1
|
Kim KS, Lee R, Park I, Hwang SH, Kim Y, Jang JW, Kim HS, Choi SM, Kim SJ, Cho HJ, Cho IH, Kim JH, Kim DG, Nah SY. Gintonin Binds to Reduced LPA4 Receptor Subtype in Human Cortical Neurons in Alzheimer's Disease Brains. Biomolecules 2025; 15:179. [PMID: 40001482 PMCID: PMC11853258 DOI: 10.3390/biom15020179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Ginseng, a traditional herbal medicine with a long history of use, is known to support human health, particularly by influencing brain function. Recent studies have identified gintonin, a lysophosphatidic acid (LPA) receptor ligand derived from ginseng, as a key bioactive. However, the specific LPA receptor subtypes targeted by gintonin in the human brain to exert its anti-Alzheimer's (AD) effects remain unclear. This study aimed to elucidate the LPA receptor subtype targeted by gintonin in the human cortex. Using a fluorescent gintonin conjugate, we investigated receptor binding in cortical samples from healthy individuals (n = 4) and AD patients (n = 4). Our results demonstrated that fluorescent gintonin selectively binds to human cortical neurons rather than glial cells and that gintonin-binding sites are co-localized with the LPA4 receptor subtype. Furthermore, the expression of LPA4 receptors was significantly reduced in the cortical neurons of AD patients. These results suggest that the LPA4 receptor may serve as a novel histopathological marker for AD and represent a promising therapeutic target for gintonin-based prevention and treatment strategies.
Collapse
Affiliation(s)
- Kyu-Sung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea;
- Neuroimmunology Laboratory, Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Rami Lee
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Inyeong Park
- Neuroimmunology Laboratory, Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju 26339, Republic of Korea;
| | - Yeshin Kim
- Department of Neurology, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea; (Y.K.), (J.-W.J.)
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University Hospital, Chuncheon 24289, Republic of Korea; (Y.K.), (J.-W.J.)
| | - Hyung-Seok Kim
- Department of Neurosurgery, Chonnam National University Medical School, Research Institute of Medical Sciences, Gwangju 61469, Republic of Korea;
| | - Seong-Min Choi
- Department of Neurology, Chonnam National University Medical School, Jebong-ro, Gwangju 61469, Republic of Korea;
| | - Sang Jin Kim
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan 47392, Republic of Korea;
- Dementia and Neurodegenerative Disease Research Center, Inje University, Busan 47392, Republic of Korea
| | - Hwa Jin Cho
- Busan & Gyeongnam Reference Laboratory, Department of Pathology, Seegene Medical Foundation, Busan 48792, Republic of Korea;
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Iksan-City 54596, Republic of Korea;
| | - Do-Geun Kim
- Neuroimmunology Laboratory, Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
2
|
Chen X, Han D, Zeng Y, Li H, Wang X, Huang Z, Yang L, Wagenaar GTM, Lin B, Yang C. Inhibition of lysophosphatidic acid receptor 2 attenuates neonatal chronic lung disease in mice by preserving vascular and alveolar development. Eur J Pharmacol 2024; 985:177120. [PMID: 39522686 DOI: 10.1016/j.ejphar.2024.177120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
AIM Bronchopulmonary dysplasia (BPD) is a common morbidity in extremely premature infants. Previous studies demonstrated the important role of lysophosphatidic acid (LPA) in inflammation in BPD. However, the role of LPA and its receptors in hyperoxia-induced vascular malformations in BPD remains to be elucidated. METHODS AND RESULTS Elevated plasma LPA levels were observed in mice with BPD compared to controls (792 vs. 607 ng/mL, p < 0.05). Inhibition of LPA signaling protected against hyperoxia-induced lung injury in neonatal mice, demonstrated by a 2.8-fold increase in pulmonary vascular density and a 14% reduction in alveolar enlargement. In vitro studies showed that LPA suppressed tube formation in human umbilical vein endothelial cells (HUVECs) by approximately 50%. LPA receptor 2 (LPA2) was identified as a functional LPA receptor in primary endothelial cells from the lungs of hyperoxic mice and in HUVECs under hyperoxic conditions. The LPA2 antagonist H2L5186303 enhanced the tube formation ability of HUVECs exposed to LPA, both under normoxia (4-fold) and hyperoxia (5-fold). Moreover, H2L5186303 significantly protected against hyperoxia-induced vascular malformation (2-fold) and improved alveolarization in neonatal mice (12% decrease in mean linear intercept, MLI). Early growth response 1 (EGR1) was characterized as a downstream target of LPA2, silencing EGR1 restored tube formation in HUVECs exposed to LPA and hyperoxia. CONCLUSIONS Our in vitro and in vivo findings demonstrate that the inhibition of LPA/LPA2 signaling mitigates hyperoxia-induced pulmonary vascular malformations, suggesting the LPA/LPA2-dependent signaling pathway has therapeutic potential for extremely premature infants with BPD.
Collapse
Affiliation(s)
- Xueyu Chen
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China; Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China
| | - Dongshan Han
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China
| | - Yali Zeng
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China
| | - Huitao Li
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China
| | - Xuan Wang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China
| | - Zilu Huang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China
| | - Lingling Yang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China
| | | | - Bingchun Lin
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China.
| | - Chuanzhong Yang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Medicine, Southern Medical University, Shenzhen, China; Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China.
| |
Collapse
|
3
|
Iwadare T, Kimura T, Yamashita Y, Okumura T, Wakabayashi SI, Kobayashi H, Sugiura A, Yamazaki T, Shimamoto S, Igarashi K, Joshita S, Umemura T. Serum Autotaxin Levels Predict Liver-Related Events in Patients With Primary Biliary Cholangitis: A Long-Term Multicenter Observational Study. Clin Transl Gastroenterol 2024; 15:e00779. [PMID: 39466702 PMCID: PMC11671064 DOI: 10.14309/ctg.0000000000000779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
INTRODUCTION A straightforward, reliable, and noninvasive method for predicting the development of liver-related events (LRE) in primary biliary cholangitis (PBC) has not been attained thus far. This study investigated whether serum autotaxin (ATX) could predict LRE in patients with PBC. METHODS This retrospective multicenter investigation included 190 biopsy-proven untreated patients with PBC. All subjects were followed for at least 1 year, during which time the prevalence of LRE, including newly developing hepatocellular carcinoma, esophagogastric varices, ascites, and hepatic encephalopathy, was investigated in relation with ATX levels at the time of liver biopsy. RESULTS During the median follow-up period of 9.7 years, LRE were observed in 22 patients (11.6%). The area under the receiver operating characteristic curve and cutoff value of serum ATX for predicting LRE were 0.80 and 1.086 mg/L, respectively. Patients with serum ATX ≥1.086 had a significantly higher cumulative incidence of LRE compared with patients with ATX < 1.086 (33.3% vs 3.6%, P < 0.00001). Notably, the predictive capability of ATX for LRE in patients with PBC surpassed that of FIB-4, ALBI, APRI, and Mac-2-binding protein glycan isomer. A multivariate Cox proportional hazards model revealed ATX as an independent associated factor for LRE (hazard ratio 6.24, 95% confidence interval 1.87-20.80, P = 0.003) along with Nakanuma stage (hazard ratio 2.75, 95% confidence interval 1.52-4.99, P < 0.001). These results were closely replicated in a serologically diagnosed PBC validation cohort (n = 32). DISCUSSION Serum ATX levels may serve as a predictive marker for LRE in patients with PBC.
Collapse
Affiliation(s)
- Takanobu Iwadare
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
| | - Takefumi Kimura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan;
| | - Yuki Yamashita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan;
| | - Taiki Okumura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
| | - Shun-ichi Wakabayashi
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
| | - Hiroyuki Kobayashi
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
| | - Ayumi Sugiura
- Department of Internal Medicine, Sato Hospital, Nakano, Japan;
| | - Tomoo Yamazaki
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
- Department of Medicine, University of California San Diego, La Jolla, USA;
| | | | - Koji Igarashi
- Bioscience Division, TOSOH Corporation, Ayase, Japan;
| | - Satoru Joshita
- Department of Medicine, Yodakubo Hospital, Nagawa, Japan.
| | - Takeji Umemura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan;
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan;
| |
Collapse
|
4
|
Mulder IA, Abbinanti M, Woller SA, Ruschel J, Coutinho JM, de Vries HE, van Bavel E, Rosen K, McKerracher L, Ayata C. The novel ROCK2 selective inhibitor NRL-1049 preserves the blood-brain barrier after acute injury. J Cereb Blood Flow Metab 2024; 44:1238-1252. [PMID: 38833563 PMCID: PMC11542141 DOI: 10.1177/0271678x241238845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 06/06/2024]
Abstract
Endothelial blood-brain barrier (BBB) dysfunction is critical in the pathophysiology of brain injury. Rho-associated protein kinase (ROCK) activation disrupts BBB integrity in the injured brain. We aimed to test the efficacy of a novel ROCK2 inhibitor in preserving the BBB after acute brain injury. We characterized the molecular structure and pharmacodynamic and pharmacokinetic properties of a novel selective ROCK2 inhibitor, NRL-1049, and its first metabolite, 1-hydroxy-NRL-1049 (referred to as NRL-2017 hereon) and tested the efficacy of NRL-1049 on the BBB integrity in rodent models of acute brain injury. Our data show that NRL-1049 and NRL-2017 both inhibit ROCK activity and are 44-fold and 17-fold more selective towards ROCK2 than ROCK1, respectively. When tested in a mouse model of cortical cryoinjury, NRL-1049 significantly attenuated the increase in water content. Interestingly, 60% of the mice in the vehicle arm developed seizures within 2 hours after cryoinjury versus none in the NRL-1049 arm. In spontaneously hypertensive rats, NRL-1049 attenuated the dramatic surge in Evans Blue extravasation compared with the vehicle arm after transient middle cerebral artery occlusion. Hemorrhagic transformation was also reduced. We show that NRL-1049, a selective ROCK2 inhibitor, is a promising drug candidate to preserve the BBB after brain injury.
Collapse
Affiliation(s)
- Inge A Mulder
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Biomedical Engineering and Physics, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
| | | | | | | | - Jonathan M Coutinho
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
- Department of Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Helga E de Vries
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit, Amsterdam, the Netherlands
| | - Ed van Bavel
- Department of Biomedical Engineering and Physics, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
- Amsterdam Neurosciences, Neurovascular Disorders, Amsterdam, the Netherlands
| | | | - Lisa McKerracher
- BioAxone BioSciences Inc, Boston, MA, USA
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Stroke Service, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Fan X, Chu R, Jiang X, Ma P, Chu Y, Hua T, Yang M, Ding R, Li J, Xiang Z, Yuan H. LPAR6 Participates in Neuropathic Pain by Mediating Astrocyte Cells via ROCK2/NF-κB Signal Pathway. Mol Neurobiol 2024; 61:8402-8413. [PMID: 38509397 DOI: 10.1007/s12035-024-04108-5] [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: 09/03/2023] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Neuropathic pain (NPP) is a common type of chronic pain. Glial cells, including astrocytes (AS), are believed to play an important role in the progression of NPP. AS cells can be divided into various types based on their expression profiles, among which A1 and A2 types have clear functions. A1-type AS cells are neurotoxic, while A2-type AS cells exert neuroprotective functions. Some types of lysophosphatidic acid receptors (LPAR) have been shown to play a role in NPP. However, it remains unclear how AS cells and LPAR6 affect the occurrence and progression of NPP. In this study, we established a mouse model of chronic constriction injury (CCI) to simulate NPP. It was found that the expression of LPAR6 in AS cells of the spinal dorsal horn was increased in the CCI model, and the thresholds of mechanical and thermal pain were elevated after knocking out LPAR6, indicating that LPAR6 and AS cells participated in the occurrence of NPP. The experiment involved culturing primary AS cells and knocking down LPAR6 by Lentivirus. The results showed that the NF-κB signal pathway was activated and the number of A1-type AS cells increased in the CCI model. However, LPAR6 knockdown inhibited the NF-κB signal pathway and A1-type AS cells. The results of the mRNA sequencing and immunoprecipitation test indicate an interaction between LPAR6 and ROCK2. Inhibiting ROCK2 by Y-27632 increased mechanical and thermal pain thresholds and alleviated NPP at the molecular level. The study presents evidence that LPAR6 activates the NF-κB pathway through ROCK2 and contributes to the progression of NPP by increasing A1-type AS and decreasing A2-type AS. This suggests that LPAR6 could be a potential therapeutic target for alleviating NPP. Clinical applications that are successful can offer new therapeutic options, enhance the quality of life for patients, and potentially uncover new mechanisms for pain modulation.
Collapse
Affiliation(s)
- Xiaoyi Fan
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Ruitong Chu
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Xin Jiang
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Peng Ma
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Yan Chu
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Tong Hua
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Mei Yang
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Ruifeng Ding
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Jian Li
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Zhenghua Xiang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology, Ministry of Education, Naval Medical University, No.800 Xiangyin Road, Shanghai, 200433, People's Republic of China.
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, People's Republic of China.
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology, Ministry of Education, Naval Medical University, No.800 Xiangyin Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
6
|
Iwadare T, Kimura T, Okumura T, Wakabayashi SI, Nakajima T, Kondo S, Kobayashi H, Yamashita Y, Sugiura A, Fujimori N, Yamazaki T, Kunimoto H, Shimamoto S, Igarashi K, Joshita S, Tanaka N, Umemura T. Serum autotaxin is a prognostic indicator of liver-related events in patients with non-alcoholic fatty liver disease. COMMUNICATIONS MEDICINE 2024; 4:73. [PMID: 38627520 PMCID: PMC11021564 DOI: 10.1038/s43856-024-00499-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Circulating autotaxin (ATX) levels have been reported to correlate with liver inflammation activity and liver fibrosis severity in patients with non-alcoholic fatty liver disease (NAFLD). The objective of this study is to investigate whether serum ATX could predict liver-related events (LRE) in NAFLD patients. METHODS This retrospective investigation includes 309 biopsy-proven NAFLD patients registered at Shinshu University Hospital. All patients are followed for at least 1 year, during which time the prevalence of LRE, including newly developing hepatocellular carcinoma, hepatic encephalopathy, ascites, and esophagogastric varices, is investigated in relation to ATX levels at the time of liver biopsy. RESULTS During the median follow-up period of 7.0 years, LRE are observed in 20 patients (6.5%). The area under the receiver operating characteristic curve and cut-off value of serum ATX for predicting LRE are 0.81 and 1.227 mg/l, respectively. Multivariate Cox proportional hazards models for LRE determine ATX and advanced fibrosis as independently associated factors. Furthermore, in a competing risk analysis that considered non-liver-related death as a competing event, ATX (HR 2.29, 95% CI 1.22-4.30, p = 0.010) is identified as an independent factor associated with LRE, along with advanced fibrosis (HR 8.01, 95% CI 2.10-30.60, p = 0.002). The predictive utility of ATX for LRE is validated in an independent cohort. CONCLUSIONS Serum ATX may serve as a predictive marker for LRE in patients with NAFLD.
Collapse
Affiliation(s)
- Takanobu Iwadare
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takefumi Kimura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan.
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan.
| | - Taiki Okumura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shun-Ichi Wakabayashi
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Taro Nakajima
- Department of Gastroenterology, Maruko Central Hospital, Ueda, Japan
| | - Shohei Kondo
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroyuki Kobayashi
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yuki Yamashita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Ayumi Sugiura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoyuki Fujimori
- Department of Hepatology, Shinshu Ueda Medical Center, Ueda, Japan
| | - Tomoo Yamazaki
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hideo Kunimoto
- Department of Hepatology, Nagano Municipal Hospital, Nagano, Japan
| | | | - Koji Igarashi
- Bioscience Division, TOSOH Corporation, Ayase, Kanagawa, Japan
| | - Satoru Joshita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoki Tanaka
- Department of Global Medical Research Promotion, Shinshu University Graduate School of Medicine, Matsumoto, Japan
- International Relations Office, Shinshu University School of Medicine, Matsumoto, Japan
- Research Center for Social Systems, Shinshu University, Matsumoto, Japan
| | - Takeji Umemura
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
- Consultation Center for Liver Diseases, Shinshu University Hospital, Matsumoto, Japan
| |
Collapse
|
7
|
Malar DS, Verma K, Prasanth MI, Tencomnao T, Brimson JM. Network analysis-guided drug repurposing strategies targeting LPAR receptor in the interplay of COVID, Alzheimer's, and diabetes. Sci Rep 2024; 14:4328. [PMID: 38383841 PMCID: PMC10882047 DOI: 10.1038/s41598-024-55013-9] [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: 09/15/2023] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has greatly affected global health. Emerging evidence suggests a complex interplay between Alzheimer's disease (AD), diabetes (DM), and COVID-19. Given COVID-19's involvement in the increased risk of other diseases, there is an urgent need to identify novel targets and drugs to combat these interconnected health challenges. Lysophosphatidic acid receptors (LPARs), belonging to the G protein-coupled receptor family, have been implicated in various pathological conditions, including inflammation. In this regard, the study aimed to investigate the involvement of LPARs (specifically LPAR1, 3, 6) in the tri-directional relationship between AD, DM, and COVID-19 through network analysis, as well as explore the therapeutic potential of selected anti-AD, anti-DM drugs as LPAR, SPIKE antagonists. We used the Coremine Medical database to identify genes related to DM, AD, and COVID-19. Furthermore, STRING analysis was used to identify the interacting partners of LPAR1, LPAR3, and LPAR6. Additionally, a literature search revealed 78 drugs on the market or in clinical studies that were used for treating either AD or DM. We carried out docking analysis of these drugs against the LPAR1, LPAR3, and LPAR6. Furthermore, we modeled the LPAR1, LPAR3, and LPAR6 in a complex with the COVID-19 spike protein and performed a docking study of selected drugs with the LPAR-Spike complex. The analysis revealed 177 common genes implicated in AD, DM, and COVID-19. Protein-protein docking analysis demonstrated that LPAR (1,3 & 6) efficiently binds with the viral SPIKE protein, suggesting them as targets for viral infection. Furthermore, docking analysis of the anti-AD and anti-DM drugs against LPARs, SPIKE protein, and the LPARs-SPIKE complex revealed promising candidates, including lupron, neflamapimod, and nilotinib, stating the importance of drug repurposing in the drug discovery process. These drugs exhibited the ability to bind and inhibit the LPAR receptor activity and the SPIKE protein and interfere with LPAR-SPIKE protein interaction. Through a combined network and targeted-based therapeutic intervention approach, this study has identified several drugs that could be repurposed for treating COVID-19 due to their expected interference with LPAR(1, 3, and 6) and spike protein complexes. In addition, it can also be hypothesized that the co-administration of these identified drugs during COVID-19 infection may not only help mitigate the impact of the virus but also potentially contribute to the prevention or management of post-COVID complications related to AD and DM.
Collapse
Affiliation(s)
- Dicson Sheeja Malar
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Kanika Verma
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
- Department of Molecular Epidemiology, ICMR- National Institute of Malaria Research (NIMR), New Delhi, India.
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - James Michael Brimson
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Research Unit for Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
8
|
Petersen-Cherubini CL, Liu Y, Deffenbaugh JL, Murphy SP, Xin M, Rau CN, Yang Y, Lovett-Racke AE. Dysregulated autotaxin expression by T cells in multiple sclerosis. J Neuroimmunol 2024; 387:578282. [PMID: 38183947 PMCID: PMC10923181 DOI: 10.1016/j.jneuroim.2023.578282] [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: 11/13/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Multiple sclerosis (MS) is a demyelinating disease characterized by infiltration of autoreactive T cells into the central nervous system (CNS). In order to understand how activated, autoreactive T cells are able to cross the blood brain barrier, the unique molecular characteristics of pathogenic T cells need to be more thoroughly examined. In previous work, our laboratory found autotaxin (ATX) to be upregulated by activated autoreactive T cells in the mouse model of MS. ATX is a secreted glycoprotein that promotes T cell chemokinesis and transmigration through catalysis of lysophoshphatidic acid (LPA). ATX is elevated in the serum of MS patients during active disease phases, and we previously found that inhibiting ATX decreases severity of neurological deficits in the mouse model. In this study, ATX expression was found to be lower in MS patient immune cells during rest, but significantly increased during early activation in a manner not seen in healthy controls. The ribosomal binding protein HuR, which stabilizes ATX mRNA, was also increased in MS patients in a similar pattern to that of ATX, suggesting it may be helping regulate ATX levels after activation. The proinflammatory cytokine interleukin-23 (IL-23) was shown to induce prolonged ATX expression in MS patient Th1 and Th17 cells. Finally, through ChIP, re-ChIP analysis, we show that IL-23 may be signaling through pSTAT3/pSTAT4 heterodimers to induce expression of ATX. Taken together, these findings elucidate cell types that may be contributing to elevated serum ATX levels in MS patients and identify potential drivers of sustained expression in encephalitogenic T cells.
Collapse
Affiliation(s)
- Cora L Petersen-Cherubini
- The Ohio State University, Neuroscience Graduate Program, 460 West 12th Avenue, Biomedical Research Tower 6894, Columbus, OH 43210, USA; The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA
| | - Yue Liu
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Joshua L Deffenbaugh
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Shawn P Murphy
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Matthew Xin
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA
| | - Christina N Rau
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Yuhong Yang
- The Ohio State University, Wexner Medical Center, Department of Neurology, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA
| | - Amy E Lovett-Racke
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA; The Ohio State University, Wexner Medical Center, Department of Neuroscience, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| |
Collapse
|
9
|
Hajipour S, Farbood Y, Dianat M, Nesari A, Sarkaki A. Effect of Berberine against Cognitive Deficits in Rat Model of Thioacetamide-Induced Liver Cirrhosis and Hepatic Encephalopathy (Behavioral, Biochemical, Molecular and Histological Evaluations). Brain Sci 2023; 13:944. [PMID: 37371422 DOI: 10.3390/brainsci13060944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/17/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Liver cirrhosis (LC) is one of the chronic liver diseases with high disability and mortality accompanying hepatic encephalopathy (HE) followed by cognitive dysfunctions. In this work, the effect of berberine (Ber) on spatial cognition was studied in a rat model of LC induced by thioacetamide (TAA). MATERIALS AND METHODS Male Wistar rats (200-250 g) were divided into six groups: (1) control; (2) TAA, 200 mg/kg/day, i.p.; (3-5) TAA + Ber; received Ber (10, 30, and 60 mg/kg, i.p., daily after last TAA injection); (6) Dizocilpine (MK-801) + TAA, received MK-801 (2 mg/kg/day, i.p.) 30 m before TAA injection. The spatial memory, BBB permeability, brain edema, liver enzymes, urea, serum and brain total bilirubin, oxidative stress and cytokine markers in the hippocampus were measured. Furthermore, a histological examination of the hippocampus was carried out. RESULTS The BBB permeability, brain edema, liver enzymes, urea, total bilirubin levels in serum and hippocampal MDA and TNF-α increased significantly after TAA injection (p < 0.001); the spatial memory was impaired (p < 0.001), and hippocampal IL-10 decreased (p < 0.001). Ber reversed all the above parameters significantly (p < 0.05, p < 0.01 and p < 0.001). MK-801 prevented the development of LC via TAA (p < 0.001). CONCLUSION Results showed that Ber improves spatial learning and memory in TAA-induced LC by improving the BBB function, oxidative stress and neuroinflammation. Ber might be a promising therapeutic agent for cognitive improvement in LC.
Collapse
Affiliation(s)
- Somayeh Hajipour
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Yaghoob Farbood
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Mahin Dianat
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Ali Nesari
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Alireza Sarkaki
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- National Institute for Medical Research Development "NIMAD", Tehran 1419693111, Iran
| |
Collapse
|
10
|
Sepehrinezhad A, Stolze Larsen F, Ashayeri Ahmadabad R, Shahbazi A, Sahab Negah S. The Glymphatic System May Play a Vital Role in the Pathogenesis of Hepatic Encephalopathy: A Narrative Review. Cells 2023; 12:cells12070979. [PMID: 37048052 PMCID: PMC10093707 DOI: 10.3390/cells12070979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Hepatic encephalopathy (HE) is a neurological complication of liver disease resulting in cognitive, psychiatric, and motor symptoms. Although hyperammonemia is a key factor in the pathogenesis of HE, several other factors have recently been discovered. Among these, the impairment of a highly organized perivascular network known as the glymphatic pathway seems to be involved in the progression of some neurological complications due to the accumulation of misfolded proteins and waste substances in the brain interstitial fluids (ISF). The glymphatic system plays an important role in the clearance of brain metabolic derivatives and prevents aggregation of neurotoxic agents in the brain ISF. Impairment of it will result in aggravated accumulation of neurotoxic agents in the brain ISF. This could also be the case in patients with liver failure complicated by HE. Indeed, accumulation of some metabolic by-products and agents such as ammonia, glutamine, glutamate, and aromatic amino acids has been reported in the human brain ISF using microdialysis technique is attributed to worsening of HE and correlates with brain edema. Furthermore, it has been reported that the glymphatic system is impaired in the olfactory bulb, prefrontal cortex, and hippocampus in an experimental model of HE. In this review, we discuss different factors that may affect the function of the glymphatic pathways and how these changes may be involved in HE.
Collapse
Affiliation(s)
- Ali Sepehrinezhad
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
| | - Fin Stolze Larsen
- Department of Gastroenterology and Hepatology, Rigshospitalet, Copenhagen University Hospital, 999017 Copenhagen, Denmark
| | | | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1449614535, Iran
| |
Collapse
|
11
|
Spencer SA, Suárez-Pozos E, Verdugo JS, Wang H, Afshari FS, Li G, Manam S, Yasuda D, Ortega A, Lister JA, Ishii S, Zhang Y, Fuss B. Lysophosphatidic acid signaling via LPA 6 : A negative modulator of developmental oligodendrocyte maturation. J Neurochem 2022; 163:478-499. [PMID: 36153691 PMCID: PMC9772207 DOI: 10.1111/jnc.15696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/14/2023]
Abstract
The developmental process of central nervous system (CNS) myelin sheath formation is characterized by well-coordinated cellular activities ultimately ensuring rapid and synchronized neural communication. During this process, myelinating CNS cells, namely oligodendrocytes (OLGs), undergo distinct steps of differentiation, whereby the progression of earlier maturation stages of OLGs represents a critical step toward the timely establishment of myelinated axonal circuits. Given the complexity of functional integration, it is not surprising that OLG maturation is controlled by a yet fully to be defined set of both negative and positive modulators. In this context, we provide here first evidence for a role of lysophosphatidic acid (LPA) signaling via the G protein-coupled receptor LPA6 as a negative modulatory regulator of myelination-associated gene expression in OLGs. More specifically, the cell surface accessibility of LPA6 was found to be restricted to the earlier maturation stages of differentiating OLGs, and OLG maturation was found to occur precociously in Lpar6 knockout mice. To further substantiate these findings, a novel small molecule ligand with selectivity for preferentially LPA6 and LPA6 agonist characteristics was functionally characterized in vitro in primary cultures of rat OLGs and in vivo in the developing zebrafish. Utilizing this approach, a negative modulatory role of LPA6 signaling in OLG maturation could be corroborated. During development, such a functional role of LPA6 signaling likely serves to ensure timely coordination of circuit formation and myelination. Under pathological conditions as seen in the major human demyelinating disease multiple sclerosis (MS), however, persistent LPA6 expression and signaling in OLGs can be seen as an inhibitor of myelin repair. Thus, it is of interest that LPA6 protein levels appear elevated in MS brain samples, thereby suggesting that LPA6 signaling may represent a potential new druggable pathway suitable to promote myelin repair in MS.
Collapse
Affiliation(s)
- Samantha A Spencer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Edna Suárez-Pozos
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jazmín Soto Verdugo
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Huiqun Wang
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Fatemah S Afshari
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Guo Li
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Susmita Manam
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Daisuke Yasuda
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - James A Lister
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Satoshi Ishii
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
12
|
Tang Y, Cheng Y, Wang S, Wang Y, Liu P, Wu H. Review: The Development of Risk Factors and Cytokines in Retinal Vein Occlusion. Front Med (Lausanne) 2022; 9:910600. [PMID: 35783660 PMCID: PMC9240302 DOI: 10.3389/fmed.2022.910600] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/23/2022] [Indexed: 12/13/2022] Open
Abstract
Retinal vein occlusion (RVO) is the second most prevalent retinal disease. Despite this, the pathogenic mechanisms and risk factors are not entirely clear. In this article, we review recent publications on the classification, pathogenesis, risk factors, ischemic changes, cytokines, and vital complications of RVO. Risk factors and cytokines are important for exploring the mechanisms and new treatment targets. Furthermore, risk factors are interrelated, making RVO mechanisms more complex. Cytokines act as powerful mediators of pathological conditions, such as inflammation, neovascularization, and macular edema. This review aims to summarize the updated knowledge on risk factors, cytokines of RVO and signaling in order to provide valuable insight on managing the disease.
Collapse
Affiliation(s)
- Yi Tang
- Eye Center of Second Hospital, Jilin University, Changchun, China
| | - Yan Cheng
- Eye Center of Second Hospital, Jilin University, Changchun, China
| | - Shuo Wang
- Eye Center of Second Hospital, Jilin University, Changchun, China
| | - Yongjie Wang
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Pengjia Liu
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, Australia
| | - Hong Wu
- Eye Center of Second Hospital, Jilin University, Changchun, China
- *Correspondence: Hong Wu
| |
Collapse
|
13
|
Devasani K, Yao Y. Expression and functions of adenylyl cyclases in the CNS. Fluids Barriers CNS 2022; 19:23. [PMID: 35307032 PMCID: PMC8935726 DOI: 10.1186/s12987-022-00322-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/07/2022] [Indexed: 12/27/2022] Open
Abstract
Adenylyl cyclases (ADCYs), by generating second messenger cAMP, play important roles in various cellular processes. Their expression, regulation and functions in the CNS, however, remain largely unknown. In this review, we first introduce the classification and structure of ADCYs, followed by a discussion of the regulation of mammalian ADCYs (ADCY1-10). Next, the expression and function of each mammalian ADCY isoform are summarized in a region/cell-specific manner. Furthermore, the effects of GPCR-ADCY signaling on blood-brain barrier (BBB) integrity are reviewed. Last, current challenges and future directions are discussed. We aim to provide a succinct review on ADCYs to foster new research in the future.
Collapse
Affiliation(s)
- Karan Devasani
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC 8, Tampa, FL, 33612, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC 8, Tampa, FL, 33612, USA.
| |
Collapse
|
14
|
Lee R, Lee BH, Choi SH, Cho YJ, Cho HS, Kim HC, Rhim H, Cho IH, Rhee MH, Nah SY. Effects of Gintonin-enriched fraction on the gene expression of six lysophosphatidic receptor subtypes. J Ginseng Res 2021; 45:583-590. [PMID: 34803428 PMCID: PMC8587509 DOI: 10.1016/j.jgr.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/17/2021] [Indexed: 11/03/2022] Open
Abstract
Background Gintonin, isolated from ginseng, acts as a ginseng-derived lysophosphatidic acid (LPA) receptor ligand and elicits the [Ca2+]i transient through six LPA receptor subtypes (LPARSs). However, the long-term effects of gintonin-enriched fraction (GEF) on the gene expression of six LPARSs remain unknown. We examined changes in the gene expression of six LPA receptors in the mouse whole brain, heart, lungs, liver, kidneys, spleen, small intestine, colon, and testis after long-term oral GEF administration. Methods C57BL/6 mice were divided into two groups: control vehicle and GEF (100 mg/kg, p.o.). After 21-day saline or GEF treatment, total RNA was extracted from nine mouse organs. Quantitative-real-time PCR (qRT-PCR) and western blot were performed to quantify changes in the gene and protein expression of the six LPARSs, respectively. Results qRT-PCR analysis before GEF treatment revealed that the LPA6 RS was predominant in all organs except the small intestine. The LPA2 RS was most abundant in the small intestine. Long-term GEF administration differentially regulated the six LPARSs. Upon GEF treatment, the LPA6 RS significantly increased in the liver, small intestine, colon, and testis but decreased in the whole brain, heart, lungs, and kidneys. Western blot analysis of the LPA6 RS confirmed the differential effects of GEF on LPA6 receptor protein levels in the whole brain, liver, small intestine, and testis. Conclusion The LPA6 receptor was predominantly expressed in all nine organs examined; long-term oral GEF administration differentially regulated LPA3, LPA4, and LPA6 receptors in the whole brain, heart, lungs, liver, kidneys, small intestine, and testis.
Collapse
Affiliation(s)
- Rami Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Byung-Hwan Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Yeon-Jin Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Han-Sung Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Man Hee Rhee
- Laboratory of Veterinary Physiology and Cell Signaling, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Takata F, Nakagawa S, Matsumoto J, Dohgu S. Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. Front Cell Neurosci 2021; 15:661838. [PMID: 34588955 PMCID: PMC8475767 DOI: 10.3389/fncel.2021.661838] [Citation(s) in RCA: 283] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is involved in the onset or progression of various neurodegenerative diseases. Initiation of neuroinflammation is triggered by endogenous substances (damage-associated molecular patterns) and/or exogenous pathogens. Activation of glial cells (microglia and astrocytes) is widely recognized as a hallmark of neuroinflammation and triggers the release of proinflammatory cytokines, leading to neurotoxicity and neuronal dysfunction. Another feature associated with neuroinflammatory diseases is impairment of the blood-brain barrier (BBB). The BBB, which is composed of brain endothelial cells connected by tight junctions, maintains brain homeostasis and protects neurons. Impairment of this barrier allows trafficking of immune cells or plasma proteins into the brain parenchyma and subsequent inflammatory processes in the brain. Besides neurons, activated glial cells also affect BBB integrity. Therefore, BBB dysfunction can amplify neuroinflammation and act as a key process in the development of neuroinflammation. BBB integrity is determined by the integration of multiple signaling pathways within brain endothelial cells through intercellular communication between brain endothelial cells and brain perivascular cells (pericytes, astrocytes, microglia, and oligodendrocytes). For prevention of BBB disruption, both cellular components, such as signaling molecules in brain endothelial cells, and non-cellular components, such as inflammatory mediators released by perivascular cells, should be considered. Thus, understanding of intracellular signaling pathways that disrupt the BBB can provide novel treatments for neurological diseases associated with neuroinflammation. In this review, we discuss current knowledge regarding the underlying mechanisms involved in BBB impairment by inflammatory mediators released by perivascular cells.
Collapse
Affiliation(s)
- Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Junichi Matsumoto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
16
|
Meduri B, Pujar GV, Durai Ananda Kumar T, Akshatha HS, Sethu AK, Singh M, Kanagarla A, Mathew B. Lysophosphatidic acid (LPA) receptor modulators: Structural features and recent development. Eur J Med Chem 2021; 222:113574. [PMID: 34126459 DOI: 10.1016/j.ejmech.2021.113574] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023]
Abstract
Lysophosphatidic acid (LPA) activates six LPA receptors (LPAR1-6) and regulates various cellular activities such as cell proliferation, cytoprotection, and wound healing. Many studies elucidated the pathological outcomes of LPA are due to the alteration in signaling pathways, which include migration and invasion of cancer cells, fibrosis, atherosclerosis, and inflammation. Current pathophysiological research on LPA and its receptors provides a means that LPA receptors are new therapeutic targets for disorders associated with LPA. Various chemical modulators are developed and are under investigation to treat a wide range of pathological complications. This review summarizes the physiological and pathological roles of LPA signaling, development of various LPA modulators, their structural features, patents, and their clinical outcomes.
Collapse
Affiliation(s)
- Bhagyalalitha Meduri
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015 India
| | - Gurubasavaraj Veeranna Pujar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015 India.
| | - T Durai Ananda Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015 India
| | - H S Akshatha
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015 India
| | - Arun Kumar Sethu
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015 India
| | - Manisha Singh
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015 India
| | - Abhinav Kanagarla
- Department of Pharmaceutical Chemistry, Andhra University, Visakhapatnam, Andhra Pradesh, 530003, India
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
17
|
Geraldo LHM, Spohr TCLDS, Amaral RFD, Fonseca ACCD, Garcia C, Mendes FDA, Freitas C, dosSantos MF, Lima FRS. Role of lysophosphatidic acid and its receptors in health and disease: novel therapeutic strategies. Signal Transduct Target Ther 2021; 6:45. [PMID: 33526777 PMCID: PMC7851145 DOI: 10.1038/s41392-020-00367-5] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidic acid (LPA) is an abundant bioactive phospholipid, with multiple functions both in development and in pathological conditions. Here, we review the literature about the differential signaling of LPA through its specific receptors, which makes this lipid a versatile signaling molecule. This differential signaling is important for understanding how this molecule can have such diverse effects during central nervous system development and angiogenesis; and also, how it can act as a powerful mediator of pathological conditions, such as neuropathic pain, neurodegenerative diseases, and cancer progression. Ultimately, we review the preclinical and clinical uses of Autotaxin, LPA, and its receptors as therapeutic targets, approaching the most recent data of promising molecules modulating both LPA production and signaling. This review aims to summarize the most update knowledge about the mechanisms of LPA production and signaling in order to understand its biological functions in the central nervous system both in health and disease.
Collapse
Affiliation(s)
- Luiz Henrique Medeiros Geraldo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | | | | | | | - Celina Garcia
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio de Almeida Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Catarina Freitas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Fabio dosSantos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavia Regina Souza Lima
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
19
|
Czarnecka AM, Milewski K, Albrecht J, Zielińska M. The Status of Bile Acids and Farnesoid X Receptor in Brain and Liver of Rats with Thioacetamide-Induced Acute Liver Failure. Int J Mol Sci 2020; 21:ijms21207750. [PMID: 33092050 PMCID: PMC7589210 DOI: 10.3390/ijms21207750] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
Acute liver failure (ALF) leads to neurological symptoms defined as hepatic encephalopathy (HE). Although accumulation of ammonia and neuroinflammation are generally accepted as main contributors to HE pathomechanism, a buildup of bile acids (BA) in the blood is a frequent component of liver injury in HE patients. Recent studies have identified the nuclear farnesoid X receptor (FXR) acting via small heterodimer partner (SHP) as a mediator of BA-induced effects in the brain of ALF animals. The present study investigated the status of the BA–FXR axis in the brain and the liver, including selective changes in pertinent genes in thioacetamide (TAA)-induced ALF in Sprague–Dawley rats. FXR was found in rat neurons, confirming earlier reports for mouse and human brain. BA accumulated in blood but not in the brain tissue. Expression of mRNAs coding for Fxr and Shp was reduced in the hippocampus and of Fxr mRNA also in the cerebellum. Changes in Fxr mRNA levels were not followed by changes in FXR protein. The results leave open the possibility that mobilization of the BA–FXR axis in the brain may not be necessarily pathognomonic to HE but may depend upon ALF-related confounding factors.
Collapse
Affiliation(s)
- Anna Maria Czarnecka
- Correspondence: (A.M.C.); (M.Z.); Tel.: +48-22-6086416 (A.M.C.); +48-22-6086470 (M.Z.)
| | | | | | - Magdalena Zielińska
- Correspondence: (A.M.C.); (M.Z.); Tel.: +48-22-6086416 (A.M.C.); +48-22-6086470 (M.Z.)
| |
Collapse
|
20
|
Bolla BS, Erdei L, Urbán E, Burián K, Kemény L, Szabó K. Cutibacterium acnes regulates the epidermal barrier properties of HPV-KER human immortalized keratinocyte cultures. Sci Rep 2020; 10:12815. [PMID: 32733073 PMCID: PMC7393503 DOI: 10.1038/s41598-020-69677-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Our skin provides a physical barrier to separate the internal part of our body from the environment. Maintenance of complex barrier functions is achieved through anatomical structures in the skin, the stratified squamous epithelium specialized junctional organelles, called tight junctions (TJs). Several members of our microbial communities are known to affect the differentiation state and function of the colonized organ. Whether and how interactions between skin cells and cutaneous microbes, including Cutibacterium acnes (C. acnes), modify the structure and/or function of our skin is currently only partly understood. Thus, in our studies, we investigated whether C. acnes may affect the epidermal barrier using in vitro model systems. Real-time cellular analysis showed that depending on the keratinocyte differentiation state, the applied C. acnes strains and their dose, the measured impedance values change, together with the expression of selected TJ proteins. These may reflect barrier alterations, which can be partially restored upon antibiotic–antimycotic treatment. Our findings suggest that C. acnes can actively modify the barrier properties of cultured keratinocytes, possibly through alteration of tight cell-to-cell contacts. Similar events may play important roles in our skin, in the maintenance of cutaneous homeostasis.
Collapse
Affiliation(s)
- Beáta Szilvia Bolla
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Lilla Erdei
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Edit Urbán
- Department of Public Health, University of Szeged, Szeged, Hungary
| | - Katalin Burián
- Institute of Clinical Microbiology, University of Szeged, Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Kornélia Szabó
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary. .,MTA-SZTE Dermatological Research Group, Szeged, Hungary.
| |
Collapse
|
21
|
Pluimer BR, Colt M, Zhao Z. G Protein-Coupled Receptors in the Mammalian Blood-Brain Barrier. Front Cell Neurosci 2020; 14:139. [PMID: 32581715 PMCID: PMC7283493 DOI: 10.3389/fncel.2020.00139] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
The mammalian neurovascular unit (NVU) is comprised of neurons, glia, and vascular cells. The NVU is the nexus between the cardiovascular and central nervous system (CNS). The central component of the NVU is the blood-brain barrier (BBB) which consists of a monolayer of tightly connected endothelial cells covered by pericytes and further surrounded by astrocytic endfeet. In addition to preventing the diffusion of toxic species into the CNS, the BBB endothelium serves as a dynamic regulatory system facilitating the transport of molecules from the bloodstream to the brain and vis versa. The structural integrity and transport functions of the BBB are maintained, in part, by an orchestra of membrane receptors and transporters including members of the superfamily of G protein-coupled receptors (GPCRs). Here, we provide an overview of GPCRs known to regulate mammalian BBB structure and function and discuss how dysregulation of these pathways plays a role in various neurodegenerative diseases.
Collapse
Affiliation(s)
- Brock R. Pluimer
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Mark Colt
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
22
|
Cerebral Ischemia-Reperfusion Injury: Lysophosphatidic Acid Mediates Inflammation by Decreasing the Expression of Liver X Receptor. J Mol Neurosci 2020; 70:1376-1384. [PMID: 32424512 DOI: 10.1007/s12031-020-01554-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/13/2020] [Indexed: 10/24/2022]
Abstract
Lysophosphatidic acid (LPA), a ubiquitous phospholipid, plays a crucial role in the pathogenesis and pathophysiological process of neurological diseases, which constitute the pathological course after cerebral ischemia. Nevertheless, the molecular mechanisms associated with the pathogenic roles of LPA remain elusive. In this study, we evaluated the expression of the liver X receptor (LXR) and nuclear factor kappa B (NFκB) by Western blotting, quantified the levels of IL-1β, IL-6, TNF-α, and LPA by ELISA, and evaluated apoptosis and infarct by TUNEL (terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling) and TTC (triphenyltetrazolium chloride) staining respectively in Sprague-Dawley (SD) rats after middle cerebral artery occlusion (MCAO). The levels of LPA, an extracellular signaling molecule, increased after ischemia and caused neurological injury effect, decreased the expression level of LXR, and increased the expression level of inflammatory factors (IL-1β, IL-6, and TNF-α) via the NFκB signaling pathway. This elevated LPA-induced pathological process is one of the pathological reactions associated with ischemic brain injury. We present a direct or indirect connection between LPA and LXR in the pathophysiological process. In conclusion, we speculate that the inhibition of LPA generation and administration of LXR agonist may be explored as potential cerebral infarction treatment strategies.
Collapse
|
23
|
Sepehrinezhad A, Zarifkar A, Namvar G, Shahbazi A, Williams R. Astrocyte swelling in hepatic encephalopathy: molecular perspective of cytotoxic edema. Metab Brain Dis 2020; 35:559-578. [PMID: 32146658 DOI: 10.1007/s11011-020-00549-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/12/2020] [Indexed: 02/06/2023]
Abstract
Hepatic encephalopathy (HE) may occur in patients with liver failure. The most critical pathophysiologic mechanism of HE is cerebral edema following systemic hyperammonemia. The dysfunctional liver cannot eliminate circulatory ammonia, so its plasma and brain levels rise sharply. Astrocytes, the only cells that are responsible for ammonia detoxification in the brain, are dynamic cells with unique phenotypic properties that enable them to respond to small changes in their environment. Any pathological changes in astrocytes may cause neurological disturbances such as HE. Astrocyte swelling is the leading cause of cerebral edema, which may cause brain herniation and death by increasing intracranial pressure. Various factors may have a role in astrocyte swelling. However, the exact molecular mechanism of astrocyte swelling is not fully understood. This article discusses the possible mechanisms of astrocyte swelling which related to hyperammonia, including the possible roles of molecules like glutamine, lactate, aquaporin-4 water channel, 18 KDa translocator protein, glial fibrillary acidic protein, alanine, glutathione, toll-like receptor 4, epidermal growth factor receptor, glutamate, and manganese, as well as inflammation, oxidative stress, mitochondrial permeability transition, ATP depletion, and astrocyte senescence. All these agents and factors may be targeted in therapeutic approaches to HE.
Collapse
Affiliation(s)
- Ali Sepehrinezhad
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Asadollah Zarifkar
- Shiraz Neuroscience Research Center and Department of Physiology, Shiraz University of Medical Sciences (SUMS), Shiraz, Iran
| | - Gholamreza Namvar
- Department of Neuroscience and Cognition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Roger Williams
- The Institute of Hepatology London and Foundation for Liver Research, 111 Coldharbour Lane, London, SE5 9NT, UK.
- Faculty of Life Sciences & Medicine, King's College London, London, UK.
| |
Collapse
|
24
|
Xiang H, Lu Y, Shao M, Wu T. Lysophosphatidic Acid Receptors: Biochemical and Clinical Implications in Different Diseases. J Cancer 2020; 11:3519-3535. [PMID: 32284748 PMCID: PMC7150451 DOI: 10.7150/jca.41841] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Lysophosphatidic acid (LPA, 1-acyl-2-hemolytic-sn-glycerol-3-phosphate) extracted from membrane phospholipid is a kind of simple bioactive glycophospholipid, which has many biological functions such as stimulating cell multiplication, cytoskeleton recombination, cell survival, drug-fast, synthesis of DNA and ion transport. Current studies have shown that six G-coupled protein receptors (LPAR1-6) can be activated by LPA. They stimulate a variety of signal transduction pathways through heterotrimeric G-proteins (such as Gα12/13, Gαq/11, Gαi/o and GαS). LPA and its receptors play vital roles in cancers, nervous system diseases, cardiovascular diseases, liver diseases, metabolic diseases, etc. In this article, we discussed the structure of LPA receptors and elucidated their functions in various diseases, in order to better understand them and point out new therapeutic schemes for them.
Collapse
Affiliation(s)
- Hongjiao Xiang
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifei Lu
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingmei Shao
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tao Wu
- Center of Chinese Medical Therapy and Systems Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
25
|
Zhou Y, Little PJ, Ta HT, Xu S, Kamato D. Lysophosphatidic acid and its receptors: pharmacology and therapeutic potential in atherosclerosis and vascular disease. Pharmacol Ther 2019; 204:107404. [DOI: 10.1016/j.pharmthera.2019.107404] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
|
26
|
Cong X, Kong W. Endothelial tight junctions and their regulatory signaling pathways in vascular homeostasis and disease. Cell Signal 2019; 66:109485. [PMID: 31770579 DOI: 10.1016/j.cellsig.2019.109485] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
Endothelial tight junctions (TJs) regulate the transport of water, ions, and molecules through the paracellular pathway, serving as an important barrier in blood vessels and maintaining vascular homeostasis. In endothelial cells (ECs), TJs are highly dynamic structures that respond to multiple external stimuli and pathological conditions. Alterations in the expression, distribution, and structure of endothelial TJs may lead to many related vascular diseases and pathologies. In this review, we provide an overview of the assessment methods used to evaluate endothelial TJ barrier function both in vitro and in vivo and describe the composition of endothelial TJs in diverse vascular systems and ECs. More importantly, the direct phosphorylation and dephosphorylation of TJ proteins by intracellular kinases and phosphatases, as well as the signaling pathways involved in the regulation of TJs, including and the protein kinase C (PKC), PKA, PKG, Ras homolog gene family member A (RhoA), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)/Akt, and Wnt/β-catenin pathways, are discussed. With great advances in this area, targeting endothelial TJs may provide novel treatment for TJ-related vascular pathologies.
Collapse
Affiliation(s)
- Xin Cong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
27
|
Xu Y. Targeting Lysophosphatidic Acid in Cancer: The Issues in Moving from Bench to Bedside. Cancers (Basel) 2019; 11:E1523. [PMID: 31658655 PMCID: PMC6826372 DOI: 10.3390/cancers11101523] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022] Open
Abstract
Since the clear demonstration of lysophosphatidic acid (LPA)'s pathological roles in cancer in the mid-1990s, more than 1000 papers relating LPA to various types of cancer were published. Through these studies, LPA was established as a target for cancer. Although LPA-related inhibitors entered clinical trials for fibrosis, the concept of targeting LPA is yet to be moved to clinical cancer treatment. The major challenges that we are facing in moving LPA application from bench to bedside include the intrinsic and complicated metabolic, functional, and signaling properties of LPA, as well as technical issues, which are discussed in this review. Potential strategies and perspectives to improve the translational progress are suggested. Despite these challenges, we are optimistic that LPA blockage, particularly in combination with other agents, is on the horizon to be incorporated into clinical applications.
Collapse
Affiliation(s)
- Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut Street R2-E380, Indianapolis, IN 46202, USA.
| |
Collapse
|
28
|
Pleotropic Roles of Autotaxin in the Nervous System Present Opportunities for the Development of Novel Therapeutics for Neurological Diseases. Mol Neurobiol 2019; 57:372-392. [PMID: 31364025 DOI: 10.1007/s12035-019-01719-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/23/2019] [Indexed: 12/23/2022]
Abstract
Autotaxin (ATX) is a soluble extracellular enzyme that is abundant in mammalian plasma and cerebrospinal fluid (CSF). It has two known enzymatic activities, acting as both a phosphodiesterase and a phospholipase. The majority of its biological effects have been associated with its ability to liberate lysophosphatidic acid (LPA) from its substrate, lysophosphatidylcholine (LPC). LPA has diverse pleiotropic effects in the central nervous system (CNS) and other tissues via the activation of a family of six cognate G protein-coupled receptors. These LPA receptors (LPARs) are expressed in some combination in all known cell types in the CNS where they mediate such fundamental cellular processes as proliferation, differentiation, migration, chronic inflammation, and cytoskeletal organization. As a result, dysregulation of LPA content may contribute to many CNS and PNS disorders such as chronic inflammatory or neuropathic pain, glioblastoma multiforme (GBM), hemorrhagic hydrocephalus, schizophrenia, multiple sclerosis, Alzheimer's disease, metabolic syndrome-induced brain damage, traumatic brain injury, hepatic encephalopathy-induced cerebral edema, macular edema, major depressive disorder, stress-induced psychiatric disorder, alcohol-induced brain damage, HIV-induced brain injury, pruritus, and peripheral nerve injury. ATX activity is now known to be the primary biological source of this bioactive signaling lipid, and as such, represents a potentially high-value drug target. There is currently one ATX inhibitor entering phase III clinical trials, with several additional preclinical compounds under investigation. This review discusses the physiological and pathological significance of the ATX-LPA-LPA receptor signaling axis and summarizes the evidence for targeting this pathway for the treatment of CNS diseases.
Collapse
|
29
|
Keep RF, Jones HC, Drewes LR. The year in review: progress in brain barriers and brain fluid research in 2018. Fluids Barriers CNS 2019; 16:4. [PMID: 30717760 PMCID: PMC6362595 DOI: 10.1186/s12987-019-0124-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 12/17/2022] Open
Abstract
This editorial focuses on the progress made in brain barrier and brain fluid research in 2018. It highlights some recent advances in knowledge and techniques, as well as prevalent themes and controversies. Areas covered include: modeling, the brain endothelium, the neurovascular unit, the blood–CSF barrier and CSF, drug delivery, fluid movement within the brain, the impact of disease states, and heterogeneity.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | - Hazel C Jones
- Gagle Brook House, Chesterton, Bicester, OX26 1UF, UK
| | - Lester R Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN, 55812, USA
| |
Collapse
|