to be added to the LGMD family. The unique variabilities of LGMD stem from genetic mutations, which then lead to protein and ultimately muscle dysfunction. Herein, we review the LGMD pathway, starting with the genetic mutations that encode proteins involved in muscle maintenance and repair, and including the genotype–phenotype relationship of the disease, the epidemiology, disease progression, burden of illness, and emerging treatments.
Collapse
|
13
|
Donati C, Cencetti F, Bernacchioni C, Vannuzzi V, Bruni P. Role of sphingosine 1-phosphate signalling in tissue fibrosis. Cell Signal 2020; 78:109861. [PMID: 33253915 DOI: 10.1016/j.cellsig.2020.109861] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Fibrosis is characterized by the excessive accumulation of extracellular matrix components, leading to loss of tissue function in affected organs. Although the majority of fibrotic diseases have different origins, they have in common a persistent inflammatory stimulus and lymphocyte-monocyte interactions that determine the production of numerous fibrogenic cytokines. Treatment to contrast fibrosis is urgently needed, since some fibrotic diseases lead to systemic fibrosis and represent a major cause of death. In this article, the role of the bioactive sphingolipid sphingosine 1-phosphate (S1P) and its signalling pathway in the fibrosis of different tissue contexts is extensively reviewed, highlighting that it may represent an innovative and promising pharmacological therapeutic target for treating this devastating multifaceted disease. In multiple tissues S1P influences different aspects of fibrosis modulating the recruitment of inflammatory cells, as well as cell proliferation, migration and transdifferentiation into myofibroblasts, the cell type mainly involved in fibrosis development. Moreover, at the level of fibrotic lesions, S1P metabolism is profoundly influenced by multiple cross-talk with profibrotic mediators, such as transforming growth factor β, thus finely regulating the development of fibrosis. This article is part of a Special Issue entitled "Physiological and pathological roles of bioactive sphingolipids".
Collapse
Affiliation(s)
- Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy.
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| | - Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| | - Valentina Vannuzzi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
14
|
Jourdain ML, Sergheraert J, Braux J, Guillaume C, Gangloff SC, Hubert D, Velard F, Jacquot J. Osteoclastogenesis and sphingosine-1-phosphate secretion from human osteoclast precursor monocytes are modulated by the cystic fibrosis transmembrane conductance regulator. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166010. [PMID: 33188942 DOI: 10.1016/j.bbadis.2020.166010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/09/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
Osteopenia and increased fracture rates are well-recognized in patients with cystic fibrosis (CF) disease. In CF pathology, F508del is the most common CFTR mutation, with more than 85% of patients carrying it on at least one allele. The underlying molecular defect in CFTR caused by the F508del-CFTR mutation in osteoclastogenesis, i.e., on the generation and bone-resorption activity of osteoclasts (OCs) from peripheral blood-derived monocytes (PBMCs) remained unexplored. We therefore investigated whether the F508del mutation could affect the osteoclastogenic capacity of PBMCs collected from 15 adult patients bearing the F508del-CFTR mutation, to modulate their bone-resorptive abilities and the level of sphingosine-1-phosphate (S1P) produced by OCs, a key factor in the bone mineral density and formation. In the present study, a severe, defective differentiation of CF-F508del PBMCs to CF-F508del OCs without any significant difference in nuclei number per OC was found compared to non-CF healthy PBMCs from 13 subjects after 7-14-days culture periods. We observed a reduced number of formed non-CF healthy OCs in the presence of a selective inhibitor of CFTR chloride conductance (CFTR-Inh172). Our data regarding OCs resorptive capabilites revealed that a loss of CFTR chloride activity in OCs led to a marked reduction in their trench-resorption mode. A 7-fold increase of the S1P release by CF-F508del OCs was found compared to non-CF healthy OCs after a 21-days culture period. We hypothesize that defective maturation of F508del-OCs precursor monocytes associated with high S1P production in the bone environment might contribute to low bone mineral density observed in the CF population.
Collapse
Affiliation(s)
- Marie-Laure Jourdain
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France; CHU de Reims, Pôle de Médecine Bucco-Dentaire, 51100 Reims, France
| | - Johan Sergheraert
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France; CHU de Reims, Pôle de Médecine Bucco-Dentaire, 51100 Reims, France
| | - Julien Braux
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France; CHU de Reims, Pôle de Médecine Bucco-Dentaire, 51100 Reims, France
| | - Christine Guillaume
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France
| | - Sophie C Gangloff
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France
| | - Dominique Hubert
- Service de Pneumologie, Adult Cystic Fibrosis Centre, Hôpital Cochin, 75012 Paris, France
| | - Frédéric Velard
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France.
| | - Jacky Jacquot
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France.
| |
Collapse
|
15
|
Liessi N, Pesce E, Braccia C, Bertozzi SM, Giraudo A, Bandiera T, Pedemonte N, Armirotti A. Distinctive lipid signatures of bronchial epithelial cells associated with cystic fibrosis drugs, including Trikafta. JCI Insight 2020; 5:138722. [PMID: 32673287 PMCID: PMC7455125 DOI: 10.1172/jci.insight.138722] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
In recent years, a number of drugs have been approved for the treatment of cystic fibrosis (CF). Among them, newly released Trikafta, a combination of 3 drugs (VX-661/VX-445/VX-770), holds great promise to radically improve the quality of life for a large portion of patients with CF carrying 1 copy of F508del, the most frequent CF transmembrane conductance regulator (CFTR) mutation. Currently available disease-modifying CF drugs work by rescuing the function of the mutated CFTR anion channel. Recent research has shown that membrane lipids, and the cell lipidome in general, play a significant role in the mechanism of CFTR-defective trafficking and, on the other hand, its rescue. In this paper, by using untargeted lipidomics on CFBE41o- cells, we identified distinctive changes in the bronchial epithelial cell lipidome associated with treatment with Trikafta and other CF drugs. Particularly interesting was the reduction of levels of ceramide, a known molecular player in the induction of apoptosis, which appeared to be associated with a decrease in the susceptibility of cells to undergo apoptosis. This evidence could account for additional beneficial roles of the triple combination of drugs on CF phenotypes.
Collapse
Affiliation(s)
- Nara Liessi
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Genova, Italy
| | - Emanuela Pesce
- L'Unità Operativa Complessa (UOC) Genetica Medica, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Giannina Gaslini, Genova, Italy
| | - Clarissa Braccia
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Genova, Italy
| | | | | | - Tiziano Bandiera
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Genova, Italy
| | - Nicoletta Pedemonte
- L'Unità Operativa Complessa (UOC) Genetica Medica, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Giannina Gaslini, Genova, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
16
|
Liessi N, Pedemonte N, Armirotti A, Braccia C. Proteomics and Metabolomics for Cystic Fibrosis Research. Int J Mol Sci 2020; 21:ijms21155439. [PMID: 32751630 PMCID: PMC7432297 DOI: 10.3390/ijms21155439] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/18/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022] Open
Abstract
The aim of this review article is to introduce the reader to the state-of-the-art of the contribution that proteomics and metabolomics sciences are currently providing for cystic fibrosis (CF) research: from the understanding of cystic fibrosis transmembrane conductance regulator (CFTR) biology to biomarker discovery for CF diagnosis. Our work particularly focuses on CFTR post-translational modifications and their role in cellular trafficking as well as on studies that allowed the identification of CFTR molecular interactors. We also show how metabolomics is currently helping biomarker discovery in CF. The most recent advances in these fields are covered by this review, as well as some considerations on possible future scenarios for new applications.
Collapse
Affiliation(s)
- Nara Liessi
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy;
| | - Nicoletta Pedemonte
- U.O.C. Genetica Medica, IRCCS Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Andrea Armirotti
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy;
- Correspondence: ; Tel.: +39-010-2896-938
| | - Clarissa Braccia
- D3PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy;
| |
Collapse
|
17
|
Plasma Levels of the Bioactive Sphingolipid Metabolite S1P in Adult Cystic Fibrosis Patients: Potential Target for Immunonutrition? Nutrients 2020; 12:nu12030765. [PMID: 32183316 PMCID: PMC7146441 DOI: 10.3390/nu12030765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 01/03/2023] Open
Abstract
Recent research has linked sphingolipid (SL) metabolism with cystic fibrosis transmembrane conductance regulator (CFTR) activity, affecting bioactive lipid mediator sphingosine-1-phosphate (S1P). We hypothesize that loss of CFTR function in cystic fibrosis (CF) patients influenced plasma S1P levels. Total and unbound plasma S1P levels were measured in 20 lung-transplanted adult CF patients and 20 healthy controls by mass spectrometry and enzyme-linked immunosorbent assay (ELISA). S1P levels were correlated with CFTR genotype, routine laboratory parameters, lung function and pathogen colonization, and clinical symptoms. Compared to controls, CF patients showed lower unbound plasma S1P, whereas total S1P levels did not differ. A positive correlation of total and unbound S1P levels was found in healthy controls, but not in CF patients. Higher unbound S1P levels were measured in ΔF508-homozygous compared to ΔF508-heterozygous CF patients (p = 0.038), accompanied by higher levels of HDL in ΔF508-heterozygous patients. Gastrointestinal symptoms were more common in ΔF508 heterozygotes compared to ΔF508 homozygotes. This is the first clinical study linking plasma S1P levels with CFTR function and clinical presentation in adult CF patients. Given the emerging role of immunonutrition in CF, our study might pave the way for using S1P as a novel biomarker and nutritional target in CF.
Collapse
|
18
|
Druggable Sphingolipid Pathways: Experimental Models and Clinical Opportunities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:101-135. [PMID: 32894509 DOI: 10.1007/978-3-030-50621-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intensive research in the field of sphingolipids has revealed diverse roles in cell biological responses and human health and disease. This immense molecular family is primarily represented by the bioactive molecules ceramide, sphingosine, and sphingosine 1-phosphate (S1P). The flux of sphingolipid metabolism at both the subcellular and extracellular levels provides multiple opportunities for pharmacological intervention. The caveat is that perturbation of any single node of this highly regulated flux may have effects that propagate throughout the metabolic network in a dramatic and sometimes unexpected manner. Beginning with S1P, the receptors for which have thus far been the most clinically tractable pharmacological targets, this review will describe recent advances in therapeutic modulators targeting sphingolipids, their chaperones, transporters, and metabolic enzymes.
Collapse
|
19
|
Lidington D, Fares JC, Uhl FE, Dinh DD, Kroetsch JT, Sauvé M, Malik FA, Matthes F, Vanherle L, Adel A, Momen A, Zhang H, Aschar-Sobbi R, Foltz WD, Wan H, Sumiyoshi M, Macdonald RL, Husain M, Backx PH, Heximer SP, Meissner A, Bolz SS. CFTR Therapeutics Normalize Cerebral Perfusion Deficits in Mouse Models of Heart Failure and Subarachnoid Hemorrhage. JACC Basic Transl Sci 2019; 4:940-958. [PMID: 31909302 PMCID: PMC6939007 DOI: 10.1016/j.jacbts.2019.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 01/01/2023]
Abstract
Heart failure (HF) and subarachnoid hemorrhage (SAH) chronically reduce cerebral perfusion, which negatively affects clinical outcome. This work demonstrates a strong relationship between cerebral artery cystic fibrosis transmembrane conductance regulator (CFTR) expression and altered cerebrovascular reactivity in HF and SAH. In HF and SAH, CFTR corrector compounds (C18 or lumacaftor) normalize pathological alterations in cerebral artery CFTR expression, vascular reactivity, and cerebral perfusion, without affecting systemic hemodynamic parameters. This normalization correlates with reduced neuronal injury. Therefore, CFTR therapeutics have emerged as valuable clinical tools to manage cerebrovascular dysfunction, impaired cerebral perfusion, and neuronal injury.
Collapse
Key Words
- CBF, cerebral blood flow
- CFTR, cystic fibrosis transmembrane conductance regulator
- HF, heart failure
- MAP, mean arterial pressure
- MOPS, 3-morpholinopropanesulfonic acid
- MRI, magnetic resonance imaging
- NIH, National Institutes of Health
- PCA, posterior cerebral artery
- S1P, sphingosine-1-phosphate
- SAH, subarachnoid hemorrhage
- TNF, tumor necrosis factor
- TPR, total peripheral resistance
- cognitive impairment
- corrector compounds
- cystic fibrosis transmembrane conductance regulator (CFTR)
- myogenic vasoconstriction
- sphingosine-1-phosphate
- tumor necrosis factor
- vascular smooth muscle cells
Collapse
Affiliation(s)
- Darcy Lidington
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Jessica C. Fares
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Franziska E. Uhl
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Danny D. Dinh
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Jeffrey T. Kroetsch
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Meghan Sauvé
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Firhan A. Malik
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Frank Matthes
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lotte Vanherle
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Arman Adel
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Abdul Momen
- Division of Cell & Molecular Biology, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Hangjun Zhang
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | | | - Warren D. Foltz
- STTARR Innovation Centre, Department of Radiation Oncology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Hoyee Wan
- Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Research Centre for Biomedical Research and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
- Division of Neurosurgery, St. Michael’s Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Sunnybrook Research Institute, Physical Sciences Platform and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Manabu Sumiyoshi
- Division of Neurosurgery, St. Michael’s Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Biosciences, Department of Neurosurgery, University of Tokushima Graduate School, Tokushima, Japan
| | - R. Loch Macdonald
- Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Research Centre for Biomedical Research and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
- Division of Neurosurgery, St. Michael’s Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Mansoor Husain
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Division of Cell & Molecular Biology, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter H. Backx
- Division of Cardiology, University Health Network, Toronto, Ontario, Canada
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Scott P. Heximer
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
| | - Anja Meissner
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Steffen-Sebastian Bolz
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Li RS, Xu GH, Cao J, Liu B, Xie HF, Ishii Y, Zhang CF. Alpha-Mangostin Ameliorates Bleomycin-Induced Pulmonary Fibrosis in Mice Partly Through Activating Adenosine 5'-Monophosphate-Activated Protein Kinase. Front Pharmacol 2019; 10:1305. [PMID: 31798444 PMCID: PMC6863977 DOI: 10.3389/fphar.2019.01305] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Pulmonary fibrosis (PF) is a devastating interstitial lung disease and characterized by an abnormal accumulation of extracellular matrix (ECM). Nintedanib (NDN) and pirfenidone are two approved therapies for PF, but their potential side-effects have been reported. Recently, the use of natural supplements for PF is attracting attention. Alpha-mangostin (α-MG) is an active xanthone-type compound isolated from the nutritious fruit mangosteen. Purpose: In the present study, the potential effect and underlying mechanism of α-MG were evaluated in bleomycin (BLM)-induced PF and activated primary lung fibroblasts (PLFs). Methods: Histopathological changes and collagen deposition were analyzed via hematoxylin-eosin staining and Masson staining, the expression of nicotinamide adenine dinucleotide phosphate oxidase-4 (NOX4) involved in oxidative stress in lung tissues was analyzed by immunochemistry staining. The expressions of α-smooth muscle actin (α-SMA), collagen I (Col I), p-adenosine 5′-monophosphate-activated protein kinase (AMPK)/AMPK, and NOX4 were detected by Western blot, immunofluorescence or RT-PCR, and effects of α-MG on cell viability were detected using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide. Results:In vivo results demonstrated that α-MG treatment (10 mg/kg/day) significantly ameliorated BLM-induced deposition of ECM in lung tissues. Moreover, α-MG could inhibit protein expressions of α-SMA and Col I as well as its mRNA levels. In addition, α-MG also significantly inhibited transforming growth factor-β1/Smad2/3 pathway and regulated the protein expression of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in lung tissues. In vitro results demonstrated that α-MG significantly increased p-AMPK/AMPK but reduced the protein expression level of α-SMA and Col I as well as NOX4 in activated PLFs. Further study demonstrated that these improvement effects were significantly blocked by compound C. Conclusion: α-MG treatment significantly decreased oxidative stress in lungs partly by activating AMPK mediated signaling pathway in BLM-induced PF and activated PLFs and decreased the deposition of ECM. The present study provides pharmacological evidence to support therapeutic application of α-MG in the treatment of PF.
Collapse
Affiliation(s)
- Ren-Shi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Gong-Hao Xu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Juan Cao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Bei Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Hai-Feng Xie
- Research and Development Department, Chengdu Biopurify Phytochemicals Ltd., Chengdu, China
| | - Yuji Ishii
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Chao-Feng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
21
|
Dennhardt S, Finke KR, Huwiler A, Coldewey SM. Sphingosine-1-phosphate promotes barrier-stabilizing effects in human microvascular endothelial cells via AMPK-dependent mechanisms. Biochim Biophys Acta Mol Basis Dis 2019; 1865:774-781. [PMID: 30660683 DOI: 10.1016/j.bbadis.2018.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023]
Abstract
Breakdown of the endothelial barrier is a critical step in the development of organ failure in severe inflammatory conditions such as sepsis. Endothelial cells from different tissues show phenotypic variations which are often neglected in endothelial research. Sphingosine-1-phosphate (S1P) and AMP-dependent kinase (AMPK) have been shown to protect the endothelium and phosphorylation of AMPK by S1P was shown in several cell types. However, the role of the S1P-AMPK interrelationship for endothelial barrier stabilization has not been investigated. To assess the role of the S1P-AMPK signalling axis in this context, we established an in vitro model allowing real-time monitoring of endothelial barrier function in human microvascular endothelial cells (HMEC-1) and murine glomerular endothelial cells (GENCs) with the electric cell-substrate impedance sensing (ECIS™) system. Following the disruption of the cell barrier by co-administration of LPS, TNF-α, IL-1ß, IFN-γ, and IL-6, we demonstrated self-recovery of the disrupted barrier in HMEC-1, while the barrier remained compromised in GENCs. Under physiological conditions we observed a rapid phosphorylation of AMPK in HMEC-1 stimulated with S1P, but not in GENCs. Consistently, S1P enhanced the basal endothelial barrier in HMEC-1 exclusively. siRNA-mediated knockdown of AMPK in HMEC-1 led to a less pronounced barrier enhancement. Thus we present evidence for a functional role of AMPK in S1P-mediated barrier stabilization in HMEC-1 and we provide insight into cell-type specific differences of the S1P-AMPK-interrelationship, which might influence the development of interventional strategies targeting endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Sophie Dennhardt
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Septomics Research Centre, Jena University Hospital, Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Karl R Finke
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, Bern, Switzerland
| | - Sina M Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Septomics Research Centre, Jena University Hospital, Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.
| |
Collapse
|
22
|
Molinski SV, Shahani VM, Subramanian AS, MacKinnon SS, Woollard G, Laforet M, Laselva O, Morayniss LD, Bear CE, Windemuth A. Comprehensive mapping of cystic fibrosis mutations to CFTR protein identifies mutation clusters and molecular docking predicts corrector binding site. Proteins 2018; 86:833-843. [DOI: 10.1002/prot.25496] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/07/2018] [Accepted: 03/19/2018] [Indexed: 01/01/2023]
Affiliation(s)
| | | | | | | | | | | | - Onofrio Laselva
- Programme in Molecular Structure and Function; Research Institute, Hospital for Sick Children; Toronto Ontario M5G 0A4 Canada
| | | | - Christine E. Bear
- Programme in Molecular Structure and Function; Research Institute, Hospital for Sick Children; Toronto Ontario M5G 0A4 Canada
- Department of Physiology; University of Toronto; Toronto Ontario M5S 1A8 Canada
- Department of Biochemistry; University of Toronto; Toronto Ontario M5S 1A8 Canada
| | | |
Collapse
|
23
|
Chiricozzi E, Loberto N, Schiumarini D, Samarani M, Mancini G, Tamanini A, Lippi G, Dechecchi MC, Bassi R, Giussani P, Aureli M. Sphingolipids role in the regulation of inflammatory response: From leukocyte biology to bacterial infection. J Leukoc Biol 2018; 103:445-456. [PMID: 29345379 DOI: 10.1002/jlb.3mr0717-269r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 10/13/2017] [Accepted: 11/07/2017] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids (SLs) are amphiphilic molecules mainly associated with the external leaflet of eukaryotic plasma membrane, and are structural membrane components with key signaling properties. Since the beginning of the last century, a large number of papers described the involvement of these molecules in several aspects of cell physiology and pathology. Several lines of evidence support the critical role of SLs in inflammatory diseases, by acting as anti- or pro-inflammatory mediators. They are involved in control of leukocyte activation and migration, and are recognized as essential players in host response to pathogenic infection. We propose here a critical overview of current knowledge on involvement of different classes of SLs in inflammation, focusing on the role of simple and complex SLs in pathogen-mediated inflammatory response.
Collapse
Affiliation(s)
- Elena Chiricozzi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Nicoletta Loberto
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Domitilla Schiumarini
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Maura Samarani
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Giulia Mancini
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Anna Tamanini
- Laboratorio di Patologia Molecolare-Laboratorio Analisi, Dipartimento di Patologia e Diagnostica, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Giuseppe Lippi
- Sezione di Biochimica Clinica, Università degli Studi di Verona, Verona, Italy
| | - Maria Cristina Dechecchi
- Laboratorio di Patologia Molecolare-Laboratorio Analisi, Dipartimento di Patologia e Diagnostica, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Rosaria Bassi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Paola Giussani
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Massimo Aureli
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
24
|
Jiang S, Li T, Yang Z, Yi W, Di S, Sun Y, Wang D, Yang Y. AMPK orchestrates an elaborate cascade protecting tissue from fibrosis and aging. Ageing Res Rev 2017; 38:18-27. [PMID: 28709692 DOI: 10.1016/j.arr.2017.07.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 01/10/2023]
Abstract
Fibrosis is a common process characterized by excessive extracellular matrix (ECM) accumulation after inflammatory injury, which is also a crucial cause of aging. The process of fibrosis is involved in the pathogenesis of most diseases of the heart, liver, kidney, lung, and other organs/tissues. However, there are no effective therapies for this pathological alteration. Annually, fibrosis represents a huge financial burden for the USA and the world. 5'-AMP-activated protein kinase (AMPK) is a pivotal energy sensor that alleviates or delays the process of fibrogenesis. In this review, we first present basic background information on AMPK and fibrogenesis and describe the protective roles of AMPK in three fibrogenic phases. Second, we analyze the protective action of AMPK during fibrosis in myocardial, hepatic, renal, pulmonary, and other organs/tissues. Third, we present a comprehensive discussion of AMPK during fibrosis and draw a conclusion. This review highlights recent advances, vital for basic research and clinical drug design, in the regulation of AMPK during fibrosis.
Collapse
Affiliation(s)
- Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
25
|
Wang Y, Aoki H, Yang J, Peng K, Liu R, Li X, Qiang X, Sun L, Gurley EC, Lai G, Zhang L, Liang G, Nagahashi M, Takabe K, Pandak WM, Hylemon PB, Zhou H. The role of sphingosine 1-phosphate receptor 2 in bile-acid-induced cholangiocyte proliferation and cholestasis-induced liver injury in mice. Hepatology 2017; 65:2005-2018. [PMID: 28120434 PMCID: PMC5444993 DOI: 10.1002/hep.29076] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/14/2016] [Accepted: 01/19/2017] [Indexed: 12/13/2022]
Abstract
UNLABELLED Bile duct obstruction is a potent stimulus for cholangiocyte proliferation, especially for large cholangiocytes. Our previous studies reported that conjugated bile acids (CBAs) activate the protein kinase B (AKT) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling pathways through sphingosine 1-phosphate receptor (S1PR) 2 in hepatocytes and cholangiocarcinoma cells. It also has been reported that taurocholate (TCA) promotes large cholangiocyte proliferation and protects cholangiocytes from bile duct ligation (BDL)-induced apoptosis. However, the role of S1PR2 in bile-acid-mediated cholangiocyte proliferation and cholestatic liver injury has not been elucidated. Here, we report that S1PR2 is the predominant S1PR expressed in cholangiocytes. Both TCA- and sphingosine-1-phosphate (S1P)-induced activation of ERK1/2 and AKT were inhibited by JTE-013, a specific antagonist of S1PR2, in cholangiocytes. In addition, TCA- and S1P-induced cell proliferation and migration were inhibited by JTE-013 and a specific short hairpin RNA of S1PR2, as well as chemical inhibitors of ERK1/2 and AKT in mouse cholangiocytes. In BDL mice, expression of S1PR2 was up-regulated in whole liver and cholangiocytes. S1PR2 deficiency significantly reduced BDL-induced cholangiocyte proliferation and cholestatic injury, as indicated by significant reductions in inflammation and liver fibrosis in S1PR2 knockout mice. Treatment of BDL mice with JTE-013 significantly reduced total bile acid levels in serum and cholestatic liver injury. CONCLUSION This study suggests that CBA-induced activation of S1PR2-mediated signaling pathways plays a critical role in obstructive cholestasis and may represent a novel therapeutic target for cholestatic liver diseases. (Hepatology 2017;65:2005-2018).
Collapse
Affiliation(s)
- Yongqing Wang
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University,Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Hiroaki Aoki
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Jing Yang
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,China Pharmaceutical University
| | - Kesong Peng
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,College of Pharmaceutical Science, Wenzhou Medical University
| | - Runping Liu
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Xiaojiaoyang Li
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,China Pharmaceutical University
| | - Xiaoyan Qiang
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,China Pharmaceutical University
| | - Lixin Sun
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,China Pharmaceutical University
| | - Emily C Gurley
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Guanhua Lai
- Department of Pathology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, 23298
| | | | - Guang Liang
- College of Pharmaceutical Science, Wenzhou Medical University
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City 951-8510
| | - Kazuaki Takabe
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,Breast Surgery, Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - William M Pandak
- McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Phillip B. Hylemon
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298,McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298,College of Pharmaceutical Science, Wenzhou Medical University
| |
Collapse
|
26
|
Mohammed S, Harikumar KB. Sphingosine 1-Phosphate: A Novel Target for Lung Disorders. Front Immunol 2017; 8:296. [PMID: 28352271 PMCID: PMC5348531 DOI: 10.3389/fimmu.2017.00296] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/01/2017] [Indexed: 01/11/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is involved in a wide range of cellular processes, which include proliferation, apoptosis, lymphocyte egress, endothelial barrier function, angiogenesis, and inflammation. S1P is produced by two isoenzymes, namely, sphingosine kinase 1 and 2 (SphK1 and 2) and once produced, S1P can act both in an autocrine and paracrine manner. S1P can be dephosphorylated back to sphingosine by two phosphatases (SGPP 1 and 2) or can be irreversibly cleaved by S1P lyase. S1P has a diverse range of functions, which is mediated in a receptor dependent, through G-protein coupled receptors (S1PR1-5) or receptor independent manner, through intracellular targets such as HDACs and TRAF2. The involvement of S1P signaling has been confirmed in various disease conditions including lung diseases. The SphK inhibitors and S1PR modulators are currently under clinical trials for different pathophysiological conditions. There is a significant effort in targeting various components of S1P signaling for several diseases. This review focuses on the ways in which S1P signaling can be therapeutically targeted in lung disorders.
Collapse
Affiliation(s)
- Sabira Mohammed
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , India
| | - K B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , India
| |
Collapse
|
27
|
Abstract
Sphingosine-1-phosphate (S1P), a simple, bioactive sphingolipid metabolite, plays a key role, both intracellularly and extracellularly, in various cellular processes such as proliferation, survival, migration, inflammation, angiogenesis, and endothelial barrier integrity. The cellular S1P level is low and is tightly regulated by its synthesis and degradation. Sphingosine Kinases (SphKs) 1 and 2, catalyze the ATP-dependent phosphorylation of sphingosine to S1P, while the degradation is mediated by the reversible dephosphorylation catalyzed by the S1P phosphatases and lipid phosphate phosphatases and the irreversible degradation to hexadecenal and ethanolamine phosphate by sphingosine-1-phosphate lyase (S1PL). As a ligand for specific G-protein-coupled receptors, S1P1-5, which are differentially expressed in different cell types, S1P generates downstream signals that play crucial role in developmental and disease related pathologies. In addition to acting extracellularly on receptors located on the plasma membrane, S1P can also act intracellularly, independently of S1P1-5, affecting calcium homeostasis and cell proliferation. The SphKs /S1P /S1PL metabolic pathway is implicated in numerous human pathologies including respiratory disorders, thereby raising the possibility that manipulating intracellular S1P levels could offer therapeutic potential in ameliorating lung diseases. This review focuses on the prospects of targeting S1P signaling and S1P metabolizing enzymes using small molecule inhibitors, receptor agonists, and antagonists in the treatment of lung diseases.
Collapse
Affiliation(s)
- David L Ebenezer
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, IL, USA
| | - Panfeng Fu
- Department of Pharmacology, University of Illinois at Chicago, IL, USA
| | - Viswanathan Natarajan
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, IL, USA; Department of Pharmacology, University of Illinois at Chicago, IL, USA; Department of Medicine, University of Illinois at Chicago, IL, USA; Department of Bioengineering, University of Illinois at Chicago, IL, USA.
| |
Collapse
|
28
|
Aureli M, Schiumarini D, Loberto N, Bassi R, Tamanini A, Mancini G, Tironi M, Munari S, Cabrini G, Dechecchi MC, Sonnino S. Unravelling the role of sphingolipids in cystic fibrosis lung disease. Chem Phys Lipids 2016; 200:94-103. [DOI: 10.1016/j.chemphyslip.2016.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/22/2016] [Accepted: 08/25/2016] [Indexed: 12/13/2022]
|
29
|
Jacquot J, Delion M, Gangloff S, Braux J, Velard F. Bone disease in cystic fibrosis: new pathogenic insights opening novel therapies. Osteoporos Int 2016; 27:1401-1412. [PMID: 26431978 DOI: 10.1007/s00198-015-3343-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/24/2015] [Indexed: 01/17/2023]
Abstract
Mutations within the gene encoding for the chloride ion channel cystic fibrosis transmembrane conductance regulator (CFTR) results in cystic fibrosis (CF), the most common lethal autosomal recessive genetic disease that causes a number of long-term health problems, as the bone disease. Osteoporosis and increased vertebral fracture risk associated with CF disease are becoming more important as the life expectancy of patients continues to improve. The etiology of low bone density is multifactorial, most probably a combination of inadequate peak bone mass during puberty and increased bone losses in adults. Body mass index, male sex, advanced pulmonary disease, malnutrition and chronic therapies are established additional risk factors for CF-related bone disease (CFBD). Consistently, recent evidence has confirmed that CFTR plays a major role in the osteoprotegerin (OPG) and COX-2 metabolite prostaglandin E2 (PGE2) production, two key regulators in the bone formation and regeneration. Several others mechanisms were also recognized from animal and cell models contributing to malfunctions of osteoblast (cell that form bone) and indirectly of bone-resorpting osteoclasts. Understanding such mechanisms is crucial for the development of therapies in CFBD. Innovative therapeutic approaches using CFTR modulators such as C18 have recently shown in vitro capacity to enhance PGE2 production and normalized the RANKL-to-OPG ratio in human osteoblasts bearing the mutation F508del-CFTR and therefore potential clinical utility in CFBD. This review focuses on the recently identified pathogenic mechanisms leading to CFBD and potential future therapies for treating CFBD.
Collapse
Affiliation(s)
- J Jacquot
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR CAP-Santé (FED 4231), Université Reims Champagne Ardenne, 1, Avenue du Maréchal Juin, 51095, Reims, France.
| | | | | | | | | |
Collapse
|
30
|
Hui S, Levy AS, Slack DL, Burnstein MJ, Errett L, Bonneau D, Latter D, Rotstein OD, Bolz SS, Lidington D, Voigtlaender-Bolz J. Sphingosine-1-Phosphate Signaling Regulates Myogenic Responsiveness in Human Resistance Arteries. PLoS One 2015; 10:e0138142. [PMID: 26367262 PMCID: PMC4569583 DOI: 10.1371/journal.pone.0138142] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/25/2015] [Indexed: 01/06/2023] Open
Abstract
We recently identified sphingosine-1-phosphate (S1P) signaling and the cystic fibrosis transmembrane conductance regulator (CFTR) as prominent regulators of myogenic responsiveness in rodent resistance arteries. However, since rodent models frequently exhibit limitations with respect to human applicability, translation is necessary to validate the relevance of this signaling network for clinical application. We therefore investigated the significance of these regulatory elements in human mesenteric and skeletal muscle resistance arteries. Mesenteric and skeletal muscle resistance arteries were isolated from patient tissue specimens collected during colonic or cardiac bypass surgery. Pressure myography assessments confirmed endothelial integrity, as well as stable phenylephrine and myogenic responses. Both human mesenteric and skeletal muscle resistance arteries (i) express critical S1P signaling elements, (ii) constrict in response to S1P and (iii) lose myogenic responsiveness following S1P receptor antagonism (JTE013). However, while human mesenteric arteries express CFTR, human skeletal muscle resistance arteries do not express detectable levels of CFTR protein. Consequently, modulating CFTR activity enhances myogenic responsiveness only in human mesenteric resistance arteries. We conclude that human mesenteric and skeletal muscle resistance arteries are a reliable and consistent model for translational studies. We demonstrate that the core elements of an S1P-dependent signaling network translate to human mesenteric resistance arteries. Clear species and vascular bed variations are evident, reinforcing the critical need for further translational study.
Collapse
Affiliation(s)
- Sonya Hui
- Toronto Centre for Microvascular Medicine, University of Toronto and St. Michael’s Hospital, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Andrew S. Levy
- Toronto Centre for Microvascular Medicine, University of Toronto and St. Michael’s Hospital, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Daniel L. Slack
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Marcus J. Burnstein
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Surgery, St. Michael’s Hospital and University of Toronto, Toronto, Canada
| | - Lee Errett
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Surgery, St. Michael’s Hospital and University of Toronto, Toronto, Canada
| | - Daniel Bonneau
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Surgery, St. Michael’s Hospital and University of Toronto, Toronto, Canada
| | - David Latter
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Surgery, St. Michael’s Hospital and University of Toronto, Toronto, Canada
| | - Ori D. Rotstein
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Surgery, St. Michael’s Hospital and University of Toronto, Toronto, Canada
| | - Steffen-Sebastian Bolz
- Toronto Centre for Microvascular Medicine, University of Toronto and St. Michael’s Hospital, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Heart & Stroke / Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Canada
- * E-mail:
| | - Darcy Lidington
- Toronto Centre for Microvascular Medicine, University of Toronto and St. Michael’s Hospital, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Julia Voigtlaender-Bolz
- Toronto Centre for Microvascular Medicine, University of Toronto and St. Michael’s Hospital, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Department of Anaesthesia, St. Michael’s Hospital and University of Toronto, Toronto, Canada
| |
Collapse
|