1
|
Valeros J, Jerome M, Tseyang T, Vo P, Do T, Fajardo Palomino D, Grotehans N, Kunala M, Jerrett AE, Hathiramani NR, Mireku M, Magesh RY, Yenilmez B, Rosen PC, Mann JL, Myers JW, Kunchok T, Manning TL, Boercker LN, Carr PE, Munim MB, Lewis CA, Sabatini DM, Kelly M, Xie J, Czech MP, Gao G, Shepherd JN, Walker AK, Kim H, Watson EV, Spinelli JB. Rhodoquinone carries electrons in the mammalian electron transport chain. Cell 2025; 188:1084-1099.e27. [PMID: 39909039 PMCID: PMC11845293 DOI: 10.1016/j.cell.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/19/2024] [Accepted: 12/09/2024] [Indexed: 02/07/2025]
Abstract
Ubiquinone (UQ), the only known electron carrier in the mammalian electron transport chain (ETC), preferentially delivers electrons to the terminal electron acceptor oxygen (O2). In hypoxia, ubiquinol (UQH2) diverts these electrons onto fumarate instead. Here, we identify rhodoquinone (RQ), an electron carrier detected in mitochondria purified from certain mouse and human tissues that preferentially delivers electrons to fumarate through the reversal of succinate dehydrogenase, independent of environmental O2 levels. The RQ/fumarate ETC is strictly present in vivo and is undetectable in cultured mammalian cells. Using genetic and pharmacologic tools that reprogram the ETC from the UQ/O2 to the RQ/fumarate pathway, we establish that these distinct ETCs support unique programs of mitochondrial function and that RQ confers protection upon hypoxia exposure in vitro and in vivo. Thus, in discovering the presence of RQ in mammals, we unveil a tractable therapeutic strategy that exploits flexibility in the ETC to ameliorate hypoxia-related conditions.
Collapse
Affiliation(s)
- Jonathan Valeros
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Madison Jerome
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Tenzin Tseyang
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Paula Vo
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Thang Do
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Diana Fajardo Palomino
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nils Grotehans
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Manisha Kunala
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Alexandra E Jerrett
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nicolai R Hathiramani
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA; Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Michael Mireku
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Rayna Y Magesh
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Batuhan Yenilmez
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Paul C Rosen
- Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jessica L Mann
- Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jacob W Myers
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | | | - Tanner L Manning
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Lily N Boercker
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Paige E Carr
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | | | - Caroline A Lewis
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - David M Sabatini
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague, Czech Republic; Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Boston Branch, 840 Memorial Drive, Cambridge, MA 02139, USA
| | - Mark Kelly
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Division of Cardiovascular Medicine, Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Michael P Czech
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Disease Research, UMass Chan Medical School, Worcester, MA 01655, USA; Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jennifer N Shepherd
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Amy K Walker
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Hahn Kim
- Small Molecule Screening Center, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Emma V Watson
- Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jessica B Spinelli
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
2
|
Devi B, Jangid K, Kumar N, Kumar V, Kumar V. Identification of potential JNK3 inhibitors through virtual screening, molecular docking and molecular dynamics simulation as therapeutics for Alzheimer's disease. Mol Divers 2024; 28:4361-4380. [PMID: 38573427 DOI: 10.1007/s11030-024-10820-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/30/2024] [Indexed: 04/05/2024]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder and no effective drug is available for its treatment. Numerous pathological conditions are believed to be responsible for the initiation and development of AD including c-Jun N-terminal kinases (JNKs). The JNKs are one of the enzymes from the mitogen-activated protein kinase (MAPK) family that controls the phosphorylation of various transcription factors on serine and threonine residues, and hold significant responsibilities in tasks like gene expression, cell proliferation, differentiation, and apoptosis. Since, JNK3 is primarily expressed in the brain hence its increased levels in the brain are associated with the AD pathology promoting neurofibrillary tangles, senile plaques, neuroinflammation, and nerve cell apoptosis. The current research work is focused on the development of novel JNK inhibitors as therapeutics for AD employing a structure-based virtual screening (SBVS) approach. The ZINC database (14634052 compounds) was investigated after employing pan assay interference (PAINs), drug-likeness, and diversity picking filter to distinguish molecules interacting with JNK3 by following three docking precision criteria: High Throughput Virtual Screening (HTVS), Standard Precision (SP), and Extra Precision (XP) & MMGBSA. Five lead molecules showed a better docking score in the range of -13.091 to -14.051 kcal/mol better than the reference compound (- 11.828 kcal/mol). The lead compounds displayed acceptable pharmacokinetic properties and were subjected to molecular dynamic simulations of 100 ns and binding free energy calculations. All the lead molecules showed stable RMSD and hydrogen bond interactions throughout the trajectory. The ∆GMM/PBSA_total score for the lead compounds ZINC220382956, ZINC147071339, ZINC207081127, ZINC205151456, ZINC1228819126, and CC-930 was calculated and found to be - 31.39, - 42.8, - 37.04, - 39.01, - 36.5, - 34.16 kcal/mol, respectively. Thus, it was concluded that the lead molecules identified in these studies have the potential to be explored as potent JNK3 inhibitors.
Collapse
Affiliation(s)
- Bharti Devi
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Kailash Jangid
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry and Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Naveen Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Vinay Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India
| | - Vinod Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Ghudda, 151401, India.
| |
Collapse
|
3
|
Yao Z, Lu Y, Wang P, Chen Z, Zhou L, Sang X, Yang Q, Wang K, Hao M, Cao G. The role of JNK signaling pathway in organ fibrosis. J Adv Res 2024:S2090-1232(24)00431-4. [PMID: 39366483 DOI: 10.1016/j.jare.2024.09.029] [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: 07/25/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Fibrosis is a tissue damage repair response caused by multiple pathogenic factors which could occur in almost every apparatus and leading to the tissue structure damage, physiological abnormality, and even organ failure until death. Up to now, there is still no specific drugs or strategies can effectively block or changeover tissue fibrosis. JNKs, a subset of mitogen-activated protein kinases (MAPK), have been reported that participates in various biological processes, such as genetic expression, DNA damage, and cell activation/proliferation/death pathways. Increasing studies indicated that abnormal regulation of JNK signal pathway has strongly associated with tissue fibrosis. AIM OF REVIEW This review designed to sum up the molecular mechanism progresses in the role of JNK signal pathway in organ fibrosis, hoping to provide a novel therapy strategy to tackle tissue fibrosis. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent evidence shows that JNK signaling pathway could modulates inflammation, immunoreaction, oxidative stress and Multiple cell biological functions in organ fibrosis. Therefore, targeting the JNK pathway may be a useful strategy in cure fibrosis.
Collapse
Affiliation(s)
- Zhouhui Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yandan Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Pingping Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Licheng Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xianan Sang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiao Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Kuilong Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Min Hao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Songyang Research Institute of Zhejiang Chinese Medical University, Songyang, 323400, China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
4
|
Cheng C, Zhang Z, Wang J, Wang C, Liu T, Yang C, Wang G, Huang H, Li Y. CircPGM5 regulates Foxo3a phosphorylation via MiR-21-5p/MAPK10 axis to inhibit bladder cancer progression. Cell Signal 2024; 121:111297. [PMID: 39004326 DOI: 10.1016/j.cellsig.2024.111297] [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: 01/28/2024] [Revised: 06/08/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Bladder cancer (BC) is one of the most prevalent malignant tumors worldwide, and the incidence is especially higher in males. Extensive evidence has demonstrated the pivotal role of circular RNAs (circRNAs) in BC progression. However, the exact regulatory mechanism of circRNAs in BC remains incompletely elucidated and warrants further exploration. This study screened a novel circRNA-circPGM5 from thousands of circRNAs by high-throughput sequencing. We found that circPGM5, originating from the PGM5 gene, was significantly lower expressed in BC tissues. Quantitative real-time PCR (qRT-PCR) verified that circPGM5 showed relatively low expression in 50 pairs of BC tissues and EJ and T24 cells. Notably, circPGM5 expression was correlated with stage, grade, and lymphatic metastasis of BC. Through RNA-FISH assay, we confirmed that circPGM5 predominantly localized in the cytoplasm. Functionally, overexpression of circPGM5 inhibited the proliferation, migration, and invasion of BC cells in vitro. Remarkably, circPGM5 demonstrated markedly significant tumor growth and metastasis suppression in vivo. Mechanistically, we discovered that circPGM5 upregulated the mitogen-activated protein kinase 10 (MAPK10) expression by influencing the oncogenic miR-21-5p activity through miR-21-5p absorption. This modulation of MAPK10 impacted the phosphorylation of the tumor suppressor Foxo3a in BC. In conclusion, our findings uncovered the tumor-suppressing role of circPGM5 in BC via the miR-21-5p/MAPK10/Foxo3a axis.
Collapse
Affiliation(s)
- Chao Cheng
- Department of Urology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China; Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China; Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, PR China
| | - Ze Zhang
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Jiawei Wang
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Chong Wang
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Tiantian Liu
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Chenglin Yang
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Guowei Wang
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Houbao Huang
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Yawei Li
- Department of Urology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China.
| |
Collapse
|
5
|
Ueno K, Kurazumi H, Suzuki R, Yanagihara M, Mizoguchi T, Harada T, Morikage N, Hamano K. miR-709 exerts an angiogenic effect through a FGF2 upregulation induced by a GSK3B downregulation. Sci Rep 2024; 14:11372. [PMID: 38762650 PMCID: PMC11102560 DOI: 10.1038/s41598-024-62340-4] [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: 11/15/2022] [Accepted: 05/15/2024] [Indexed: 05/20/2024] Open
Abstract
The aim of this study was to identify angiogenic microRNAs (miRNAs) that could be used in the treatment of hindlimb ischemic tissues. miRNAs contained in extracellular vesicles (EVs) deriving from the plasma were analyzed in C57BL/6 mice, which have ischemia tolerance, and in BALB/c mice without ischemia tolerance as part of a hindlimb ischemia model; as a result 43 angiogenic miRNA candidates were identified. An aortic ring assay was employed by using femoral arteries isolated from BALC/c mice and EVs containing miRNA; as a result, the angiogenic miRNA candidates were limited to 14. The blood flow recovery was assessed after injecting EVs containing miRNA into BALB/c mice with hindlimb ischemia, and miR-709 was identified as a promising angiogenic miRNA. miR-709-encapsulating EVs were found to increase the expression levels of the fibroblast growth factor 2 (FGF2) mRNA in the thigh tissues of hindlimb ischemia model BALB/c mice. miR-709 was also found to bind to the 3'UTR of glycogen synthase kinase 3 beta (GSK3B) in three places. GSK3B-knockdown human artery-derived endothelial cells were found to express high levels of FGF2, and were characterized by increased cell proliferation. These findings indicate that miR-709 induces an upregulation of FGF2 through the downregulation of GSK3B.
Collapse
Affiliation(s)
- Koji Ueno
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan.
- Division of Advanced Cell Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan.
| | - Hiroshi Kurazumi
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Ryo Suzuki
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Masashi Yanagihara
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Takahiro Mizoguchi
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Takasuke Harada
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Graduate School of Medicine, Yamaguchi University, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
- Division of Advanced Cell Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi, Japan
| |
Collapse
|
6
|
Rajan RK, Kumar RP, Ramanathan M. Piceatannol improved cerebral blood flow and attenuated JNK3 and mitochondrial apoptotic pathway in a global ischemic model to produce neuroprotection. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:479-496. [PMID: 37470802 DOI: 10.1007/s00210-023-02616-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Cerebral ischemia is one of the leading causes of death and disability worldwide. The only FDA-approved treatment is recanalization with systemic tissue plasminogen activators like alteplase, although reperfusion caused by recanalization can result in neuroinflammation, which can cause brain cell apoptosis. Therefore, after an ischemic/reperfusion injury, interventions are needed to minimize the neuroinflammatory cascade. In the present study, piceatannol (PCT) was studied for its neuroprotective efficacy in a rat model of global ischemic injury by attenuating c-Jun N-terminal kinase 3 (JNK3) downstream signaling. PCT is a resveratrol analog and a polyphenolic stilbenoid naturally occurring in passion fruit and grapes. The neuroprotective efficacy of PCT (1, 5, 10 mg/kg) in ischemic conditions was assessed through pre- and post-treatment. Cerebral blood flow (CBF) and tests for functional recovery were assessed. Protein and gene expression were done for JNK3 and other inflammatory markers. A docking study was performed to identify the amino acid interaction. The results showed that PCT improved motor and memory function as measured by a functional recovery test believed to be due to an increase in cerebral blood flow. Also, the caspase signaling which promotes apoptosis was found to be down-regulated; however, nitric oxide synthase expression was up-regulated, which could explain the enhanced cerebral blood flow (CBF). According to our findings, PCT impeded c-Jun N-terminal kinase 3 (JNK3) signaling by suppressing phosphorylation and disrupting the mitochondrial apoptotic pathway, which resulted in the neuroprotective effect. Molecular docking analysis was performed to investigate the atomic-level interaction of JNK3 and PCT, which reveals that Met149, Leu206, and Lys93 amino acid residues are critical for the interaction of PCT and JNK3. According to our current research, JNK3 downstream signaling and the mitochondrial apoptosis pathway are both inhibited by PCT, which results in neuroprotection under conditions of global brain ischemia. Piceatannol attenuated JNK3 phosphorylation during the ischemic condition and prevented neuronal apoptosis.
Collapse
Affiliation(s)
- Ravi Kumar Rajan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India.
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Girijananda Chowdhury University, Dekargaon, Tezpur, 784501, Assam, India.
| | - Ram Pravin Kumar
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India
| | - M Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, 641004, Tamilnadu, India
| |
Collapse
|
7
|
Guo L, Yang Q, Wei R, Zhang W, Yin N, Chen Y, Xu C, Li C, Carney RP, Li Y, Feng M. Enhanced pericyte-endothelial interactions through NO-boosted extracellular vesicles drive revascularization in a mouse model of ischemic injury. Nat Commun 2023; 14:7334. [PMID: 37957174 PMCID: PMC10643472 DOI: 10.1038/s41467-023-43153-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Despite improvements in medical and surgical therapies, a significant portion of patients with critical limb ischemia (CLI) are considered as "no option" for revascularization. In this work, a nitric oxide (NO)-boosted and activated nanovesicle regeneration kit (n-BANK) is constructed by decorating stem cell-derived nanoscale extracellular vesicles with NO nanocages. Our results demonstrate that n-BANKs could store NO in endothelial cells for subsequent release upon pericyte recruitment for CLI revascularization. Notably, n-BANKs enable endothelial cells to trigger eNOS activation and form tube-like structures. Subsequently, eNOS-derived NO robustly recruits pericytes to invest nascent endothelial cell tubes, giving rise to mature blood vessels. Consequently, n-BANKs confer complete revascularization in female mice following CLI, and thereby achieve limb preservation and restore the motor function. In light of n-BANK evoking pericyte-endothelial interactions to create functional vascular networks, it features promising therapeutic potential in revascularization to reduce CLI-related amputations, which potentially impact regeneration medicine.
Collapse
Affiliation(s)
- Ling Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China.
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, P. R. China.
| | - Qiang Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Runxiu Wei
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Wenjun Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Na Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Yuling Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Chao Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, P. R. China
| | - Changrui Li
- Guangzhou Zhixin High School, Zhixin South Road, Guangzhou, 510080, P.R. China
| | - Randy P Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, USA.
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, 95616, USA.
| | - Min Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China.
| |
Collapse
|
8
|
Pk N, Rajan RK, Nanchappan V, Karuppaiah A, Chandrasekaran J, Jayaraman S, Gunasekaran V. C-Glucosyl Xanthone derivative Mangiferin downregulates the JNK3 mediated caspase activation in Almal induced neurotoxicity in differentiated SHSY-5Y neuroblastoma cells. Toxicol Mech Methods 2023; 33:707-718. [PMID: 37455591 DOI: 10.1080/15376516.2023.2237106] [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: 01/22/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION C-Glucosyl Xanthone derivatives were assessed to inhibit the JNK3 mediated Caspase pathway in Almal (Aluminum Maltolate) induced neurotoxicity in SHSY-5Y cells. METHODS Mangiferin was selected among 200 C-Glucosyl Xanthones based on molecular interaction, docking score (-10.22 kcal/mol), binding free energy (-71.12 kcal/mol), ADME/tox properties and by molecular dynamic studies. Further, it was noticed that glycone moiety of Mangiferin forms H-bond with ASN 194, SER 193, GLY 76, and OH group in the first position of the aglycone moiety shows interaction at Met 149 which is exceptionally crucial for JNK3 inhibitory activity. RESULTS AND DISCUSSION Mangiferin (0.5, 1, 10, 20 and 30 µM) and standard SP600125 (20 µM) treatment increased the cell survival rate against Almal 200 µM, with EC50 of Mangiferin (8 µM) and standard SP600125 (4.9 µM) respectively. Mangiferin significantly impedes kinase activation, indicating suppression of JNK3 signaling with IC50 (98.26 nM). Mangiferin (10 and 15 µM) dose-dependently inhibits the caspase 3, 8, and 9 enzyme activation in comparison to Almal group. CONCLUSION Mangiferin demonstrated neuroprotection in SHSY-5Y cells against apoptosis induced by Almal by adapting the architecture of the neurons and increasing their density. Among all Xanthone derivatives, Mangiferin could improve neuronal toxicity by inhibiting JNK3 and down-regulating the Caspase activation.
Collapse
Affiliation(s)
- Nafila Pk
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, India
| | - Ravi Kumar Rajan
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Sciences, Tezpur, India
| | | | - Arjunan Karuppaiah
- Department of Pharmaceutics, Karpagam College of Pharmacy, Coimbatore, India
| | - Jaikanth Chandrasekaran
- Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher education and Research (Deemed to be University) Porur, Chennai, India
| | - Saravanan Jayaraman
- JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | |
Collapse
|
9
|
Priego M, Noriega L, Kalinin S, Hoffman LM, Feinstein DL, Morfini G. Genetic deletion of c-Jun amino-terminal kinase 3 (JNK3) modestly increases disease severity in a mouse model of multiple sclerosis. J Neuroimmunol 2023; 382:578152. [PMID: 37454525 PMCID: PMC10527920 DOI: 10.1016/j.jneuroim.2023.578152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/01/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
The c-Jun amino terminal kinases (JNKs) regulate transcription, and studies suggest they contribute to neuropathology in the EAE model of MS. To examine the role of the JNK3 isoform, we compared EAE in JNK3 null mice to wild type (WT) littermates. Although disease severity was similar in female mice, in male JNK3 null mice the day of onset and time to reach 100% incidence occurred sooner, and disease severity was increased. While glial activation in spinal cord was similar, white matter lesions were increased in JNK3 null mice. These results suggest JNK3 normally limits EAE disease in a sex-dependent manner.
Collapse
Affiliation(s)
- Mercedes Priego
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612, United States of America
| | - Lorena Noriega
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612, United States of America
| | - Sergey Kalinin
- Department of Research, Jesse Brown VA Medical Center, Chicago, IL 60612, United States of America
| | - Lisa M Hoffman
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612, United States of America
| | - Douglas L Feinstein
- Department of Research, Jesse Brown VA Medical Center, Chicago, IL 60612, United States of America; Department of Anesthesiology, University of Illinois, Chicago, IL 60612, United States of America.
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612, United States of America.
| |
Collapse
|
10
|
Zeemering S, Isaacs A, Winters J, Maesen B, Bidar E, Dimopoulou C, Guasch E, Batlle M, Haase D, Hatem SN, Kara M, Kääb S, Mont L, Sinner MF, Wakili R, Maessen J, Crijns HJGM, Fabritz L, Kirchhof P, Stoll M, Schotten U. Atrial fibrillation in the presence and absence of heart failure enhances expression of genes involved in cardiomyocyte structure, conduction properties, fibrosis, inflammation, and endothelial dysfunction. Heart Rhythm 2022; 19:2115-2124. [PMID: 36007727 DOI: 10.1016/j.hrthm.2022.08.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 07/29/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Little is known about genome-wide changes in the atrial transcriptome as a cause or consequence of atrial fibrillation (AF), and the effect of its common and clinically relevant comorbidity-heart failure (HF). OBJECTIVE The purpose of this study was to explore candidate disease processes for AF by investigating gene expression changes in atrial tissue samples from patients with and without AF, stratified by HF. METHODS RNA sequencing was performed in right and left atrial appendage tissue in 195 patients undergoing open heart surgery from centers participating in the CATCH-ME consortium (no history of AF, n = 91; paroxysmal AF, n = 53; persistent/permanent AF, n = 51). Analyses were stratified into patients with/without HF (n = 75/120) and adjusted for age, sex, atrial side, and a combination of clinical characteristics. RESULTS We identified 35 genes associated with persistent AF compared to patients without a history of AF, both in the presence or absence of HF (false discovery rate <0.05). These were mostly novel associations, including 13 long noncoding RNAs. Genes were involved in regulation of cardiomyocyte structure, conduction properties, fibrosis, inflammation, and endothelial dysfunction. Gene set enrichment analysis identified mainly inflammatory gene sets to be enriched in AF patients without HF, and gene sets involved in cellular respiration in AF patients with HF. CONCLUSION Analysis of atrial gene expression profiles identified numerous novel genes associated with persistent AF, in the presence or absence of HF. Interestingly, no consistent transcriptional changes were associated with paroxysmal AF, suggesting that AF-induced changes in gene expression predominate other changes.
Collapse
Affiliation(s)
- Stef Zeemering
- Department of Physiology, Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, the Netherlands
| | - Aaron Isaacs
- Department of Physiology, Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Joris Winters
- Department of Physiology, Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, the Netherlands
| | - Bart Maesen
- Department of Cardiothoracic Surgery, Maastricht University Medical Centre, University Maastricht, Maastricht, the Netherlands
| | - Elham Bidar
- Department of Cardiothoracic Surgery, Maastricht University Medical Centre, University Maastricht, Maastricht, the Netherlands
| | | | - Eduard Guasch
- Cardiovascular Institute, Hospital Clinic Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; CIBERCV, Madrid, Spain
| | - Montserrat Batlle
- Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; CIBERCV, Madrid, Spain
| | | | - Stéphane N Hatem
- INSERM UMRS1166, Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France; Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Mansour Kara
- Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Stefan Kääb
- Department of Medicine I, University Hospital, Munich, Germany; German Centre for Cardiovascular Research, partner site Munich Heart, Munich, Germany
| | - Lluis Mont
- European Society of Cardiology, Sophia Antipolis, France; Cardiovascular Institute, Hospital Clinic Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; CIBERCV, Madrid, Spain
| | - Moritz F Sinner
- Department of Medicine I, University Hospital, Munich, Germany; German Centre for Cardiovascular Research, partner site Munich Heart, Munich, Germany
| | - Reza Wakili
- German Centre for Cardiovascular Research, partner site Munich Heart, Munich, Germany; Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, Essen, Germany
| | - Jos Maessen
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Harry J G M Crijns
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Larissa Fabritz
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Cardiology, UHB and SWBH NHS Trusts, Birmingham, United Kingdom
| | - Paulus Kirchhof
- INSERM UMRS1166, Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France; Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; University Heart and Vascular Center UKE Hamburg, Hamburg, Germany; German Center for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Germany
| | - Monika Stoll
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands; Institute of Human Genetics, University of Muenster, Muenster, Germany
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, University Maastricht, Maastricht, the Netherlands; INSERM UMRS1166, Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France.
| |
Collapse
|
11
|
Niu R, Wang L, Yang W, Sun L, Tao J, Sun H, Mei S, Wang W, Feng K, Qian D, Bai X. MicroRNA-582-5p targeting Creb1 modulates apoptosis in cardiomyocytes hypoxia/reperfusion-induced injury. Immun Inflamm Dis 2022; 10:e708. [PMID: 36301033 PMCID: PMC9601879 DOI: 10.1002/iid3.708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/10/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) caused by the reperfusion therapy of myocardial ischemic diseases is a kind of major disease that threatens human health and lives severely. There are lacking of effective therapeutic measures for MIRI. MicroRNAs (miRNAs) are abundant in mammalian species and play a critical role in the initiation, promotion, and progression of MIRI. However, the biological role and molecular mechanism of miRNAs in MIRI are not entirely clear. METHODS We used bioinformatics analysis to uncover the significantly different miRNA by analyzing transcriptome sequencing data from myocardial tissue in the mouse MIRI model. Multiple miRNA-related databases, including miRdb, PicTar, and TargetScan were used to forecast the downstream target genes of the differentially expressed miRNA. Then, the experimental models, including male C57BL/6J mice and HL-1 cell line, were used for subsequent experiments including quantitative real-time polymerase chain reaction analysis, western blot analysis, hematoxylin and eosin staining, flow cytometry, luciferase assay, gene interference, and overexpression. RESULTS MiR-582-5p was found to be differentially upregulated from the transcriptome sequencing data. The elevated levels of miR-582-5p were verified in MIRI mice and hypoxia/reperfusion (H/R)-induced HL-1 cells. Functional experiments revealed that miR-582-5p promoted apoptosis of H/R-induced HL-1 cells via downregulating cAMP-response element-binding protein 1 (Creb1). The inhibiting action of miR-582-5p inhibitor on H/R-induced apoptosis was partially reversed after Creb1 interference. CONCLUSIONS Collectively, the research findings reported that upregulation of miR-582-5p promoted H/R-induced cardiomyocyte apoptosis by inhibiting Creb1. The potential diagnostic and therapeutic strategies targeting miR-582-5p and Creb1 could be beneficial for the MIRI treatment.
Collapse
Affiliation(s)
- Rui‐Ze Niu
- Department of Cardiac SurgeryKunming Medical University First Affiliated HospitalKunmingYunnanChina
- Department of Animal ZoologyKunming Medical UniversityKunmingYunnanChina
| | - Lu‐Qiao Wang
- Department of CardiologyKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Wei Yang
- Department of AnesthesiologyKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Li‐Zhong Sun
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel DiseasesCapital Medical UniversityBeijingChina
| | - Jie Tao
- Department of Cardiac SurgeryKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Huang Sun
- Department of CardiologyKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Song Mei
- Department of Cardiac SurgeryKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Wen‐Jie Wang
- Department of Cardiac SurgeryKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Ke‐Xiang Feng
- Department of Cardiac SurgeryKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Dian‐Lun Qian
- Department of Cardiac SurgeryKunming Medical University First Affiliated HospitalKunmingYunnanChina
| | - Xiang‐Feng Bai
- Department of Cardiac SurgeryKunming Medical University First Affiliated HospitalKunmingYunnanChina
| |
Collapse
|
12
|
Choi J, Kim YS, Kim MH, Kim HJ, Yoon BE. Maternal lead exposure induces sex-dependent cerebellar glial alterations and repetitive behaviors. Front Cell Neurosci 2022; 16:954807. [PMID: 36072563 PMCID: PMC9442054 DOI: 10.3389/fncel.2022.954807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Lead (Pb) is one of the most prevalent heavy metals we encounter daily. Although there are many reports regarding their toxic effects on humans, the effects of exposure to low lead concentrations throughout the pregnancy period on the offspring are not fully elucidated yet. This study aimed to investigate the cellular mechanisms that occur in response to lead exposure. To this end, we administered lead-containing water to pregnant mice from the day of conception till delivery or till day 28 postnatally. Furthermore, we performed neurodevelopmental disorder-related behavior tests and RNA-sequencing analysis. We used both genders for all experiments because neurodevelopmental disorders usually show several sex-dependent differences. The results revealed increased levels of gliosis in the cerebella of lead-exposed pups compared to those in littermates belonging to the control group. Additionally, we observed altered behaviors of male mice in the autism spectrum disorder-related tests. RNA-sequencing results revealed changes in gamma-aminobutyric acid (GABA) signaling in the lead-exposed mouse model. Specifically, the lead-exposed male mice showed decreased monoamine oxidase B and increased levels of diamine oxidase enzyme, which is related to the synthesis of GABA in astrocytes. These findings demonstrate sex-dependent basal developmental changes in glial cells and an increased prevalence of autistic-like behaviors in the young pups of mothers exposed to lead during pregnancy.
Collapse
Affiliation(s)
- Juwon Choi
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
| | - Mi-Hye Kim
- Department of Physiology, College of Medicine, Dankook University, Cheonan, South Korea
- Department of Medical Laser, Graduate School, Dankook University, Cheonan, South Korea
| | - Hee Jung Kim
- Department of Physiology, College of Medicine, Dankook University, Cheonan, South Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
- *Correspondence: Bo-Eun Yoon,
| |
Collapse
|
13
|
Yu P, Zhou J, Ge C, Fang M, Zhang Y, Wang H. Differential expression of placental 11β-HSD2 induced by high maternal glucocorticoid exposure mediates sex differences in placental and fetal development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 827:154396. [PMID: 35259391 DOI: 10.1016/j.scitotenv.2022.154396] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/20/2022] [Accepted: 03/04/2022] [Indexed: 06/14/2023]
Abstract
A variety of adverse environmental factors during pregnancy cause maternal chronic stress. Caffeine is a common stressor, and its consumption during pregnancy is widespread. Our previous study showed that prenatal caffeine exposure (PCE) increased maternal blood glucocorticoid levels and caused abnormal development of offspring. However, the placental mechanism for fetal development inhibition caused by PCE-induced high maternal glucocorticoid has not been reported. This study investigated the effects of PCE-induced high maternal glucocorticoid level on placental and fetal development by regulating placental 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) expression and its underlying mechanism. First, human placenta and umbilical cord blood samples were collected from women without prenatal use of synthetic glucocorticoids. We found that placental 11β-HSD2 expression was significantly correlated with umbilical cord blood cortisol level and birth weight in male newborns but not in females. Furthermore, we established a rat model of high maternal glucocorticoids induced by PCE (caffeine, 60 mg/kg·d, ig), and found that the expression of 11β-HSD2 in male PCE placenta was decreased and negatively correlated with the maternal/fetal/placental corticosterone levels. Meanwhile, we found abnormal placental structure and nutrient transporter expression. In vitro, BeWo cells were used and confirm that 11β-HSD2 mediated inhibition of placental nutrient transporter expression induced by high levels of glucocorticoid. Finally, combined with the animal and cell experiments, we further confirmed that high maternal glucocorticoid could activate the GR-C/EBPα-Egr1 signaling pathway, leading to decreased expression of 11β-HSD2 in males. However, there was no significant inhibition of placental 11β-HSD2 expression, placental and fetal development in females. In summary, we confirmed that high maternal glucocorticoids could regulate placental 11β-HSD2 expression in a sex-specific manner, leading to differences in placental and fetal development. This study provides the theoretical and experimental basis for analyzing the inhibition of fetoplacental development and its sex difference caused by maternal stress.
Collapse
Affiliation(s)
- Pengxia Yu
- Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Jin Zhou
- Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Caiyun Ge
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Man Fang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China
| | - Yuanzhen Zhang
- Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, China; Department of Obstetrics and Gynaecology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
14
|
Kamel R, El Morsy EM, Elsherbiny ME, Nour-Eldin M. Chrysin promotes angiogenesis in rat hindlimb ischemia: Impact on PI3K/Akt/mTOR signaling pathway and autophagy. Drug Dev Res 2022; 83:1226-1237. [PMID: 35662099 DOI: 10.1002/ddr.21954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/01/2022] [Accepted: 05/17/2022] [Indexed: 11/09/2022]
Abstract
Limb ischemia occurs due to obstruction of blood perfusion to lower limbs, a manifestation that is associated with peripheral artery disease (PAD). Angiogenesis is important for adequate oxygen delivery. The present study investigated a potential role for chrysin, a naturally occurring flavonoid, in promoting angiogenesis in hindlimb ischemia (HLI) rat model. Rats were allocated into four groups: (1) sham-operated control, (2) HLI: subjected to unilateral femoral artery ligation, (3) HLI + chrysin: received 100 mg/kg, i.p. chrysin immediately after HLI, and (4) HLI + chrysin + rapamycin: received 6 mg/kg/day rapamycin i.p. for 5 days then subjected to HLI and dosed with 100 mg/kg chrysin, i.p. Rats were killed 18 h later and gastrocnemius muscles were collected and divided into parts for (1) immunohistochemistry detection of CD31 and CD105, (2) qRT-PCR analysis of eNOS and VEGFR2, (3) colorimetric analysis of NO, (4) ELISA estimation of TGF-β, VEGF, ATG5 and Beclin-1, and (5) Western blot analysis of p-PI3K, PI3K, p-Akt, Akt, p-mTOR, mTOR, and HIF-1α. Chrysin significantly enhanced microvessels growth in HLI muscles as indicated by increased CD31 and CD105 levels and decreased TGF-β. Chrysin's proangiogenic effect is potentially mediated by increased VEGF, VEGFR2 and activation of PI3K/AKT/mTOR pathway, which promoted eNOS and NO levels as it was reversed by the mTOR inhibitor, rapamycin. Chrysin also inhibited autophagy as it decreased ATG5 and Beclin-1. The current study shows that chrysin possesses a proangiogenic effect in HLI rats and might be useful in patients with PAD.
Collapse
Affiliation(s)
- Rehab Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Engy M El Morsy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Marwa E Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Mahmoud Nour-Eldin
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), Menoufia, Egypt
| |
Collapse
|
15
|
Luo Z, Hu Q, Tang Y, Leng Y, Tian T, Tian S, Huang C, Liu A, Deng X, Shen L. Construction and investigation of β3GNT2-associated regulatory network in esophageal carcinoma. Cell Mol Biol Lett 2022; 27:8. [PMID: 35073841 PMCID: PMC8903709 DOI: 10.1186/s11658-022-00306-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/04/2022] [Indexed: 12/24/2022] Open
Abstract
Background Glycosyltransferases play a crucial role in various cancers. β1, 3-N-acetylglucosaminyltransferase 2, a polylactosamine synthase, is an important member of the glycosyltransferase family. However, the biological function and regulatory mechanism of β3GNT2 in esophageal carcinoma (ESCA) is still poorly understood. Methods The Cancer Genome Atlas and Genotype-Tissue Expression databases were used for gene expression and prognosis analysis. Quantitative real-time PCR, Western blot, and immunohistochemistry were performed to detect the expression of β3GNT2 in ESCA cell lines and tissues. In vitro assays and xenograft tumor models were utilized to evaluate the impact of β3GNT2 on ESCA progression. The downstream effectors and upstream regulators of β3GNT2 were predicted by online software and verified by functional experiments. Results We found that β3GNT2 was highly expressed in ESCA tissues and positively correlated with poor prognosis in ESCA patients. β3GNT2 expression was closely associated with the tumor size, TNM stage, and overall survival of ESCA patients. Functionally, β3GNT2 promoted ESCA cell growth, migration, and invasion in vitro, as well as tumorigenesis in vivo. Mechanistically, β3GNT2 knockdown decreased the expression of the polylactosamine on EGFR. Knockdown of β3GNT2 also inhibited the JAK/STAT signaling pathway. Meanwhile, the JAK/STAT inhibitor could partly reverse the biological effects caused by β3GNT2 overexpression. Moreover, β3GNT2 expression was positively regulated by CREB1 and negatively regulated by miR-133b. Both CREB1 and miR-133b was involved in the β3GNT2-mediated ESCA progression. Conclusions Our study, for the first time, reveals the importance of β3GNT2 in ESCA progression and offers a potential therapeutic target for ESCA. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00306-y.
Collapse
|
16
|
Ren Q, Pan J, Chen Y, Shen Z, Yang Z, Kwon K, Guo Y, Wang Y, Ji F. Melatonin-Medicated Neural JNK3 Up-Regulation Promotes Ameloblastic Mineralization. Front Cell Dev Biol 2022; 9:749642. [PMID: 35004671 PMCID: PMC8740296 DOI: 10.3389/fcell.2021.749642] [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: 08/12/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Melatonin, an endogenous neurohormone, modulates the biological circadian rhythms of vertebrates. It functions have been reported in previous stomatological studies as anti-inflammation, antioxidant, osseointegration of dental implants and stimulation to dental pulp stem cells differentiation, but its role in ameloblastic differentiation and mineralization has been rarely studied. Objective: To reveal the effects of melatonin on the mineralization of ameloblast lineage cells (ALCs), and to identify the change in gene expression and the potential mechanism based on ribonucleic acid sequencing (RNA-seq) analysis. Method: ALCs were induced in melatonin-conditioned medium. After 7-days culture, Western blot, real-time PCR, alkaline phosphatase (ALP) activity test, RNA-seq were accordingly used to detect the change in molecular level. After 1-month odontogenic induction in melatonin medium, Alizarin Red-S (ARS) staining showed the changes of mineral nodules. Differentially expressed genes (DEGs), enrichment of functions and signaling pathways analysis based on Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) database were performed. The JNK3 antagonist (JNK3 inhibitor IX, SR3576) and β-arrestin1 (Arrb1) overexpression were applied to confirm the fluctuation of melatonin-medicated JNK3 and Arrb1 expression. Results: In this study, we found out melatonin contributed to the ameloblastic mineralization, from which we can observed the elevated expression of enamel matrix protein, and increased ALP activity and mineralized nodules formation. RNA-seq analysis showed the up-regulation of neural JNK3 and down-regulation of Arrb1 in ALCs. Meanwhile, phosphorylated JNK3 deficiency (phosphorylated JNK3 inhibitor---SR3576 added to culture medium) led to mineralization delay, and Arrb1 overexpression proved Arrb1 takes bridge between melatonin receptors (MTNR) and JNK3 in MAPK signaling pathway.
Collapse
Affiliation(s)
- Qianhui Ren
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Pan
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yunshuo Chen
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital, Shanghai Institute of Hematology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhecheng Shen
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhao Yang
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Kubin Kwon
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Guo
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yueying Wang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital, Shanghai Institute of Hematology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Ji
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Wang Q, Xie Y, He Q, Geng Y, Xu J. LncRNA-Cox2 regulates macrophage polarization and inflammatory response through the CREB-C/EBPβ signaling pathway in septic mice. Int Immunopharmacol 2021; 101:108347. [PMID: 34799287 DOI: 10.1016/j.intimp.2021.108347] [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: 03/04/2021] [Revised: 07/09/2021] [Accepted: 11/03/2021] [Indexed: 12/20/2022]
Abstract
LncRNA-Cox2 has been reported to regulate macrophage polarization, and the activation of macrophages is a major participant in the pathogenesis of sepsis. Therefore, we explored whether lncRNA-Cox2 was involved in the progression of sepsis. In this study, we established a cecal ligation and puncture (CLP) mouse model and found that silencing lncRNA-Cox2 in CLP mice improved the 7-day survival rate, and alleviated the increase of blood bacterial burdens, systemic inflammatory response, and pulmonary dysfunction induced by CLP. Besides, interference with lncRNA-Cox2 declined the percentage of M1 macrophages and increased the percentage of M2 macrophages in the spleens of CLP mice. In vitro, the knockdown of lncRNA-Cox2 suppressed LPS-induced inflammation and M1 macrophage marker expression, and promoted M2 macrophage marker expression in primary peritoneal macrophages and RAW264.7 cells. Moreover, lncRNA-Cox2 induced CREB phosphorylation by binding to CREB, and increased phosphorylated-CREB enrichment in the C/EBPβ promoter region, so as to promote C/EBPβ transcription, thereby activating the CREB-C/EBPβ cascade. In addition, overexpressing lncRNA-Cox2 enhanced the effect of LPS on inflammation and macrophage polarization, which was reversed by treatment with 666-15 (an inhibitor of CREB). In conclusion, silencing lncRNA-Cox2 restrained the progression of sepsis in mice by modulating macrophage polarization and inflammatory response through suppressing CREB-C/EBPβ pathway.
Collapse
Affiliation(s)
- Qi Wang
- Department of Microbiology and Immunology, School of Medicine, Xi'an Jiaotong University, Xi'an, China; Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Xie
- Medical Laboratory Center, Northwest Women's and Children's Hospital Affiliated to Xi'an Jiaotong University, Xi'an, China
| | - Qian He
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Geng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiru Xu
- Department of Microbiology and Immunology, School of Health Science Center, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
18
|
Santiago FS, Li Y, Khachigian LM. Serine 26 in Early Growth Response-1 Is Critical for Endothelial Proliferation, Migration, and Network Formation. J Am Heart Assoc 2021; 10:e020521. [PMID: 34476983 PMCID: PMC8649526 DOI: 10.1161/jaha.120.020521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Vascular endothelial cell proliferation, migration, and network formation are key proangiogenic processes involving the prototypic immediate early gene product, Egr‐1 (early growth response‐1). Egr‐1 undergoes phosphorylation at a conserved Ser26 but its function is completely unknown in endothelial cells or any other cell type. Methods and Results A CRISPR/Cas9 strategy was used to introduce a homozygous Ser26>Ala mutation into endogenous Egr‐1 in human microvascular endothelial cells. In the course of generating mutant cells, we produced cells with homozygous deletion in Egr‐1 caused by frameshift and premature termination. We found that Ser26 mutation in Egr‐1, or Egr‐1 deletion, perturbed endothelial cell proliferation in models of cell counting or real‐time growth using the xCELLigence System. We found that Ser26 mutation or Egr‐1 deletion ameliorated endothelial cell migration toward VEGF‐A165 (vascular endothelial growth factor‐A) in a dual‐chamber model. On solubilized basement membrane preparations, Ser26 mutation or Egr‐1 deletion prevented endothelial network (or tubule) formation, an in vitro model of angiogenesis. Flow cytometry further revealed that Ser26 mutation or Egr‐1 deletion elevated early and late apoptosis. Finally, we demonstrated that Ser26 mutation or Egr‐1 deletion increased VE‐cadherin (vascular endothelial cadherin) expression, a regulator of endothelial adhesion and signaling, permeability, and angiogenesis. Conclusions These findings not only indicate that Egr‐1 is essential for endothelial cell proliferation, migration, and network formation, but also show that point mutation in Ser26 is sufficient to impair each of these processes and trigger apoptosis as effectively as the absence of Egr‐1. This highlights the importance of Ser26 in Egr‐1 for a range of proangiogenic processes.
Collapse
Affiliation(s)
- Fernando S Santiago
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| | - Yue Li
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research School of Medical Sciences UNSW Medicine and HealthUniversity of New South Wales Sydney NSW Australia
| |
Collapse
|
19
|
Li H, Li Y, Zhang Y, Tan B, Huang T, Xiong J, Tan X, Ermolaeva MA, Fu L. MAPK10 Expression as a Prognostic Marker of the Immunosuppressive Tumor Microenvironment in Human Hepatocellular Carcinoma. Front Oncol 2021; 11:687371. [PMID: 34408980 PMCID: PMC8366563 DOI: 10.3389/fonc.2021.687371] [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: 03/29/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains a devastating malignancy worldwide due to lack of effective therapy. The immune-rich contexture of HCC tumor microenvironment (TME) makes this tumor an appealing target for immune-based therapies; however, the immunosuppressive TME is still a major challenge for more efficient immunotherapy in HCC. Using bioinformatics analysis based on the TCGA database, here we found that MAPK10 is frequently down-regulated in HCC tumors and significantly correlates with poor survival of HCC patients. HCC patients with low MAPK10 expression have lower expression scores of tumor infiltration lymphocytes (TILs) and stromal cells in the TME and increased scores of tumor cells than those with high MAPK10 expression. Further transcriptomic analyses revealed that the immune activity in the TME of HCC was markedly reduced in the low-MAPK10 group of HCC patients compared to the high-MAPK10 group. Additionally, we identified 495 differentially expressed immune-associated genes (DIGs), with 482 genes down-regulated and 13 genes up-regulated in parallel with the decrease of MAPK10 expression. GO enrichment and KEGG pathway analyses indicated that the biological functions of these DIGs included cell chemotaxis, leukocyte migration and positive regulation of the response to cytokine–cytokine receptor interaction, T cell receptor activation and MAPK signaling pathway. Protein–protein interaction (PPI) analyses of the 495 DIGs revealed five potential downstream hub genes of MAPK10, including SYK, CBL, VAV1, LCK, and CD3G. Several hub genes such as SYK, LCK, and VAV1 could respond to the immunological costimulatory signaling mediated by the transmembrane protein ICAM1, which was identified as a down-regulated DIG associated with low-MAPK10 expression. Moreover, ectopic overexpression or knock-down of MAPK10 could up-regulate or down-regulate ICAM1 expression via phosphorylation of c-jun at Ser63 in HCC cell lines, respectively. Collectively, our results demonstrated that MAPK10 down-regulation likely contributes to the immunosuppressive TME of HCC, and this gene might serve as a potential immunotherapeutic target and a prognostic factor for HCC patients.
Collapse
Affiliation(s)
- Huahui Li
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China.,Group of Homeostasis and Stress Tolerance, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany.,Shenzhen University-Friedrich Schiller Universitat Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuting Li
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China.,Group of Homeostasis and Stress Tolerance, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany.,Shenzhen University-Friedrich Schiller Universitat Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, Shenzhen, China
| | - Ying Zhang
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Binbin Tan
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Tuxiong Huang
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Jixian Xiong
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Xiangyu Tan
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Maria A Ermolaeva
- Group of Homeostasis and Stress Tolerance, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Li Fu
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
20
|
Cheng C, Wang X, Jiang Y, Li Y, Liao Z, Li W, Yu Z, Whalen MJ, Lok J, Dumont AS, Liu N, Wang X. Recombinant Annexin A2 Administration Improves Neurological Outcomes After Traumatic Brain Injury in Mice. Front Pharmacol 2021; 12:708469. [PMID: 34400908 PMCID: PMC8363504 DOI: 10.3389/fphar.2021.708469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Microvascular failure is one of the key pathogenic factors in the dynamic pathological evolution after traumatic brain injury (TBI). Our laboratory and others previously reported that Annexin A2 functions in blood-brain barrier (BBB) development and cerebral angiogenesis, and recombinant human Annexin A2 (rA2) protected against hypoxia plus IL-1β-induced cerebral trans-endothelial permeability in vitro, and cerebral angiogenesis impairment of AXNA2 knock-out mice in vivo. We thereby hypothesized that ANXA2 might be a cerebrovascular therapy candidate that targets early BBB integrity disruption, and subacute/delayed cerebrovascular remodeling after TBI, ultimately improve neurological outcomes. In a controlled cortex impact (CCI) mice model, we found rA2 treatment (1 mg/kg) significantly reduced early BBB disruption at 24 h after TBI; and rA2 daily treatment for 7 days augmented TBI-induced mRNA levels of pro-angiogenic and endothelial-derived trophic factors in cerebral microvessels. In cultured human brain microvascular endothelial cells (HBMEC), through MAPKs array, we identified that rA2 significantly activated Akt, ERK, and CREB, and the activated CREB might be responsible for the rA2-induced VEGF and BDNF expression. Moreover, rA2 administration significantly increased cerebral angiogenesis examined at 14 days and vessel density at 28 days after TBI in mice. Consistently, our results validated that rA2 significantly induced angiogenesis in vitro, evidenced by tube formation and scratched migration assays in HBMEC. Lastly, we demonstrated that rA2 improved long-term sensorimotor and cognitive function, and reduced brain tissue loss at 28 days after TBI. Our findings suggest that rA2 might be a novel vascular targeting approach for treating TBI.
Collapse
Affiliation(s)
- Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Xiaoshu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Yinghua Jiang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Yadan Li
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Zhengbu Liao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Wenlu Li
- Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Zhanyang Yu
- Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Michael J Whalen
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Josephine Lok
- Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States.,Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ning Liu
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
21
|
Chen J, Liang ZQ, Hu C, Gao Y, Wang YK, Yang JW, Zhao C, Cao YM, Cao YB. Protection against peripheral artery disease injury by Ruan Jian Qing Mai formula via metabolic programming. Biotechnol Appl Biochem 2021; 68:366-380. [PMID: 32374895 DOI: 10.1002/bab.1934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022]
Abstract
Ruan Jian Qing Mai formula (RJQM), a multicomponent herbal formula, has been widely used to treat peripheral arterial disease (PAD) in China. However, its active compounds and mechanisms of action are still unknown. First, RNA sequencing analysis of 15 healthy and 16 PAD samples showed that 524 PAD differential genes were significantly enriched in Go Ontology (ribonucleotide metabolic process, oxidoreductase complex, and electron transfer activity), Kyoto Encyclopedia of Genes and Genomes (KEGG) and GSEA pathways (OXPHOS and TCA cycle), miRNA (MIR183), and kinase (PAK6). Fifty-three active ingredients in RJQM had similar structures to the seven drug molecules in CLUE. Then, network topology analysis of the 53 components-target-pathway-disease network yielded 10 active ingredients. Finally, computational toxicity estimations showed that the median lethal dose (LD50) of the 10 active ingredients was above 1000 mg/kg, and eight of them did not cause hepatotoxicity, mutagenicity, carcinogenicity, cytotoxicity, and immunotoxicity nor activate 12 toxic pathways. In conclusion, RJQM has a protection effect on PAD by regulating a complex molecular network. Part of the mechanism is associated with the regulation of OXPHOS by 10 active components, which may alleviate mitochondrial dysfunction and pathological metabolic programming.
Collapse
Affiliation(s)
- Jian Chen
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Academy of Traditional Chinese Medicine, Institute of Vascular Anomalies, Shanghai, China
| | - Zhi-Qiang Liang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Can Hu
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Gao
- Traditional Chinese Recovery and Treatment Center, Zhejiang Rehabilitation Medical Center, Hangzhou, China
| | - Yong-Kui Wang
- The Department of Orthopaedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiang-Wei Yang
- Mental Health Center, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Cheng Zhao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Academy of Traditional Chinese Medicine, Institute of Vascular Anomalies, Shanghai, China
| | - Ye-Min Cao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Academy of Traditional Chinese Medicine, Institute of Vascular Anomalies, Shanghai, China
| | - Yong-Bing Cao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Academy of Traditional Chinese Medicine, Institute of Vascular Anomalies, Shanghai, China
| |
Collapse
|
22
|
Xie L, Wang Y, Chen Z. Early Growth Response Protein 1 Knockdown Alleviates the Cerebral Injury in Rats with Intracerebral Hemorrhage (ICH) via STAT3/NF-κB Pathway by Reducing RXRα Acetylation Level. Neuroscience 2021; 487:120-130. [PMID: 33600884 DOI: 10.1016/j.neuroscience.2021.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 11/25/2022]
Abstract
Brain EGR1 (early growth response protein 1) overexpression aggravates focal ischemic brain injury, but its role in intracerebral hemorrhage (ICH) induced cerebral injury remains obscure. In this study, a rat ICH model was established by injecting type VII collagenase into the brain, and EGR1 knockdown reversed the increase of hematoma area, neurological function score, brain water content, blood-brain barrier (BBB) permeability, inflammation, p300 and retinoid a X receptor-α (RXRα) protein levels, as well as RXRα acetylation level induced by ICH. EGR1 expression was up-regulated in primary brain microvascular endothelial cells (BMECs), neurons, and astrocytes after ICH induction, and the up-regulation was most significant in BMECs. We also found that EGR1 promoted RXRα acetylation level by regulating p300 in BMECs. Subsequently, the BMECs were treated with OGD (oxygen glucose deprivation) plus hemin to simulate ICH condition. And silencing EGR1 rescued the upregulation of cell inflammation and the reduction of cell viability and TEER (transendothelial electric resistance) caused by OGD plus hemin via p300-mediated RXRα acetylation. Furthermore, the STAT3/NF-κB pathway was activated after treatment with OGD plus hemin, which was suppressed by silencing EGR1. And treatment with Stattic (an inhibitor of STAT3) restrained the effect of OGD plus hemin on NF-κB pathway activity, inflammation, cell viability and TEER. In conclusion, EGR1 increased RXRα acetylation level by regulating p300, thereby aggravating brain damage in ICH rat model and dysfunction in BMECs, which might through the STAT3/NF-κB pathway.
Collapse
Affiliation(s)
- Lijuan Xie
- Department of Vascular Surgery, China-Japan Friendship Hospital, Jilin University, China
| | - Yingying Wang
- Ward 4 of Neurology Department, China-Japan Friendship Hospital, Jilin University, China
| | - Zhuo Chen
- Ward 1 of Neurosurgery Department, China-Japan Friendship Hospital, Jilin University, China.
| |
Collapse
|
23
|
Systematical Identification of the Protective Effect of Danhong Injection and BuChang NaoXinTong Capsules on Transcription Factors in Cerebral Ischemia Mice Brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2020:5879852. [PMID: 33414894 PMCID: PMC7755463 DOI: 10.1155/2020/5879852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/20/2020] [Accepted: 11/17/2020] [Indexed: 11/17/2022]
Abstract
Cerebral ischemia has led to a high rate of both disability and mortality with massive healthcare costs. Although transcriptional regulation is typically mediated by different combinations of TFs, a combined regulatory unit to synergistically activate transcription has remained unclear in cerebral ischemia, especially in different drug treatments. In this study, TFs alterations after 6 h cerebral ischemic injury and repair were performed by a concatenated tandem array of consensus transcription factor response elements (catTFREs), and vital TFs were obtained by TFs-target imbalanced network. Drug intervention used Danhong injection (DHI) and BNC (BuChang NaoXinTong Capsules), which has been widely prescribed in Chinese herb medicine for the treatment of cerebrovascular and cardiovascular diseases. There were 198 TFs identified after 6 h MCAO operation, and six TFs (Sox2, Smad3, FoxO1, Creb1, Egr,1 and Smad4) were considered as critical TFs in response to cerebral ischemia. Moreover, Smad3 was identified as a hub TF among six vital TFs, and the transcription activity of Smad3 was further verified. These 6 TFs were all reversed by DHI or BNC, indicating different medications may regulate different transcription factors through TF synergy. Moreover, validation results indicated that Smad3 was a putative target TF for DHI and BNC-mediated protection against cerebral ischemia. The observations of the present study provide a fresh understanding of biomolecules and possible new avenues for therapeutic interventions, in addition to the new intervention pattern for different treatments for ischemia stroke.
Collapse
|
24
|
Hu S, Chen Y, Liu L, Yin X, Yang Y, Tang L. PLZF and PLZF-MAPK10 can predict the prognosis of postoperative patients with hepatocellular carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:3158-3166. [PMID: 33425116 PMCID: PMC7791377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/28/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE The current study aimed to explore the expression level of promyelocytic leukaemia zinc finger (PLZF) in hepatocellular carcinoma tissues and to investigate the value of detecting the expression levels of PLZF and mitogen-activated protein kinase 10 (MAPK10) on predicting prognosis. METHODS This study selected data from 53 patients with HCC who had undergone hepatectomy in our hospital. The expressions of PLZF and MAPK10 in tumor tissues and normal tissues were compared and related clinical factors were analyzed. The clinical data including patient's gender, age, hepatitis B virus infection (HBV), alpha-fetoprotein levels (AFP), tumor size, TNM stage (AJCC), cirrhosis, portal vein tumor thrombus (PVTT), bile duct tumor thrombus (BDTT), and OS (Overall survival) was collected. RESULTS We found that PLZF expression was significantly down-regulated in HCC samples compared with that in adjacent non-tumor tissues (P=0.001). The expression level of PLZF was correlated with patients' gender (P=0.046), tumor stage (P=0.039), and OS (P=0.015). Moreover, the expression level of PLZF-MAPK10 (P-M protein) was correlated with gender (P=0.000) and tumor stage (P=0.045). Multivariate analyses showed that microsatellite nodules, PLZF, and P-M protein were independent risk factors of HCC prognosis. Postoperative patients with a normal expression level of PLZF and MAPK10 have a longer overall survival than those with abnormal levels (P=0.039). CONCLUSION PLZF expression was significantly down-regulated in HCC tissues and itself and PLZF-MAPK10 were both independent prognostic factors for the OS of patients with HCC.
Collapse
Affiliation(s)
- Shanshan Hu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Yue Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Liheng Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Xuedong Yin
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Yaying Yang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing, China
| | - Liping Tang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| |
Collapse
|
25
|
Nakano R, Nakayama T, Sugiya H. Biological Properties of JNK3 and Its Function in Neurons, Astrocytes, Pancreatic β-Cells and Cardiovascular Cells. Cells 2020; 9:cells9081802. [PMID: 32751228 PMCID: PMC7464089 DOI: 10.3390/cells9081802] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022] Open
Abstract
JNK is a protein kinase, which induces transactivation of c-jun. The three isoforms of JNK, JNK1, JNK2, and JNK3, are encoded by three distinct genes. JNK1 and JNK2 are expressed ubiquitously throughout the body. By contrast, the expression of JNK3 is limited and observed mainly in the brain, heart, and testes. Concerning the biological properties of JNKs, the contribution of upstream regulators and scaffold proteins plays an important role in the activation of JNKs. Since JNK signaling has been described as a form of stress-response signaling, the contribution of JNK3 to pathophysiological events, such as stress response or cell death including apoptosis, has been well studied. However, JNK3 also regulates the physiological functions of neurons and non-neuronal cells, such as development, regeneration, and differentiation/reprogramming. In this review, we shed light on the physiological functions of JNK3. In addition, we summarize recent advances in the knowledge regarding interactions between JNK3 and cellular reprogramming.
Collapse
Affiliation(s)
- Rei Nakano
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- Laboratory of Veterinary Radiology, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa 252-0880, Japan; (T.N.); (H.S.)
- Correspondence:
| | - Tomohiro Nakayama
- Laboratory of Veterinary Radiology, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa 252-0880, Japan; (T.N.); (H.S.)
| | - Hiroshi Sugiya
- Laboratory of Veterinary Radiology, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa 252-0880, Japan; (T.N.); (H.S.)
| |
Collapse
|
26
|
Duan Y, Cheng S, Jia L, Zhang Z, Chen L. PDRPS7 protects cardiac cells from hypoxia/reoxygenation injury through inactivation of JNKs. FEBS Open Bio 2020; 10:593-606. [PMID: 32108998 PMCID: PMC7137793 DOI: 10.1002/2211-5463.12822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/06/2020] [Accepted: 02/25/2020] [Indexed: 11/09/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a major complication of reperfusion therapy in myocardial infarction. Ischemic myocardium produces a variety of peptides. We recently identified PDRPS7 as a novel peptide in cardiomyocytes that can be induced by hypoxia. However, the role of PDRPS7 is unknown. Here, we investigated the effects of PDRPS7 on hypoxia/reoxygenation (H/R)‐induced injury in rat cardiomyoblast H9c2 cells and NRCMs. We found that PDRPS7 improved cell survival and attenuated lactate dehydrogenase leakage following H/R in H9c2 cells and NRCMs. PDRPS7 also alleviated H/R‐induced pulsation reduction in NRCMs. Moreover, H/R‐induced cell apoptosis was decreased in the presence of PDRPS7. H/R‐induced reactive oxygen species generation was reduced by PDRPS7; in addition, PDRPS7 did not impact H2O2‐induced cell injury. Signaling analysis demonstrated that H/R increased the phosphorylation levels of JNKs, ERKs, and p38 mitogen‐activated protein kinases. However, PDRPS7 only attenuated H/R‐induced JNK phosphorylation, but not phosphorylation of ERKs and p38. PDRPS7 protected cardiomyocytes from apoptosis by inhibiting JNK phosphorylation and c‐Jun phosphorylation pathways, markedly upregulating anti‐apoptotic Bcl‐2 expression and inhibiting that of pro‐apoptotic Bax and cleaved caspase‐3. Importantly, pharmacological activation of JNKs diminished the protective effect of PDRPS7 in terms of cell survival against H/R stimulation. In summary, our study identified PDRPS7 as a novel cardioprotective peptide against H/R challenge and this action was mediated, at least in part, through inactivation of JNKs.
Collapse
Affiliation(s)
- Yulian Duan
- College of Life Sciences, Nanjing Normal University, China
| | - Siyuan Cheng
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Liang Jia
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Zhao Zhang
- College of Life Sciences, Nanjing Normal University, China
| | - Leilei Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| |
Collapse
|