1
|
Zhu LR, Cui W, Liu HP. Molecular mechanisms of endoplasmic reticulum stress-mediated acute kidney injury in juvenile rats and the protective role of mesencephalic astrocyte-derived neurotrophic factor. J Pharm Pharmacol 2025; 77:609-620. [PMID: 39437337 DOI: 10.1093/jpp/rgae134] [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: 01/30/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES This study examined the role of endoplasmic reticulum stress in pediatric acute kidney injury and the therapeutic effect of midbrain astrocyte-derived neurotrophic factor. METHODS Two-week-old Sprague-Dawley rats were divided into: Sham, ischemia-reperfusion injury-induced acute kidney injury (AKI), mesencephalic astrocyte-derived neurotrophic factor (MANF)-treated, tauroursodeoxycholic acid (TUDCA)-treated. Analyses were conducted 24 h post-treatment. Serum creatinine, cystatin C, Albumin, MANF levels were measured, cytokine concentrations in serum and renal tissues were determined using a Luminex assay. Histopathology was assessed via light and electron microscopy. Western blotting and RT-qPCR analyzed markers for oxidative stress, apoptosis, endoplasmic reticulum (ER) stress, and autophagy. HK-2 cells underwent hypoxia/reoxygenation (H/R) to simulate AKI and were treated with MANF or TUDCA. RESULTS AKI rats had increased serum creatinine, cystatin C, and inflammatory cytokines, along with significant renal damage, and showed loose and swollen ER structures, reduced cell proliferation, and elevated levels of IRE1, PERK, ATF6, CHOP, LC3-II/I, KIM-1, TLR4, JNK, and NF-κB. MANF treatment reduced these biomarkers and protein levels, improved ER structure and cell proliferation, alleviated oxidative stress, apoptosis, ER stress, and inhibited JNK/TLR4/NF-κB signaling. In HK-2 cells, MANF reduced ER stress and inflammation post-H/R exposure. CONCLUSIONS MANF treatment alleviates ER stress, oxidative stress, apoptosis, and inflammation in pediatric AKI, improving renal function and morphology.
Collapse
Affiliation(s)
- Li-Ran Zhu
- Anhui Institute of Pediatric Research, Anhui Provincial Children's Hospital (Children's Hospital of Fudan University Anhui Hospital, Children's Medical Center of Anhui Medical University), Wangjiang Road, Hefei, 230051 Anhui, China
| | - Wei Cui
- Department of Scientific Research and Education, Anhui Provincial Children's Hospital (Children's Hospital of Fudan University Anhui Hospital, Children's Medical Center of Anhui Medical University), Wangjiang Road, Hefei, 230051 Anhui, China
| | - Hai-Peng Liu
- Anhui Institute of Pediatric Research, Anhui Provincial Children's Hospital (Children's Hospital of Fudan University Anhui Hospital, Children's Medical Center of Anhui Medical University), Wangjiang Road, Hefei, 230051 Anhui, China
| |
Collapse
|
2
|
Shin J, Kim JS, Jung YJ, Lee Y, Yoo H, Ju SH, Sim D, Kim Y, Bae GW, Yoon SM, Lee SJ. Activation of hepatic alcohol metabolism by enzymatic porcine placenta hydrolysate in rats. Food Sci Biotechnol 2025; 34:2025-2038. [PMID: 40196334 PMCID: PMC11972273 DOI: 10.1007/s10068-025-01822-1] [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: 09/06/2024] [Revised: 01/02/2025] [Accepted: 01/13/2025] [Indexed: 04/09/2025] Open
Abstract
Alcohol consumption causes severe liver damage and oxidative stress. This study investigated the hepatoprotective effects of enzymatic porcine placenta hydrolysate (EPPH) in Sprague-Dawley rats under acute alcohol administration. EPPH significantly reduced plasma ethanol and acetaldehyde levels in a dose-dependent manner. Furthermore, EPPH decreased the hepatic levels of malondialdehyde and thiobarbituric acid reactive substances and suppressed Cyp2e1 mRNA expression. EPPH decreased the plasma alanine transaminase and aspartate transaminase levels and increased the hepatic NAD+/NADH ratio. Hepatic transcriptome analysis revealed the significant regulation of key genes involved in inflammation, alcohol response, and apoptosis. Phosphokinase array analysis demonstrated that EPPH reduced phosphorylation of CASP9, BAX, TP53, and CHK2, thereby facilitating reactive oxygen species removal and suppressing apoptosis. Additionally, qPCR confirmed EPPH reduced Bax and Caspase9 mRNA levels, while immunoblotting showed decreased phosphorylation of TP53 and CHK2. These findings suggest that EPPH improves hepatic alcohol metabolism and reduces alcohol-induced hepatotoxicity. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-025-01822-1.
Collapse
Affiliation(s)
- Jaeeun Shin
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Ji-Sun Kim
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Young Jae Jung
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Yeonho Lee
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Haeyoung Yoo
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Seong Hun Ju
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Daehyeon Sim
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Yebean Kim
- Department of Biotechnology, Graduate School of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841 South Korea
| | - Gun Won Bae
- Unimed Pharmaceuticals Inc., UNIMED Bldg #69, Samjeon-ro, Songpa-gu, Seoul, 05567 South Korea
| | - Sun Myung Yoon
- Unimed Pharmaceuticals Inc., UNIMED Bldg #69, Samjeon-ro, Songpa-gu, Seoul, 05567 South Korea
| | - Sung-Joon Lee
- Interdisciplinary Program in Precision Public Health, BK21 Four Institute of Precision Public Health, Korea University, Seoul, 02841 South Korea
- R&D, Ector Biotherapeutics, 145 Anam-ro, Seongbuk-gu, Seoul, 02841 South Korea
| |
Collapse
|
3
|
Gao H, Liu T, Liu J, Yang L, Liu L, Cui Z, Du X, Gu Y, Huang P. Oleanolic Acid@SPIONs Alleviates Lipid-Oxidative Stress Injury of Zebrafish Blood Vessels via Regulating the Expression of JNK and MAPK Signaling Pathways in Vascular Endothelial Cells. Drug Des Devel Ther 2025; 19:2921-2940. [PMID: 40255470 PMCID: PMC12009123 DOI: 10.2147/dddt.s512752] [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: 01/14/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
Background and Aims The incidence of and mortality due to atherosclerosis, a leading cause of cardiovascular disease, is rising annually. Oleanolic acid (OA), an active component of the traditional Chinese medicine Ligustrum lucidum, has been proven to have significant anti-inflammatory and lipid-lowering potential. Methods The fli1a::EGFP+ zebrafish fed with oxidized low-density lipoprotein (oxLDL) diet were used as Atherosclerosis model. The zebrafish Atherosclerosis model were fed with oxalic acid driven by superparamagnetic ferrite nanoparticles (OA@SPIONs). Isolation and enrichment of fli1a::EGFP+ zebrafish endothelial cells (zeECs) from each group and RNA-seq to analyze changes in gene transcription. The H&E, MASSION, Oil red O staining were used to identifying pathological phenotypes. Results Pathological staining and ultrastructural identification indicated that oxLDL-treated zebrafish exhibited significant lipid plaque deposition and signs of cellular senescence that were significantly alleviated by OA@SPIONs treatment. OA@SPIONs treatment notably improved the ultrastructural integrity of myocardial, liver, and intestinal tissues in oxLDL-treated zebrafish. The RNA-seq results showed that OA@SPIONs treatment significantly altered the expression levels of multiple gene transcripts in zeECs. The KEGG analysis revealed that in the OA@SPION-treated group zeECs, key genes in the JNK and MAPK signaling pathways, such as Cacna1c, Rab1ab (Ras), Map3k1 (MEKK1), Mapk8b (JNK), and JunD, had significantly lower sequencing signals than in the oxLDL+SPION-treated group zeECs. The qPCR results were highly consistent with the RNA-sequencing data. Conclusion Therefore, our results confirm that SPIONs can effectively deliver OA for stable release in zebrafish and provide strong evidence that OA@SPION-polyethyleneimine exerts protective effects against oxLDL-induced damage in zebrafish by downregulating the expression of the JNK and MAPK signaling pathways.
Collapse
Affiliation(s)
- Hongguo Gao
- Department of Traditional Chinese Medicine, Graduate School, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330100, People’s Republic of China
- Department of Geriatric Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, People’s Republic of China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, People’s Republic of China
| | - Junfeng Liu
- Department of Nephrology, Shanghai Quyang Hospital, Shanghai, 200083, People’s Republic of China
| | - Lian Yang
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, People’s Republic of China
| | - Luxi Liu
- Class 1, Grade 8, Shanghai Wenlai Middle School, Shanghai, 201101, People’s Republic of China
| | - Zeyu Cui
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, People’s Republic of China
| | - Xiling Du
- School of Life Science and Technology, Tongji University, Shanghai, 200092, People’s Republic of China
| | - Yun Gu
- Department of Traditional Chinese Medicine, Graduate School, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330100, People’s Republic of China
- Department of Geriatric Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, People’s Republic of China
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, People’s Republic of China
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People’s Republic of China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, People’s Republic of China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200080, People’s Republic of China
| |
Collapse
|
4
|
Zhang X, Yan Q, Xiao Y, Du X, Zhang X, Lou D, Peng F, Chen D, Tang W. Integrating network pharmacology, molecular docking, and animal studies to investigate the protective effect of astragalus polysaccharide on fluoride-induced renal injury in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118109. [PMID: 40154226 DOI: 10.1016/j.ecoenv.2025.118109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Fluoride is an essential trace element required for normal physiological functions and holds significant importance for human health. However, excessive fluoride intake can lead to renal damage, for which effective prevention and therapeutic strategies remain scarce. Astragalus polysaccharide (APS), a major bioactive component of the traditional Chinese herb Astragalus membranaceus, possesses pharmacological properties including anti-inflammatory, antiviral, and antioxidant activities. In this study, we investigated the protective effects of APS against fluoride-induced renal injury in vivo experiment. Additionally, network pharmacology and molecular docking techniques were employed to predict its potential mechanisms of action, while the protein expression levels of key target molecules were validated. The results demonstrated that APS intervention significantly alleviated renal injury and oxidative stress induced by sodium fluoride (NaF) in rats. Key targets involved in the amelioration of fluoride-induced renal damage by APS included STAT3, Caspase-3, JUN, MMP1, and PTGS2. Molecular docking analysis revealed high-affinity binding between APS and these core targets. Immunohistochemical and Western blot analysis further confirmed that APS suppressed the expression of pro-apoptotic proteins STAT3, Caspase-3, JUN, and MMP1 while enhancing the expression of the anti-apoptotic protein PTGS2. Overall, our findings suggest that APS alleviates fluoride-induced renal injury by modulating multiple targets, with the potential mechanism linked to the regulation of apoptotic processes. This study provides a theoretical basis for the prevention and treatment of fluoride toxicity.
Collapse
Affiliation(s)
- Xiaoyue Zhang
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Qianda Yan
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Yuanyuan Xiao
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xingyan Du
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xuehua Zhang
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Didong Lou
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; School of Public Health, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Fang Peng
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; School of Public Health, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Daiyong Chen
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Wenchao Tang
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; School of Public Health, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
5
|
Ahn JY, Lim DW, Park SY, Lee JH. Sinhyotaklisan alleviates inflammation in LPS-activated macrophages by modulating the heme oxygenase pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119548. [PMID: 40015540 DOI: 10.1016/j.jep.2025.119548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/22/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shinhyotaklisan (SHTLS) is a time-honored traditional Korean herbal formula composed of four key herbs: Lonicerae Flos, Astragali Radix, Angelicae Gigantis Radix, and Glycyrrhizae Radix et Rhizoma. It has been extensively used to treat inflammatory diseases by expelling excessive heat, detoxifying the body, and promoting the drainage of pus from abscesses. AIM OF THE STUDY This study examines the therapeutic effects of SHTLS and elucidates its mechanisms of action in alleviating lipopolysaccharide (LPS)-induced inflammation in vitro. MATERIALS AND METHODS SHTLS was prepared by boiling four herbs in 30% ethanol, and its antioxidant and antimicrobial effects were assessed. Furthermore, SHTLS was applied to LPS-exposed RAW 264.7 cells, and its anti-inflammatory effects were evaluated using an MTT assay, nitric oxide (NO) and reactive oxygen species (ROS) assays, enzyme-linked immunosorbent assay, and western blotting. RESULTS SHTLS demonstrated potent antioxidant and antimicrobial effects. It effectively suppressed LPS-induced inflammatory cascades, resulting in a significant reduction in pro-inflammatory cytokines, including IL-6 and TNF-α, in murine macrophage cells. Moreover, SHTLS decreased COX-2 and iNOS expression levels, primarily through the inhibition of NF-κB and MAPK signaling pathways. Additionally, SHTLS significantly reduced intracellular ROS and reactive nitrogen species levels by upregulating heme oxygenase-1 (HO-1) expression. The anti-inflammatory effects of SHTLS were diminished by the addition of an HO-1 inhibitor, underscoring its strong association with intracellular antioxidant mechanisms. CONCLUSION Our results suggest that SHTLS exhibits strong antioxidant and anti-inflammatory effects, primarily by upregulating HO-1 and inhibiting the NF-κB and MAPK pathways, highlighting its potential as a therapeutic agent for inflammation-related conditions.
Collapse
Affiliation(s)
- Jung-Yun Ahn
- College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea.
| | - Dong-Woo Lim
- College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea.
| | - Sung Yun Park
- College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea.
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea.
| |
Collapse
|
6
|
Eldehna WM, Roshdy E, Abdulla MH, Zain-Alabdeen AI, Shaldam MA, Alhassan NS, Traiki TB, Al-Sanea MM, El-Hamaky AA, Al-Karmalawy AA, El Kerdawy AM, Abe M, Tawfik HO. Discovery of 1-phenyl-1,2,3-triazole ureas as dual VEGFR-2/JNK-1 type II kinase inhibitors targeting pancreatic cancer. Int J Biol Macromol 2025; 308:142372. [PMID: 40139613 DOI: 10.1016/j.ijbiomac.2025.142372] [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: 12/02/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
In oncology, pancreatic cancer (PC) continues to be a problem that requires creative approaches to therapy. This research aims to create dual kinase inhibitors that target VEGFR-2 and JNK-1, two important factors in the angiogenesis and progression of PC. We found compounds with promising anticancer action using phenyltriazolyl piperazine/(piperidine) carboxamides (PTPCs) and phenyltriazolyl phenylureas (PTPUs). Compound 12b was the most effective in inhibiting VEGFR-2 (IC50: 46 nM) and JNK-1 (IC50: 35 nM) and showed the highest activity against PANC-1 cancer cells (IC50: 1.05 μM). Furthermore, 12b altered the caspase-3, Bcl-2, and Bax apoptotic markers. The binding interactions of 12b with target kinases were discovered by in silico investigations. This study emphasizes how dual kinase inhibitors may be a viable way to improve the effectiveness of cancer treatments and deal with resistance mechanisms.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt.
| | - Eslam Roshdy
- Department of Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, Higashi-Hiroshima City, Hiroshima 739-8526, Japan; Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Maha-Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia.
| | | | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt
| | - Noura S Alhassan
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Thamer Bin Traiki
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Mohammad M Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf 72388, Saudi Arabia
| | - Anwar A El-Hamaky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Ahmed M El Kerdawy
- School of Health and Care Sciences, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, United Kingdom; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, P.O. Box 11562, Cairo, Egypt
| | - Manabu Abe
- Department of Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, Higashi-Hiroshima City, Hiroshima 739-8526, Japan
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
7
|
Xia H, He T, Li X, Zhao K, Zhang Z, Zhu G, Yang H, Yan X, Wang Q, Li Z, Jiang Z, Wang K, Yin X. Study on the mechanism of BGN in progression and metastasis of ccRCC. BMC Med Genomics 2025; 18:55. [PMID: 40108593 PMCID: PMC11924620 DOI: 10.1186/s12920-025-02124-5] [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: 03/24/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE To investigate the role of Biglycan(BGN) in the progression and metastasis of clear cell renal cell carcinoma(ccRCC). METHODS Based on multiple public databases, we investigated the expression level of BGN in ccRCC, its clinical significance, and its association with immune cells. Real-time fluorescence quantitative polymerase chain reaction(PCR) was employed to validate BGN expression in tumor and adjacent normal tissues from ten patients. We utilized RNA sequencing results for further analysis, including differential gene analysis, GO-KEGG analysis, and GSEA analysis, to identify the signaling pathways through which BGN exerts its effects. BGN knockdown cells(786-0 and Caki-1) were generated through lentiviral transfection to examine the impact of BGN on ccRCC. Cell proliferation, migration, and invasion were assessed using CCK8, colony formation, wound healing, Transwell migration, and invasion assays, respectively. RESULTS Our findings from database analysis and PCR revealed a significant upregulation of BGN expression in kidney cancer tissues compared to normal tissues. Further analysis demonstrated a correlation between high BGN expression and ccRCC progression and immune infiltration. In vitro experiments confirmed that BGN silencing effectively inhibited cell proliferation, migration, and invasion of ccRCC. Mechanistically, these effects may be mediated through the MAPK signaling pathway. CONCLUSION BGN potentially plays a pivotal role in the progression and metastasis of ccRCC, possibly acting through the MAPK signaling pathway. Therefore, BGN holds promise as a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Hanqing Xia
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Tianzhen He
- Institute of Special Environmental Medicine, Nantong University, Nantong, China
| | - Xueyu Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Kai Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zongliang Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Guanqun Zhu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Han Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xuechuan Yan
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Qinglei Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zhaofeng Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zaiqing Jiang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Ke Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xinbao Yin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China.
| |
Collapse
|
8
|
Zhang Y, Wu Y, Pei B, Sun Q, Zhang C, Yang Q, Jin Y, Wu J, Li X. Piwei Peiyuan Prescription Attenuates the Progression of Chronic Atrophic Gastritis by Eliciting MAPK10-Mediated Mitochondrial Autophagy. Cell Biol Int 2025. [PMID: 40103313 DOI: 10.1002/cbin.70016] [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: 12/11/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Piwei Peiyuan (PWPY) prescription is a traditional Chinese medicine prescription and has been efficiently used in the clinics to treat chronic atrophic gastritis (CAG) for many years. However, the mechanism of action underlying PWPY for treating CAG remains elusive. A CAG rat animal and cell model was constructed in this study to explore the action mechanism of PWPY prescription in treating CAG. Here we show that PWPY attenuates the progression of CAG by eliciting MAPK10-mediated mitochondrial autophagy. Experimental model of CAG was introduced using N-methyl-n'-nitro-n-nitroguanidine (MNNG). Our histological analyses reveal that MNNG-induced CAG in rat undergoes classical morphological alterations judged by immunohistochemistry and serum level of PGⅠ, PGⅡ, and G17. Importantly, PWPY treatment prevents the progression of MNNG-induced CAG judged by serum level of PGⅠ, PGⅡ, and G17. Interestingly, PWPY treatment inhibits MAPK10 activity judged by biochemical assays and promotes mitochondrial autophagy judged by electron microscopic analyses. Thus, we conclude that PWPY attenuates the progression of MNNG-induced CAG and prevents precancerous lesions by harnessing MAPK10-elicited mitochondrial autophagy. The MNNG-induced experimental CAG model provides a robust platform for further delineating therapeutic targets underlying PWPY regimen.
Collapse
Affiliation(s)
- Yi Zhang
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Ying Wu
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Bei Pei
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Qin Sun
- The Department of Spleen and Stomach, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Cheng Zhang
- The Research Department, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Qi Yang
- The Department of Spleen and Stomach, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Yueping Jin
- The Department of Spleen and Stomach, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Jing Wu
- The Department of Spleen and Stomach, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Xuejun Li
- The Department of Spleen and Stomach, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
9
|
Huang YT, Calvi BR. Activation of a Src-JNK pathway in unscheduled endocycling cells of the Drosophila wing disc induces a chronic wounding response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642788. [PMID: 40161657 PMCID: PMC11952448 DOI: 10.1101/2025.03.12.642788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The endocycle is a specialized cell cycle during which cells undergo repeated G / S phases to replicate DNA without division, leading to large polyploid cells. The transition from a mitotic cycle to an endocycle can be triggered by various stresses, which results in unscheduled, or induced endocycling cells (iECs). While iECs can be beneficial for wound healing, they can also be detrimental by impairing tissue growth or promoting cancer. However, the regulation of endocycling and its role in tissue growth remain poorly understood. Using the Drosophila wing disc as a model, we previously demonstrated that iEC growth is arrested through a Jun N-Terminal Kinase (JNK)-dependent, reversible senescence-like response. However, it remains unclear how JNK is activated in iECs and how iECs impact overall tissue structure. In this study, we performed a genetic screen and identified the Src42A-Shark-Slpr pathway as an upstream regulator of JNK in iECs, leading to their senescence-like arrest. We found that tissues recognize iECs as wounds, releasing wound-related signals that induce a JNK-dependent developmental delay. Similar to wound closure, this response triggers Src-JNK-mediated actomyosin remodeling, yet iECs persist rather than being eliminated. Our findings suggest that the tissue response to iECs shares key signaling and cytoskeletal regulatory mechanisms with wound healing and dorsal closure, a developmental process during Drosophila embryogenesis. However, because iECs are retained within the tissue, they create a unique system that may serve as a model for studying chronic wounds and tumor progression.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Indiana University, Bloomington, Indiana, 47405 USA
| | - Brian R. Calvi
- Department of Biology, Indiana University, Bloomington, Indiana, 47405 USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, 46202 USA
| |
Collapse
|
10
|
Bednarski IA, Dróżdż I, Ciążyńska M, Wódz K, Narbutt J, Lesiak A. Ultraviolet B Exposure Does Not Influence the Expression of YAP mRNA in Human Epidermal Keratinocytes-Preliminary Study. Biomedicines 2025; 13:596. [PMID: 40149574 PMCID: PMC11940570 DOI: 10.3390/biomedicines13030596] [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: 01/14/2025] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Background: The causal relationship between exposure to ultraviolet radiation and the development of skin cancers requires constant research for possible orchestrating mechanisms. In recent years, the Hippo pathway, along with its effector protein YAP, became implicated in cutaneous carcinogenesis; however, Hippo pathway regulation by ultraviolet radiation has not been described thoroughly. In order to address this issue, we focused on how different doses of ultraviolet B affect Hippo signaling pathway components and its upstream regulators, JNK1/2 and ABL1, in human keratinocytes. Additionally, we decided to determine how silencing of YAP influences Hippo pathway component expression. Methods: Primary epidermal keratinocytes were irradiated using UVB lamps with increasing doses of ultraviolet B radiation (including 311 nm UVB). Real-time PCR was used to determine the mRNA levels of each investigated gene. The experiment was then performed after YAP silencing using siRNA transfection. Additionally, we determined the mRNA expression of Hippo pathway components in an A431 cSCC cell line. Results: We observed that YAP mRNA expression in the A431 cell line was insignificant in comparison to control, while in the case of LATS1/2, a significant increase was noted. UVB irradiation did not change the levels of YAP mRNA expression in human epidermal keratinocytes. LATS1, LATS2, ABL1 and MAP4K4 mRNA expression was significantly upregulated after UVB irradiation in non-YAP-silenced keratinocytes in a dose-dependent manner, while after YAP silencing, only LATS2 and ABL1 showed significant mRNA upregulation. The 311 nm UVB irradiation resulted in significant, dose-dependent mRNA upregulation in non-YAP-silenced keratinocytes for LATS1, ABL1 and MAP4K4. After YAP silencing, a significant change in mRNA expression was present only in the case of ABL1. Conclusions: YAP mRNA expression does not significantly increase after exposure to UVB; however, it upregulates the expression of its proven (LATS1/2, JNK1/2) regulators, suggesting that in real-life settings, UV-induced dysregulation of the Hippo pathway may not be limited to YAP.
Collapse
Affiliation(s)
- Igor Aleksander Bednarski
- Dermatology, Pediatric Dermatology and Dermatological Oncology Clinic, Medical University of Łódź, 91-347 Łódź, Poland; (I.A.B.); (J.N.)
- Department of Neurology, Medical University of Łódź, 90-419 Łódź, Poland
| | - Izabela Dróżdż
- Department of Clinical Genetics, Medical University of Łódź, 92-213 Łódź, Poland;
| | - Magdalena Ciążyńska
- Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Medical University of Łódź, 93-513 Łódź, Poland;
| | - Karolina Wódz
- Laboratory of Molecular Biology, Vet-Lab Brudzew, 62-720 Brudzew, Poland;
| | - Joanna Narbutt
- Dermatology, Pediatric Dermatology and Dermatological Oncology Clinic, Medical University of Łódź, 91-347 Łódź, Poland; (I.A.B.); (J.N.)
| | - Aleksandra Lesiak
- Dermatology, Pediatric Dermatology and Dermatological Oncology Clinic, Medical University of Łódź, 91-347 Łódź, Poland; (I.A.B.); (J.N.)
| |
Collapse
|
11
|
Ding B, Li J, Yan JL, Jiang CY, Qian LB, Pan J. Resveratrol contributes to NK cell-mediated breast cancer cytotoxicity by upregulating ULBP2 through miR-17-5p downmodulation and activation of MINK1/JNK/c-Jun signaling. Front Immunol 2025; 16:1515605. [PMID: 39963142 PMCID: PMC11830804 DOI: 10.3389/fimmu.2025.1515605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUNDS Natural killer (NK) cell mediated cytotoxicity is a crucial form of anti-cancer immune response. Natural killer group 2 member D (NKG2D) is a prominent activating receptor of NK cell. UL16-binding protein 2 (ULBP2), always expressed or elevated on cancer cells, functions as a key NKG2D ligand. ULBP2-NKG2D ligation initiates NK cell activation and subsequent targeted elimination of cancer cells. Enhanced expression of ULBP2 on cancer cells leads to more efficient elimination of these cells by NK cells. Resveratrol (RES) is known for its multiple health benefits, while current understanding of its role in regulating cancer immunogenicity remains limited. This study aims to investigate how RES affects the expression of ULBP2 and the sensitivity of breast cancer (BC) cells to NK cell cytotoxicity, along with the underlying mechanisms. METHODS The effects of RES on ULBP2 expression were detected with qRT-PCR, western blot, flow cytometry analysis and immunohistochemistry. The effects of RES on sensitivity of BC cells to NK cell cytotoxicity were evaluated in vitro and in vivo. The target gene of miR-17-5p were predicted with different algorithms from five databases and further confirmed with dual-luciferase reporter assay. Overexpression and knockdown experiments of miR-17-5p and MINK1 were conducted to investigate their roles in regulating ULBP2 expression and subsequent JNK/c-Jun activation. The JNK inhibitor sp600125 was utilized to elucidate the specific role of JNK in modulating ULBP2 expression. RESULTS RES increased ULBP2 expression on BC cells, thereby augmenting their vulnerability to NK cell-mediated cytotoxicity both in vitro and in vivo. RES administration led to a reduction in cellular miR-17-5p level. MiR-17-5p negatively regulated ULBP2 expression. Specifically, miR-17-5p directly targeted MINK1, leading to its suppression. MINK1 played a role in facilitating the activation of JNK and its downstream effector, c-Jun. Furthermore, treatment with sp600125, a JNK inhibitor, resulted in the suppression of ULBP2 expression. CONCLUSIONS: RES potentiates ULBP2-mediated immune eradication of BC cells by NK cells through the downregulation of miR-17-5p and concurrent activation of the MINK1/JNK/c-Jun cascade. This finding identifies RES as a potentially effective therapeutic agent for inhibiting BC progression and optimizing NK cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Bisha Ding
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jie Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jia-Lin Yan
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chun-Yan Jiang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ling-Bo Qian
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jie Pan
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Guneri-Sozeri PY, Adebali O. Transcription factors, nucleotide excision repair, and cancer: A review of molecular interplay. Int J Biochem Cell Biol 2025; 179:106724. [PMID: 39672502 DOI: 10.1016/j.biocel.2024.106724] [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: 08/29/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Bulky DNA adducts are mostly formed by external factors such as UV irradiation, smoking or treatment with DNA crosslinking agents. If such DNA adducts are not removed by nucleotide excision repair, they can lead to formation of driver mutations that contribute to cancer formation. Transcription factors (TFs) may critically affect both DNA adduct formation and repair efficiency at the binding site to DNA. For example, "hotspot" mutations in melanoma coincide with UV-induced accumulated cyclobutane pyrimidine dimer (CPD) adducts and/or inhibited repair at the binding sites of some TFs. Similarly, anticancer treatment with DNA cross-linkers may additionally generate DNA adducts leading to secondary mutations and the formation of malignant subclones. In addition, some TFs are overexpressed in response to UV irradiation or chemotherapeutic treatment, activating oncogenic and anti-oncogenic pathways independently of nucleotide excision repair itself. This review focuses on the interplay between TFs and nucleotide excision repair during cancer development and progression.
Collapse
Affiliation(s)
| | - Ogün Adebali
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Türkiye.
| |
Collapse
|
13
|
Wankhede NL, Kale MB, Kyada A, M RM, Chaudhary K, Naidu KS, Rahangdale S, Shende PV, Taksande BG, Khalid M, Gulati M, Umekar MJ, Fareed M, Kopalli SR, Koppula S. Sleep deprivation-induced shifts in gut microbiota: Implications for neurological disorders. Neuroscience 2025; 565:99-116. [PMID: 39622383 DOI: 10.1016/j.neuroscience.2024.11.070] [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: 07/31/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Sleep deprivation is a prevalent issue in contemporary society, with significant ramifications for both physical and mental well-being. Emerging scientific evidence illuminates its intricate interplay with the gut-brain axis, a vital determinant of neurological function. Disruptions in sleep patterns disturb the delicate equilibrium of the gut microbiota, resulting in dysbiosis characterized by alterations in microbial composition and function. This dysbiosis contributes to the exacerbation of neurological disorders such as depression, anxiety, and cognitive decline through multifaceted mechanisms, including heightened neuroinflammation, disturbances in neurotransmitter signalling, and compromised integrity of the gut barrier. In response to these challenges, there is a burgeoning interest in therapeutic interventions aimed at restoring gut microbial balance and alleviating neurological symptoms precipitated by sleep deprivation. Probiotics, dietary modifications, and behavioural strategies represent promising avenues for modulating the gut microbiota and mitigating the adverse effects of sleep disturbances on neurological health. Moreover, the advent of personalized interventions guided by advanced omics technologies holds considerable potential for tailoring treatments to individualized needs and optimizing therapeutic outcomes. Interdisciplinary collaboration and concerted research efforts are imperative for elucidating the underlying mechanisms linking sleep, gut microbiota, and neurological function. Longitudinal studies, translational research endeavours, and advancements in technology are pivotal for unravelling the complex interplay between these intricate systems.
Collapse
Affiliation(s)
- Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences Marwadi University, Rajkot 360003, Gujarat, India
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sandip Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
14
|
HARISA GAMALELDINI, ALZHRANI RIYADF, ALLUHAIDAN ABDULRAHMANA, ALAMRI SULTANM, BAKHEIT AHMEDH, ASIRI HANADIH, ATTIA SABRYM. Chitosan capped-NLCs enhanced codelivery of gefitinib and simvastatin into MDR HCC: impact of compositions on cell death, JNK3, and Telomerase. Oncol Res 2025; 33:477-492. [PMID: 39866231 PMCID: PMC11754001 DOI: 10.32604/or.2024.053337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/08/2024] [Indexed: 01/28/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a health problem due to multi-drug resistance (MDR). Codelivery of multiple oncotherapy in one cargo as chimeric cancer therapy (CCT) is suggested as a solution for MDR. This study aims to engineer chitosan-coated nanostructure lipid carriers (NLCs) loaded with gefitinib (GF) and simvastatin (SV) as CCT for HCC. Methods Both GF and SV-loaded nanostructure lipids carriers (GFSVNLC) and chitosan-capped GF and SV-loaded nanostructure lipids carriers (CGFSVNLC) formulations were assembled by top-down techniques. Moreover, particle size (PS), zeta potential (ZP), and polydispersity index (PDI) were measured by Zetasizer. The biosafety of GFSVNLC preparations was investigated by using erythrocytes as a biological model. The cytotoxic, and apoptotic effects of the prepared GFSVNLCs were investigated using HepG2 cell lines as a substitute model for HCC. The effect of GF, SV, and NLC composition on JNK3, HDAC6, and telomerase was studied using molecular docking simulation (MDS). Results The present results revealed that the obtained GFSVNLC and CGFSVNLC have nanosized and consistent, CS coating shifts anionic ZP of GFSVNLC into CGFSVNLC with cationic ZP. Moreover, both formulations are biocompatible as indicated by their gentle effect on erythrocyte hemolysis. The treatment of HepG2 cells with GFSVNLC, and CGFSVNLC induced marked cell death compared to other groups with a decrease of IC50. Equally, the percentage of the apoptotic HepG2 cells was increased upon treatment of the cells with GFSV, GFSVNLC, and CGFSVNLC compared to the control group. Additionally, GF, SV, stearic acid (SA), and oleic acid (OA) modulate the activity of JNK3, HDAC6, and telomerase. Conclusions This study suggests CGFSVNLC achieves codelivery, selective targeting, and enhancing the synergistic effect of GF and SV for inducing HepG2 cell death. Mechanistically, CGFSVNLC inhibits key cascades implicated in MDR and HepG2 cell survival. CGFSVNLC is promising for overcoming drug resistance mechanisms and improving therapeutic outcomes against HepG2 cells.
Collapse
Affiliation(s)
- GAMALELDIN I. HARISA
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - RIYAD F. ALZHRANI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | | | - SULTAN M. ALAMRI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - AHMED H. BAKHEIT
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - HANADI H. ASIRI
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - SABRY M. ATTIA
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
15
|
Al-Mahadeen MM, Jaber AM, Al-Najjar BO, Khanfar MA, El-Abadelah MM. Novel N-Substituted Isatin-Oxoindolin-1 H-Benzo[D] Imidazole Fumarate as a New Class of JNK3 Inhibitor: Design, Synthesis, Molecular Modeling and its Biological Activity. Curr Org Synth 2025; 22:410-418. [PMID: 40259593 DOI: 10.2174/0115701794335274240910111137] [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: 06/15/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 04/23/2025]
Abstract
BACKGROUND A direct synthesis of functionalized dimethyl fumarate derivatives of 2- (2-((E)-(2-oxoindolin-3-ylidene)methyl)-1H-benzo[d]imidazol-1-yl) is achieved via one-pot reaction involving 2-methyl-1H-benzo[d]imidazole and appropriate isatin in the presence of DMAD. METHODS Conversely, this one-pot reaction furnished, upon conduction at 60 ° C, the 2-(2-((E)- (2-oxoindolin-3-ylidene)methyl)-1H-benzo[d]imidazol-1-yl) products. The biological activities were evaluated against JNK3 kinase. We chose to dock the compounds into the JNK3 binding site in order to comprehend the molecular underpinnings of the observed bioactivities. RESULTS The structures of the synthesized compound adduct were evidenced from NMR and MS spectral data and further confirmed by single-crystal X-ray diffraction. The biological activities revealing that the introduction of an alkyl group at the 1-position of the isatin moiety produced JNK3 inhibitors with IC50 values in the low micromolar range. CONCLUSION This study synthesized a unique compound using a three-component method. Compound 4d showed high antitumor activity (IC50 = 6.5 μM) against JNK3 inhibitors, while compounds 4c, 4d, and 4f exhibited high selectivity. The research highlights the effectiveness of the one-pot reaction in creating medically useful hybrid compounds, marking a significant advance in medicinal chemistry.
Collapse
Affiliation(s)
- Mohammed M Al-Mahadeen
- Department of Chemistry , Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| | - Areej M Jaber
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Belal O Al-Najjar
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, 19328 Amman, Jordan
| | - Monther A Khanfar
- Department of Chemistry , Faculty of Science, The University of Jordan, Amman, 11942, Jordan
- Department of Chemistry, Pure and Applied Chemistry Group, College of Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Mustafa M El-Abadelah
- Department of Chemistry , Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
16
|
Lin FX, Gu HY, He W. MAPK signaling pathway in spinal cord injury: Mechanisms and therapeutic potential. Exp Neurol 2025; 383:115043. [PMID: 39522804 DOI: 10.1016/j.expneurol.2024.115043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Spinal cord injury (SCI) is a severe disabling injury of the central nervous system that can lead to motor, sensory, and autonomic dysfunction below the level of the injury. According to its pathophysiological process, SCI can be divided into primary injury and secondary injury. Currently, multiple therapeutic strategies have been proposed to alleviate secondary injury and overcome the occurrence of neurodegenerative events. Although current treatment modalities have achieved varying degrees of success, they cannot effectively intervene or treat its pathological processes, which may be due to the complex treatment and protection mechanisms involved. Research has confirmed that signaling pathways play a crucial role in the pathological processes of SCI and the mechanisms of neuronal recovery. Mitogen-activated protein kinase (MAPK) signaling pathway plays a crucial role in neuronal differentiation, growth, survival and axon regeneration after central nervous system injury. Meanwhile, the MAPK signaling pathway is an important pathway closely related to the pathological processes of SCI. The MAPK signaling pathway is abnormally activated after SCI, and inhibiting the activity of MAPK pathway can effectively inhibit inflammation, oxidative stress, pain and apoptosis to promote the recovery of nerve function after SCI. Based on the role of the MAPK pathway in SCI, it may be a potential therapeutic target. This article summarizes the role and mechanism of MAPK pathway in SCI, and discusses the shortcomings and shortcomings of MAPK pathway in SCI field, as well as the potential challenges of targeting MAPK pathway in SCI treatment strategies. This article aims to elucidate the mechanism of the MAPK pathway in SCI to emphasize the role of targeting the MAPK pathway in the treatment of SCI, providing a theoretical basis for the MAPK pathway as a potential therapeutic target for SCI treatment.
Collapse
Affiliation(s)
- Fei-Xiang Lin
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China
| | - Hou-Yun Gu
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China
| | - Wei He
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou 341000, Jiangxi Province, PR China.
| |
Collapse
|
17
|
Klein JC, Mahapatra R, Hon GC, Wang RC. Identification of Associations with Dermatologic Diseases through a Focused GWAS of the UK Biobank. JID INNOVATIONS 2025; 5:100322. [PMID: 39624183 PMCID: PMC11609433 DOI: 10.1016/j.xjidi.2024.100322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 01/31/2025] Open
Abstract
The UK Biobank includes genotype information for about 500,000 patients for over 7000 phenotypes. However, owing to multiple testing correction for approximately 200 billion tests, many clinically and statistically significant associations remain unappreciated. We perform a focused analysis of the UK Biobank for 13 dermatologic conditions, including malignant melanoma, melanoma in situ, squamous cell carcinoma, basal cell carcinoma, actinic keratosis, seborrheic keratosis, psoriasis, lichen planus, systemic lupus erythematosus, hyperhidrosis, pilonidal cyst, sebaceous cyst, and lipoma. We identify 447 sentinel variants, which are enriched for protein-coding variants and an elevated combined annotation-dependent depletion (CADD) score compared with background variants. Through gene ontology enrichment analysis, we identify known pathways involved in melanoma, actinic keratoses, and squamous cell carcinoma and uncover additional pathways. We also uncover 5 protein-coding variants, which, to our knowledge, have not been previously reported, including LRP3 for lipomas, PLCD1 for sebaceous cysts, EIF3CL for lichen planus, TTK for pilonidal cysts, and MAPK15 for systemic lupus erythematosus.
Collapse
Affiliation(s)
- Jason C. Klein
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ruchika Mahapatra
- School of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Gary C. Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas Texas, USA
| | - Richard C. Wang
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
18
|
Das A, Mitra A, Ghosh S, Sarkar S, Pal PK, Bandyopadhyay D, Chattopadhyay S. Arsenic-induced transition of thymic inflammation-to-fibrosis involves Stat3-Twist1 interaction: Melatonin to the rescue. Biofactors 2025; 51:e2110. [PMID: 39096306 DOI: 10.1002/biof.2110] [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: 01/11/2024] [Accepted: 07/19/2024] [Indexed: 08/05/2024]
Abstract
Groundwater arsenic is a notorious toxicant and exposure to environmentally relevant concentrations persists as a healthcare burden across the world. Arsenic has been reported to jeopardize the normal functioning of the immune system, but there are still gaps in the understanding of thymic T cell biology. Immunotoxic influence of arsenic in thymic integrity demands a potent restorative molecule. The objectives of this study were to examine key signaling cross-talks associated with arsenic-induced immune alterations in the thymus and propose melatonin as a potential candidate against immunological complications arising from arsenic exposure. Swiss albino mice were exposed to sodium arsenite (0.05 mg/L; in drinking water) and melatonin (IP:10 mg/kg BW) for 28 days. Melatonin successfully protected thymus from arsenic-mediated tissue degeneration and maintained immune homeostasis including T cell maturation and proliferation by mitigating oxidative stress through Nrf2 upregulation. Additionally, melatonin exerted ameliorative effect against arsenic-induced apoptosis and inflammation by inhibiting p53-mediated mitochondrial cell death pathway and NF-κB-p65/STAT3-mediated proinflammatory pathway, respectively. For the first time, we showed that arsenic-induced profibrotic changes were inhibited by melatonin through targeting of inflammation-associated EMT. Our findings clearly demonstrate that melatonin can be a viable and promising candidate in combating arsenic-induced immune toxicity with no collateral damage, making it an important research target.
Collapse
Affiliation(s)
- Ankur Das
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Ankan Mitra
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Sourav Ghosh
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Swaimanti Sarkar
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Palash Kumar Pal
- Department of Physiology, University of Calcutta, Kolkata, India
| | | | - Sreya Chattopadhyay
- Department of Physiology, University of Calcutta, Kolkata, India
- Centre for Research in Nanoscience and Nanotechnology (CRNN), University of Calcutta, Kolkata, India
| |
Collapse
|
19
|
Aydemir E. Immunomodulatory activity of metoclopramide HCl on murine macrophages. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03715-2. [PMID: 39673636 DOI: 10.1007/s00210-024-03715-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Metoclopramide HCl is the active ingredient of the commonly used drugs against nausea, heartburn, and acid reflux. Other than the patients with the acid reflux and stomach problems, it has been widely used in patients going through chemotherapy and radiotherapy, as well as patients suffering from migraine with stomach issues. To our knowledge, the immunomodulatory effect of metoclopramide HCl has never been tested extensively. Since patients who are going through chemotherapy and radiotherapy are prone to developing immunodeficiencies, it is imperative to analyze the activity of this active drug ingredient on the mammalian macrophages. For this purpose, the mammalian macrophages were tested for their potential to produce the pro-inflammatory cytokines, in vitro, in the presence and absence of LPS as the stimulating agent with a range of metoclopramide concentrations. Our results suggest that metoclopramide HCl did not stimulate the macrophages by itself; but in the presence of LPS stimulation, it significantly and substantially decreased the production of the pro-inflammatory TNFα, IL-6, GMCSF, and IL12p40 cytokines. Although this study involved solely in vitro model, the results imply that in the clinical setting metoclopramide HCl might induce inflammatory reaction against potential infections.
Collapse
Affiliation(s)
- Esra Aydemir
- Department of Biomedical Engineering, Biruni University, Istanbul, 34010, Turkey.
| |
Collapse
|
20
|
Xie J, Shao Z, Li C, Zeng C, Xu B. Cuproptosis-related gene ATOX1 promotes MAPK signaling and diffuse large B-cell lymphoma proliferation via modulating copper transport. BIOMOLECULES & BIOMEDICINE 2024; 25:16-28. [PMID: 39036924 PMCID: PMC11647247 DOI: 10.17305/bb.2024.10536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/29/2024] [Accepted: 06/29/2024] [Indexed: 07/23/2024]
Abstract
Diffuse Large B-cell lymphoma (DLBCL) is a common subtype of non-Hodgkin lymphoma, highlighting the importance of studying susceptibility genes to develop personalized treatment strategies. While cuproptosis, caused by high levels of copper ions induced by ionophores, has been shown to affect cancer survival, its specific role in lymphoma is not yet clear. To investigate the involvement of upregulation-related genes in DLBCL, we employed bioinformatics techniques. Specifically, we analyzed the differentially expressed genes (DEGs) in the GSE25638 dataset using Weighted Gene Co-expression Network Analysis (WGCNA) and performed functional enrichment analysis. By building a Protein-Protein Interaction (PPI) network, candidate genes were identified. Gene Set Enrichment Analysis (GSEA) and Receiver Operating Characteristic (ROC) curve analysis were used to confirm the clinical diagnostic use of these genes. The effects of Antioxidant 1 (ATOX1) knockdown, CuCl2, and DCAC50 treatments on DLBCL cells and the activation of the Mitogen-Activated Protein Kinase (MAPK) pathway were investigated by conducting in vitro experiments. Bioinformatics and in vitro experiments confirmed elevated expression of ATOX1 in DLBCL cells and tumor samples. ATOX1 knockdown led to decreased cell proliferation and G2 cell cycle arrest in vitro. Additionally, Phosphorylated Extracellular Signal-Regulated Kinases 1 and 2 (P-ERK1/2) protein levels within the MAPK pathway were reduced as a result of ATOX1 knockdown, but these levels were recovered by CuCl2. Treatment with DCAC50 showed a dose-dependent antiproliferative effect in DLBCL cells, which was strengthened by ATOX1 knockdown. Our study demonstrated that ATOX1 may be important in DLBCL via controlling the MAPK pathway through copper transport, providing new insights into potential therapeutic strategies for DLBCL.
Collapse
Affiliation(s)
- Junjie Xie
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, China
| | - Zhixiong Shao
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, China
| | - Changjie Li
- Jiefang Kairui Medical Laboratory, Shanghai, China
| | - Cheng Zeng
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, China
| | - Biao Xu
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
| |
Collapse
|
21
|
Alkafaas SS, Khedr SA, ElKafas SS, Hafez W, Loutfy SA, Sakran M, Janković N. Targeting JNK kinase inhibitors via molecular docking: A promising strategy to address tumorigenesis and drug resistance. Bioorg Chem 2024; 153:107776. [PMID: 39276490 DOI: 10.1016/j.bioorg.2024.107776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/13/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024]
Abstract
Among members of the mitogen-activated protein kinase (MAPK) family, c-Jun N-terminal kinases (JNKs) are vital for cellular responses to stress, inflammation, and apoptosis. Recent advances have highlighted their important implications in cancer biology, where dysregulated JNK signalling plays a role in the growth, progression, and metastasis of tumors. The present understanding of JNK kinase and its function in the etiology of cancer is summarized in this review. By modifying a number of downstream targets, such as transcription factors, apoptotic regulators, and cell cycle proteins, JNKs exert diverse effects on cancer cells. Apoptosis avoidance, cell survival, and proliferation are all promoted by abnormal JNK activation in many types of cancer, which leads to tumor growth and resistance to treatment. JNKs also affect the tumour microenvironment by controlling the generation of inflammatory cytokines, angiogenesis, and immune cell activity. However, challenges remain in deciphering the context-specific roles of JNK isoforms and their intricate crosstalk with other signalling pathways within the complex tumor environment. Further research is warranted to delineate the precise mechanisms underlying JNK-mediated tumorigenesis and to develop tailored therapeutic strategies targeting JNK signalling to improve cancer management. The review emphasizes the role of JNK kinases in cancer biology, as well as their potential as pharmaceutical targets for precision oncology therapy and cancer resistance. Also, this review summarizes all the available promising JNK inhibitors that are suggested to promote the responsiveness of cancer cells to cancer treatment.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, 31527, Egypt.
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Sara Samy ElKafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt; Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, Russia
| | - Wael Hafez
- NMC Royal Hospital, 16th St - Khalifa City - SE-4 - Abu Dhabi, United Arab Emirates; Department of Internal Medicine, Medical Research and Clinical Studies Institute, The National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate 12622, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Sakran
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Nenad Janković
- Institute for Information Technologies Kragujevac, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia.
| |
Collapse
|
22
|
Sun J, Yuan H, Yu Y, Li A, Zhao Z, Tang Y, Zheng F. Immunomodulatory potential of primary cilia in the skin. Front Immunol 2024; 15:1456875. [PMID: 39676858 PMCID: PMC11638010 DOI: 10.3389/fimmu.2024.1456875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Primary cilia (PC) are essential signaling hubs for proper epithelial formation and the maintenance of skin homeostasis. Found on most cells in the human body, including skin cells, PC facilitate signal transduction that allows ciliated cells to interact with the immune system via multiple pathways, helping to maintain immune system homeostasis. PC can be altered by various microenvironmental stimuli to develop corresponding regulatory functions. Both PC and ciliary signaling pathways have been shown to be involved in the immune processes of various skin lesions. However, the mechanisms by which PC regulate cellular functions and maintain immune homeostasis in tissues are highly complex, and our understanding of them in the skin remains limited. In this paper, we discuss key ciliary signaling pathways and ciliated cells in the skin, with a focus on their immunomodulatory functions. We have compiled evidence from various cells, tissues and disease models to help explore the potential immunomodulatory effects of PC in the skin and their molecular mechanisms.
Collapse
Affiliation(s)
- Jingwei Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huimin Yuan
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yanru Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Aorou Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zihe Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fengjie Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
23
|
Lin Q, Liu HM, Wu LZ, Yu DD, Hua CY, Zou Y, Jiao WE, Li XP, Chen SM. Effect and underlying mechanism of a photochemotherapy dual-function nanodrug delivery system for head and neck squamous cell carcinoma. J Transl Med 2024; 22:1043. [PMID: 39563418 DOI: 10.1186/s12967-024-05855-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND The novel nanomaterials PNA-TN (PN) and PNA-TN-Dox (PND) have been shown to have strong inhibitory effects on breast cancer; however, it is unclear whether PN and PND have anti-head and neck squamous cell carcinoma (HNSCC) activity, and their potential mechanisms of activity are unknown. So, our study aims to explore the therapeutic effects of PN and PND on HNSCC and their possible mechanisms. METHODS We used a series of phenotypic research to evaluate the effects of PN + Laser (L) and PND + L on the biological function of HNSCC cells in vitro and in vivo. We subsequently used mechanism research to examine changes in mRNA and protein expression related to apoptosis, epithelial‒mesenchymal transition (EMT), and the JNK signalling pathway. RESULTS Our study revealed that PN and PND have strong inhibitory effects on HNSCC cells both in vitro and in vivo. In vitro, PN and PND significantly inhibited the proliferation, migration, invasion and EMT ability of HNSCC cells and promoted apoptosis; the inhibitory effect in the PND + L group was significantly greater than that in the PN + L group. In vivo, both treatments led to significant reductions in tumour volume and weight. Notably, the tumour volume and weight in the PND + L group were significantly lower than those in the PN + L group. Mechanism research confirmed that PN + L activated the expression of apoptosis-related proteins and inhibited the expression of EMT-related proteins via the JNK pathway. Furthermore, the anti-HNSCC effect of PN + L was blocked after the use of a JNK pathway inhibitor. CONCLUSION Treatment with PN + L or PND + L significantly inhibited the malignant progress of HNSCC cells, and the therapeutic effect of PND + L was significantly stronger than that of PN + L. The JNK signalling pathway is a key mechanism by which PN exerts its anti-HNSCC activity.
Collapse
Affiliation(s)
- Qian Lin
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Hui-Min Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238, Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Li-Zhi Wu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Dong-Dong Yu
- Department of Oncology, Renmin Hospital of Wuhan University, No. 238, Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Cheng-Yu Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - You Zou
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China
| | - Xiang-Pan Li
- Department of Oncology, Renmin Hospital of Wuhan University, No. 238, Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China.
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, P. R. China.
| |
Collapse
|
24
|
Shahpari M, Hashemi M, Younesirad T, Hasanzadeh A, Mosanne MM, Ahmadifard M. The functional roles of competitive endogenous RNA (ceRNA) networks in apoptosis in human cancers: The circRNA/miRNA/mRNA regulatory axis and cell signaling pathways. Heliyon 2024; 10:e37089. [PMID: 39524849 PMCID: PMC11546195 DOI: 10.1016/j.heliyon.2024.e37089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Circular RNAs are noncoding RNAs with circular conformation mainly due to backsplicing event. CircRNAs can potentially impact cell biological processes by interacting with cell signaling pathways. Numerous circRNAs have been found to be aberrantly expressed in a variety of cancers. These RNAs can act as ceRNA (competitive endogenous RNA) by sponging certain miRNAs to form circRNA/miRNA/mRNA networks. Dysregulation of ceRNA networks may lead to dysfunctions in various cell pathways, which modulate apoptosis-associated genes and ultimately result in cancer progression. Since disruption of apoptosis is one of the leading causes of cancer development, one approach for cancer treatment is to drive cells toward apoptosis. In this review, we present a summary of studies on the role of ceRNA networks in cellular signaling pathways that regulate apoptosis; these networks are suggested to be potential biomarkers for cancer treatment.
Collapse
Affiliation(s)
| | | | - Tayebeh Younesirad
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Aida Hasanzadeh
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad mahdi Mosanne
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohamadreza Ahmadifard
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
25
|
Shi S, Tian X, Gong Y, Sun M, Liu J, Zhang J, Liu Y, Li L, Jiang S. Pivotal role of JNK protein in the therapeutic efficacy of parthenolide against breast cancer: Novel and comprehensive evidences from network pharmacology, single-cell RNA sequencing and metabolomics. Int J Biol Macromol 2024; 279:135209. [PMID: 39244135 DOI: 10.1016/j.ijbiomac.2024.135209] [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: 06/12/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
This study aimed to evaluate the efficacy and therapeutic mechanism of parthenolide (PTL) in breast cancer (BC) through a comprehensive strategy integrating network pharmacology, single-cell RNA sequencing (scRNA-seq) and metabolomics. In network pharmacology, 70 therapeutic targets were identified, of which 16 core targets were filtered out through seven classical algorithms of Cytohubba plugin. Additionally, the hub module of PPI network was extracted using MCODE plugin. Molecular docking and molecular dynamics simulation showed a potent binding affinity between PTL and JNK, subsequently validated by MST and SPR assays. Further, Mendelian randomization analysis indicated that JNK was causally associated with BC. GO and KEGG enrichment analyses revealed that PTL counteracted BC via promoting ROS generation, inducing apoptosis and suppressing proliferation, which potentially involved the coordinated regulation of MAPK and FoxO1 pathways. Moreover, ssGSEA and scRNA-seq analysis suggested that PTL may act on T cell immune microenvironment of BC. Subsequently, these bioinformatics-based predictions were experimentally validated using in-vitro and in-vivo models. Finally, metabolome profiling unveiled that PTL remodeled the glycine, serine and threonine metabolism as well as biosynthesis of unsaturated fatty acids, and thereby contributed to BC inhibition. From molecular, immune and metabolic perspectives, this study not only provided a unique insight into the mechanistic details of PTL against BC, but also proposed a novel promising therapeutic strategy for BC.
Collapse
Affiliation(s)
- Shulong Shi
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China; Department of Clinical Medicine, Jining Medical University, Jining 272013, China; Cisen Pharmaceutical Co., Ltd, Jining 272000, China; School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China
| | - Yining Gong
- Department of Clinical Medicine, Jining Medical University, Jining 272013, China
| | - Mingliang Sun
- Department of Endocrinology, Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250000, China
| | - Juan Liu
- Shandong Rehabilitation Hospital, Jinan 250000, China
| | - Jiaqi Zhang
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China
| | - Yaping Liu
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Luning Li
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China.
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China.
| |
Collapse
|
26
|
Huang R, Zhang B, Ye W, Tang Z, Zheng Q. IL-4 Downregulates PD-L1 Level Via SOCS1 Upregulation-Induced JNK Deactivation to Enhance Antitumor Immunity in In Vitro Colorectal Cancer. J Interferon Cytokine Res 2024; 44:486-495. [PMID: 39364618 DOI: 10.1089/jir.2024.0110] [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] [Indexed: 10/05/2024] Open
Abstract
Interleukin-4 (IL-4) controls cell growth and immune system regulation in tumorigenesis and can inhibit the growth of colon cancer cell lines, but the possible mechanism is unclear. In this study, we investigated the possible mechanism of IL-4 in colorectal cancer (CRC) through in vitro experiments. CRC cells received treatment with IL-4 (50 ng/mL), investigating the suppressor of cytokine signaling 1 (SOCS1)-related mechanism underlying the role of IL-4 in the progression and immunosuppression of CRC. The malignant processes of CRC cells and CD8+T cell-mediated immune response in CRC cells were determined by CCK-8, Transwell, wound healing, and flow cytometry assays. Programmed death ligand 1 (PD-L1), SOCS1 expressions, and c-Jun N-terminal kinase (JNK) activation in CRC cells were analyzed by quantitative reverse transcription polymerase chain reaction and/or Western blot. IL-4 repressed the malignant processes, yet promoted the apoptosis of CRC cells. Besides, IL-4 downregulated PD-L1 level, upregulated SOCS1 level, and restrained JNK activation in CRC cells, while enhancing CRC cell-killing effect of CD8+T cells. IL-4-induced effects on the aforementioned malignant processes of CRC cells and the killing effect of CD8+T cells toward CRC cells were all reversed when SOCS1 was knocked down in the CRC cells. IL-4 downregulates PD-L1 level via SOCS1 upregulation-induced JNK deactivation to enhance antitumor immunity in in vitro CRC. The study provides a theoretical basis for the clinical application of IL-4 in antitumor immunity in CRC.
Collapse
Affiliation(s)
- Ruiyan Huang
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Baofan Zhang
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Wanchun Ye
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Zhongjie Tang
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Qingsong Zheng
- The Second Department of Oncology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
27
|
Li L, Chang J, Xu Z, Chu L, Zhang J, Xing Q, Bao Z. Functional allocation of Mitogen-activated protein kinases (MAPKs) unveils thermotolerance in scallop Argopecten irradians irradians. MARINE ENVIRONMENTAL RESEARCH 2024; 202:106750. [PMID: 39293275 DOI: 10.1016/j.marenvres.2024.106750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Global warming has significantly impacted agriculture, particularly in animal husbandry and aquaculture industry. Rising ocean temperatures due to global warming are severely affecting shellfish production, necessitating an understanding of how shellfish cope with thermal stress. The mitogen-activated protein kinases (MAPK) signaling pathway plays a crucial role in cell growth, differentiation, adaptation to environmental stress, inflammatory response, and managing high temperature stress. To investigate the function of MAPKs in bay scallops, a comparative genomics and bioinformatics approach identified three MAPK genes: AiERK, Aip38, and AiJNK. Structural and phylogenetic analyses of these proteins were conducted to determine their evolutionary relationships. Spatiotemporal expression patterns were examined at different developmental stages and in various tissues of healthy adult scallops. Additionally, the expression regulation of these genes was studied in selected tissues (hemocyte, gill, heart, mantle) following exposure to high temperatures (32 °C) for different durations (0 h, 6 h, 12 h, 24 h, 3 d, 6 d, 10 d). The spatiotemporal expressions of AiMAPKs were ubiquitous, with significant increases in AiERK expression observed at the umbo larval stage (3.09-fold), while Aip38 and AiJNK were identified as potential maternal effect genes. In adult scallops, different gene expression patterns of AiMAPKs were observed across eight tissues, with high expressions in the foot and gill, and lower expressions in the striated muscle. Following high temperature stress, AiMAPKs expressions in the gill and mantle were mainly up-regulated, while in the hemocyte, they were primarily down-regulated. These findings indicate time- and tissue-dependent expression patterns with functional allocation in response to different thermal durations. This study enhances our understanding of the function and evolution of AiMAPKs genes in shellfish and provides a theoretical basis for elucidating the energy regulation mechanism of bay scallops in response to high temperature stress.
Collapse
Affiliation(s)
- Linshu Li
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Jiaxi Chang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Zhaosong Xu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Longfei Chu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Junhao Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Qiang Xing
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| |
Collapse
|
28
|
Hsiao YT, Yoshida Y, Okuda S, Abe M, Mizuno S, Takahashi S, Nakagami H, Morishita R, Kamimura K, Terai S, Aung TM, Li J, Furihata T, Tang JY, Walsh K, Ishigami A, Minamino T, Shimizu I. PCPE-1, a brown adipose tissue-derived cytokine, promotes obesity-induced liver fibrosis. EMBO J 2024; 43:4846-4869. [PMID: 39160276 PMCID: PMC11535236 DOI: 10.1038/s44318-024-00196-0] [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: 10/20/2023] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH, previously termed non-alcoholic steatohepatitis (NASH)), is a major complication of obesity that promotes fatty liver disease. MASH is characterized by progressive tissue fibrosis and sterile liver inflammation that can lead to liver cirrhosis, cancer, and death. The molecular mechanisms of fibrosis in MASH and its systemic control remain poorly understood. Here, we identified the secreted-type pro-fibrotic protein, procollagen C-endopeptidase enhancer-1 (PCPE-1), as a brown adipose tissue (BAT)-derived adipokine that promotes liver fibrosis in a murine obesity-induced MASH model. BAT-specific or systemic PCPE-1 depletion in mice ameliorated liver fibrosis, whereas, PCPE-1 gain of function in BAT enhanced hepatic fibrosis. High-calorie diet-induced ER stress increased PCPE-1 production in BAT through the activation of IRE-1/JNK/c-Fos/c-Jun signaling. Circulating PCPE-1 levels are increased in the plasma of MASH patients, suggesting a therapeutic possibility. In sum, our results uncover PCPE-1 as a novel systemic control factor of liver fibrosis.
Collapse
Affiliation(s)
- Yung Ting Hsiao
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, 564-8565, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Advanced Senotherapeutics, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Ryuichi Morishita
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of General Medicine, Niigata University School of Medicine, Niigata, 951-8510, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Tin May Aung
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, 564-8565, Japan
| | - Ji Li
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China
| | - Takaaki Furihata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Jing Yuan Tang
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Kenneth Walsh
- Division of Cardiovascular Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, 564-8565, Japan.
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan.
| |
Collapse
|
29
|
Jalil A, Donate MM, Mattei J. Exploring resistance to immune checkpoint inhibitors and targeted therapies in melanoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:42. [PMID: 39534873 PMCID: PMC11555183 DOI: 10.20517/cdr.2024.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Melanoma is the most aggressive form of skin cancer, characterized by a poor prognosis, and its incidence has risen rapidly over the past 30 years. Recent therapies, notably immunotherapy and targeted therapy, have significantly improved the outcome of patients with metastatic melanoma. Previously dismal five-year survival rates of below 5% have shifted to over 50% of patients surviving the five-year mark, marking a significant shift in the landscape of melanoma treatment and survival. Unfortunately, about 50% of patients either do not respond to therapy or experience early or late relapses following an initial response. The underlying mechanisms for primary and secondary resistance to targeted therapies or immunotherapy and relapse patterns remain not fully identified. However, several molecular pathways and genetic factors have been associated with melanoma resistance to these treatments. Understanding these mechanisms paves the way for creating novel treatments that can address resistance and ultimately enhance patient outcomes in melanoma. This review explores the mechanisms behind immunotherapy and targeted therapy resistance in melanoma patients. Additionally, it describes the treatment strategies to overcome resistance, which have improved patients' outcomes in clinical trials and practice.
Collapse
Affiliation(s)
- Anum Jalil
- Department of Medicine, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| | - Melissa M Donate
- Long School of Medicine, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| | - Jane Mattei
- Department of Hematology Oncology, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| |
Collapse
|
30
|
Samrit T, Changklungmao N, Sangpairoj K, Buddawong A, Kueakhai P, Chuanboon K, Sobhon P, Pranweerapaiboon K. Ethanolic extract of Parkia speciosa pods exhibits antioxidant and anti-inflammatory properties in lipopolysaccharide-induced murine macrophages by inhibiting the p38 MAPK pathway. Heliyon 2024; 10:e39641. [PMID: 39506962 PMCID: PMC11538774 DOI: 10.1016/j.heliyon.2024.e39641] [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: 05/31/2024] [Revised: 09/13/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
Background Parkia speciosa (PS) is commonly used in Southeast Asian cuisine and traditional medicine to treat diabetes, hypertension, dermatitis, and kidney diseases. PS has emerged as a subject of interest because of its potential antioxidation and anti-inflammatory properties. However, despite its historically long and wide usage, a comprehensive investigation of these properties in PS pods (PSp) have not been conducted. Aims of this study This study aimed to identify the phytochemical compounds in the ethanolic extract of PSp collected from Southern Thailand and assess whether PSp exhibit antioxidant properties and mitigate inflammation in a lipopolysaccharide (LPS)-induced RAW264.7 model. Materials and methods The ethanolic extract of PSp was comprehensively analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and gas chromatography-mass spectrometry (GC/MS) to identify its phytochemical constituents. To assess the antioxidant activity, 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azino-bis-(3-ethylbenzothiazoline-6-sulfonic) acid (ABTS) assays were performed, and cytotoxicity was evaluated using the MTT assay. The effect of PSp on reactive nitrogen and oxygen species (RNS and ROS) was determined using a nitric oxide (NO) assay, and its effect on pro-inflammatory cytokines was assessed using enzyme-linked immunosorbent assay (ELISA) and real-time quatitvative polymerase chain reaction (qPCR). Morphological changes following treatment were observed using a microscope. Western blot analysis was performed to quantify MAPK pathway expression. Results PSp contain polyphenols, phytosterols, triterpenes, oxaloacetic acid, and unsaturated fatty acids. PSp demonstrated high antioxidant potential in scavenging free radicals and exhibited no cytotoxic effects on macrophages. Moreover, PSp effectively reduced NO release and inhibited pro-inflammatory cytokines such as IL1-β, TNF-α, and IL-6. PSp treatment induced notable morphological changes in macrophages, characterized by an increase in cell size and the presence of intracellular vacuoles. In addition, Western blot analysis showed the selective suppressive effect of PSp on the p38-MAPK pathway. Conclusion PSp possess strong antioxidant and anti-inflammatory properties, making it a potential therapeutic agent for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Tepparit Samrit
- Food Bioactive Compounds Research Unit, Faculty of Allied Health Sciences, Burapha University, Chonburi, 20131, Thailand
| | - Narin Changklungmao
- Food Bioactive Compounds Research Unit, Faculty of Allied Health Sciences, Burapha University, Chonburi, 20131, Thailand
| | - Kant Sangpairoj
- Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani, 12120, Thailand
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Aticha Buddawong
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Pornanan Kueakhai
- Food Bioactive Compounds Research Unit, Faculty of Allied Health Sciences, Burapha University, Chonburi, 20131, Thailand
| | - Kititpong Chuanboon
- Mahidol University-Frontier Research Facility, Research Management and Development Division, Office of the President, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Kanta Pranweerapaiboon
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| |
Collapse
|
31
|
Zheng S, Qi W, Xue T, Zao X, Xie J, Zhang P, Li X, Ye Y, Liu A. Chinese medicine in the treatment of chronic hepatitis B: The mechanisms of signal pathway regulation. Heliyon 2024; 10:e39176. [PMID: 39640799 PMCID: PMC11620126 DOI: 10.1016/j.heliyon.2024.e39176] [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: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Chronic hepatitis B (CHB) is a chronic inflammatory disease of the liver caused by infection with the hepatitis B virus (HBV), which in later stages can lead to the development of end-stage liver diseases such as cirrhosis and hepatocellular carcinoma in severe cases, jeopardizing long-term quality of life, with a poor prognosis, and placing a serious financial burden on many families around the world. The pathogenesis of the disease is complex and closely related to the immune function of the body, which has not yet been fully elucidated. The development of chronic hepatitis B is closely related to the involvement of various signaling pathways, such as JAK/STAT, PI3K/Akt, Toll-like receptor, NF-κB and MAPK signaling pathways. A large number of studies have shown that Chinese medicine has obvious advantages in anti-hepatitis B virus, and it can effectively treat the disease by modulating relevant signaling pathways, strengthening immune resistance and defense, and inhibiting inflammatory responses, and certain research progress has been made, but there is still a lack of a comprehensive review on the modulation of relevant signaling pathways in Chinese medicine for the treatment of CHB. Therefore, this article systematically combed and elaborated the relevant literature on the modulation of relevant signaling pathways by traditional Chinese medicine in recent years, with a view to providing new ideas for the treatment of CHB and further drug development.
Collapse
Affiliation(s)
- Shihao Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Wenying Qi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Tianyu Xue
- Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, 050000, China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
| | - Jinchi Xie
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Peng Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Xiaoke Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yongan Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Aimin Liu
- Shangzhuang Township Community Health Service Center, Beijing, 100094, China
| |
Collapse
|
32
|
Mahmoud ME, Ali A, Farooq M, Isham IM, Suhail SM, Herath-Mudiyanselage H, Rahimi R, Abdul-Careem MF. Cyclooxygenase-2/prostaglandin E2 pathway orchestrates the replication of infectious bronchitis virus in chicken tracheal explants. Microbiol Spectr 2024; 12:e0040724. [PMID: 39472003 PMCID: PMC11619240 DOI: 10.1128/spectrum.00407-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/09/2024] [Indexed: 12/08/2024] Open
Abstract
In this study, we investigated the localized pathogenesis of infectious bronchitis virus (IBV) in chicken tracheal organ cultures (TOCs), focusing on the role of inducible cyclooxygenase (COX-2). Two divergent IBV strains, respiratory Connecticut (Conn) A5968 and nephropathogenic Delmarva (DMV)/1639, were studied at 6, 12, 24, and 48 hours post-infection (hpi). Various treatments including exogenous prostaglandin (PGE)2, a selective COX-2 antagonist (SC-236), and inhibitors of PGE2 receptors and Janus kinase (JAK) were administered. IBV genome load and antigen expression were quantified using real-time quantitative PCR and immunohistochemistry. COX-2, interferon (IFN)-α, IFN-β, interleukin (IL)-1β, IL-6, and inducible nitric oxide synthase (iNOS) expressions were measured, along with PGE2 and COX-2 concentrations. IBV genome load and protein expression peaked at 12 and 24 hpi, respectively. Conn A5968-infected TOCs exhibited continuous COX-2 expression for up to 24 hpi, extended PGE2 production up to 48 hpi, and reduced inflammatory cytokine expression. In contrast, DMV/1639-infected TOCs displayed heightened inflammatory cytokine expression, brief COX-2 expression, and PGE2 production. Treatment with IFN-γ, SC-236, PGE2 receptor inhibitors, or JAK inhibitors reduced IBV infection and lesion scores, whereas exogenous PGE2 or IFN-γ pretreatment with a JAK-2 inhibitor augmented infection. These findings shed light on the innate immune regulation of IBV infection in the trachea, highlighting the involvement of the COX-2/PGE2 pathway. IMPORTANCE Understanding the localized pathogenesis of infectious bronchitis virus (IBV) within the trachea of chickens is crucial for developing effective control strategies against this prevalent poultry pathogen. This study sheds light on the role of inducible cyclooxygenase (COX-2) and prostaglandin (PGE)2 in IBV pathogenesis using chicken tracheal organ culture (TOC) models. The findings reveal distinct patterns of COX-2 expression, PGE2 production, and immune responses associated with different IBV strains, highlighting the complexity of host-virus interactions. Furthermore, the identification of specific inhibitors targeting the COX-2/PGE2 pathway and Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway provides potential therapeutic avenues for mitigating IBV infection in poultry. Overall, this study contributes to our understanding of the innate immune regulation of IBV infection within the trachea, laying the groundwork for the development of targeted interventions to control IBV outbreaks in poultry populations.
Collapse
Affiliation(s)
- Motamed Elsayed Mahmoud
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Animal Husbandry, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Ahmed Ali
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Muhammad Farooq
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ishara M. Isham
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sufna M. Suhail
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Ryan Rahimi
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
33
|
Wen Y, Zheng Y, Hua S, Li T, Bi X, Lu Q, Li M, Sun S. Mechanisms of Bone Morphogenetic Protein 2 in Respiratory Diseases. Curr Allergy Asthma Rep 2024; 25:1. [PMID: 39466470 DOI: 10.1007/s11882-024-01181-7] [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] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE OF REVIEW Bone morphogenetic protein 2 (BMP2) belongs to the transforming growth factor-β (TGF-β) superfamily and plays an important role in regulating embryonic development, angiogenesis, osteogenic differentiation, tissue homeostasis, and cancer invasion. Increasing studies suggest BMP2 is involved in several respiratory diseases. This study aimed to review the role and mechanisms of BMP2 in respiratory diseases. RECENT FINDINGS BMP2 signaling pathway includes the canonical and non-canonical signaling pathway. The canonical signaling pathway is the BMP2-SMAD pathway, and the non-canonical signaling pathway includes mitogen-activated protein kinase (MAPK) pathway and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway. The BMP2 is related to pulmonary hypertension (PH), lung cancer, pulmonary fibrosis (PF), asthma, and chronic obstructive pulmonary disease (COPD). BMP2 inhibits the proliferation of pulmonary artery smooth muscle cells (PASMCs), promotes the apoptosis of PASMCs to reduce pulmonary vascular remodeling in PH, which is closely related to the canonical and non-canonical pathway. In addition, BMP2 stimulates the proliferation and migration of cells to promote the occurrence, colonization, and metastasis of lung cancer through the canonical and the non-canonical pathway. Meanwhile, BMP2 exert anti-fibrotic function in PF through canonical signaling pathway. Moreover, BMP2 inhibits airway inflammation to maintain airway homeostasis in asthma. However, the signaling pathways involved in asthma are poorly understood. BMP2 inhibits the expression of ciliary protein and promotes squamous metaplasia of airway epithelial cells to accelerate the development of COPD. In conclusion, BMP2 may be a therapeutic target for several respiratory diseases.
Collapse
Affiliation(s)
- Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Yuanyuan Zheng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Tongfen Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Xiaoqing Bi
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Qiongfen Lu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Min Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China.
| |
Collapse
|
34
|
Liu C, Wei J, Wang X, Zhao Q, Lv J, Tan Z, Xin Y, Jiang X. Radiation-induced skin reactions: oxidative damage mechanism and antioxidant protection. Front Cell Dev Biol 2024; 12:1480571. [PMID: 39450273 PMCID: PMC11500330 DOI: 10.3389/fcell.2024.1480571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
According to official statistics, cancer remains the main reason of death and over 50% of patients with cancer receive radiotherapy. However, adverse consequences after radiation exposure like radiation-induced skin reactions (RISR) have negative or even fatal impact on patients' quality of life (QoL). In this review we summarize the mechanisms and managements of RISRs, a process that involve a variety of extracellular and intracellular signals, among which oxidative stress (OS) are now commonly believed to be the initial part of the occurrence of all types of RISRs. As for the management of RISRs, traditional treatments have been widely used but without satisfying outcomes while some promising therapeutic strategies related to OS still need further researches. In the context we discuss how OS leads to the happening of RISRs of different types, hoping it can shed some light on the exploration of new countermeasures.
Collapse
Affiliation(s)
- Chuchu Liu
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Jinlong Wei
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Xuanzhong Wang
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Qin Zhao
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Jincai Lv
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Zining Tan
- Key Laboratory of Pathobiology, Ministry of Education and College of Basic Medical Science, Jilin University, Changchun, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education and College of Basic Medical Science, Jilin University, Changchun, China
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| |
Collapse
|
35
|
Zhou T, Zhang C, Wang X, Lin J, Yu J, Liang Y, Guo H, Yang M, Shen X, Li J, Shi R, Wang Y, Yang J, Shu Z. Research on traditional Chinese medicine as an effective drug for promoting wound healing. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118358. [PMID: 38763370 DOI: 10.1016/j.jep.2024.118358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/26/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The incidence of skin trauma is high and the repair process is complex, often leading to poor healing and other issues, which can result in significant economic and social burdens. Traditional Chinese medicine (TCM) is a valuable resource with proven effectiveness and safety in wound repair, widely utilized in clinical practice. A systematic analysis of wound healing with a focus on TCM research progress holds both academic and clinical importance. AIM OF THE REVIEW This article reviews the research progress of TCM in promoting wound healing, and provides basic data for the development of innovative drugs that promote wound healing. MATERIALS AND METHODS This article provides a review of the literature from the past decade and conducts a thorough analysis of various databases that contain reports on the use of TCM for wound repair. The data for this systematic research was gathered from electronic databases including CNKI, SciFinder, and PubMed. The study explores and summarizes the research findings and patterns by creating relevant charts. RESULTS This study reviewed the mechanism of wound healing, experimental TCM methods to promote wound healing, the theory and mode of action of TCM to promote wound healing, the active ingredients of TCM that promote wound healing, the efficacy of TCM formulae to promote wound healing, and the potential toxicity of TCM and its antidotes. This study enriched the theory of TCM in promoting wound healing. CONCLUSION Skin wound healing is a complex process that can be influenced by various internal and external factors. This article offers a theoretical foundation for exploring and utilizing TCM resources that enhance wound repair. By analyzing a range of TCM that promote wound healing, the article highlights the clinical importance and future potential of these medicines in promoting wound healing.
Collapse
Affiliation(s)
- Tong Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Chongyang Zhang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Xiao Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Jiazi Lin
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Jiamin Yu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Yefang Liang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Huilin Guo
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Mengru Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Xuejuan Shen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Jianhua Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Ruixiang Shi
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China
| | - Yi Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Ji Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Zunpeng Shu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, PR China.
| |
Collapse
|
36
|
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
|
37
|
Yin L, Zhang E, Mao T, Zhu Y, Ni S, Li Y, Liu C, Fang Y, Ni K, Lu Y, Li H, Zhou M, Hu Q. Macrophage P2Y 6R activation aggravates psoriatic inflammation through IL-27-mediated Th1 responses. Acta Pharm Sin B 2024; 14:4360-4377. [PMID: 39525587 PMCID: PMC11544167 DOI: 10.1016/j.apsb.2024.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/07/2024] [Accepted: 06/11/2024] [Indexed: 11/16/2024] Open
Abstract
Purinergic signaling plays a causal role in the modulation of immune inflammatory response in the course of psoriasis, but its regulatory mechanism remains unclear. As a member of purinoceptors, P2Y6R mainly distributed in macrophages was significantly up-expressed in skin lesions from patients with psoriasis in the present study. Here, the severity of psoriasis was alleviated in imiquimod-treated mice with macrophages conditional knockout of P2Y6R, while the cell-chat algorithm showed there was a correlation between macrophage P2Y6R and Th1 cells mediated by IL-27. Mechanistically, P2Y6R enhanced PLC β /p-PKC/MAPK activation to induce IL-27 release dependently, which subsequently regulated the differentiation of Th1 cells, leading to erythematous and scaly plaques of psoriasis. Interestingly, we developed a novel P2Y6R inhibitor FS-6, which bonds with the ARG266 side chain of P2Y6R, exhibited remarkable anti-psoriasis effects targeting P2Y6R. Our study provides insights into the role of P2Y6R in the pathogenesis of psoriasis and suggests its potential as a target for the development of therapeutic interventions. A novel P2Y6R inhibitor FS-6 could be developed as an anti-psoriasis drug candidate for the clinic.
Collapse
Affiliation(s)
- Li Yin
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Enming Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tianqi Mao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Yifan Zhu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Shurui Ni
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yehong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chunxiao Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yafei Fang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Kexin Ni
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yuhe Lu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Mengze Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qinghua Hu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
38
|
Feng G, Yang X, Shuai W, Wang G, Ouyang L. Update on JNK inhibitor patents: 2015 to present. Expert Opin Ther Pat 2024; 34:907-927. [PMID: 39223788 DOI: 10.1080/13543776.2024.2400167] [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: 04/10/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION c-Jun N-terminal kinase (JNK) regulates various biological processes through the phosphorylation cascade and is closely associated with numerous diseases, including inflammation, cardiovascular diseases, and neurological disorders. Therefore, JNKs have emerged as potential targets for disease treatment. AREAS COVERED This review compiles the patents and literatures concerning JNK inhibitors through retrieving relevant information from the SciFinder, Google Patents databases, and PubMed from 2015 to the present. It summarizes the structure-activity relationship (SAR) and biological activity profiles of JNK inhibitors, offering valuable perspectives on their potential therapeutic applications. EXPERT OPINION The JNK kinase serves as a novel target for the treatment of neurodegenerative disorders, pulmonary fibrosis, and other illnesses. A variety of small-molecule inhibitors targeting JNKs have demonstrated promising therapeutic potential in preclinical studies, which act upon JNK kinases via distinct mechanisms, encompassing traditional ATP competitive inhibition, covalent inhibition, and bidentate inhibition. Among them, several JNK inhibitors from PregLem SA, Celegene SA, and Xigen SA have accomplished the early stage of clinical trials, and their results will guide the development and indications of future JNK inhibitors.
Collapse
Affiliation(s)
| | | | | | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and West China second Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and West China second Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Abdel-Maksoud MS, Nasser SA, Hassan RM, Abd-Allah WH. Anticancer and anti-inflammatory effects of novel ethyl pyrazole derivatives having sulfonamide terminal moiety. Bioorg Chem 2024; 153:107825. [PMID: 39317036 DOI: 10.1016/j.bioorg.2024.107825] [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: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024]
Abstract
In the present work, a new series of ethyl pyrazole-containing compounds with side sulphonamide moiety was designed and synthesized. The new derivatives were divided into four groups based on the linker between the sulphonamide and pyridine ring attached to position 4 of the pyrazole ring and the substitution on the phenyl ring at position 3 of the same ring. The linker could be ethyl or propyl linkers. The phenyl ring is substituted with a methoxy group or hydroxyl group at position 3. The aim compounds were tested for their JNK1, JNK2, JNK3, and BRAF(V600E) activities. Compounds 23b, 23c, and 23d showed the highest activity with nanomolar IC50s. The most potent compound over JNK1 was 23d with an IC502 nM. While compound 23c was the most potent over JNK2 with an IC5057 nM. Finally, compound 23b was the most potent over JNK2 and BRAF(V600E) with IC50s of125 nM and 98 nM, respectively. After obtaining kinase inhibitory activity, the compounds were submitted to NCI to test their activity over different cell lines. Compound 23b showed the highest activity over most tested cell lines. In the second part of the present study, the final target compounds were tested for their anti-inflammatory effect. The anti-inflammatory effect of the new final compounds was performed by measuring their ability to inhibit inducible nitric oxide release and prostaglandin E2 production inhibition. Compound 23c showed the highest activity regarding nitric oxide release with IC50 0.63 μM, while compound 21d had the highest activity regarding prostaglandin E2 production with IC50 0.52 μM. The effect of the most potent compounds was tested by western blot against iNOS, COX-1, and COX-2.
Collapse
Affiliation(s)
- Mohammed S Abdel-Maksoud
- Medicinal &Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), P.O. 12622, Dokki, Giza, Egypt.
| | - Shaimaa A Nasser
- Medicinal &Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), P.O. 12622, Dokki, Giza, Egypt
| | - Rasha M Hassan
- Medicinal &Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), P.O. 12622, Dokki, Giza, Egypt
| | - Walaa H Abd-Allah
- Pharmaceutical Chemistry Department, Collage of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, P.O. 77, 6th of October City, Giza, Egypt
| |
Collapse
|
40
|
Turley J, Robertson F, Chenchiah IV, Liverpool TB, Weavers H, Martin P. Deep learning reveals a damage signalling hierarchy that coordinates different cell behaviours driving wound re-epithelialisation. Development 2024; 151:dev202943. [PMID: 39177163 PMCID: PMC11449448 DOI: 10.1242/dev.202943] [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: 04/10/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
One of the key tissue movements driving closure of a wound is re-epithelialisation. Earlier wound healing studies describe the dynamic cell behaviours that contribute to wound re-epithelialisation, including cell division, cell shape changes and cell migration, as well as the signals that might regulate these cell behaviours. Here, we have used a series of deep learning tools to quantify the contributions of each of these cell behaviours from movies of repairing wounds in the Drosophila pupal wing epithelium. We test how each is altered after knockdown of the conserved wound repair signals Ca2+ and JNK, as well as after ablation of macrophages that supply growth factor signals believed to orchestrate aspects of the repair process. Our genetic perturbation experiments provide quantifiable insights regarding how these wound signals impact cell behaviours. We find that Ca2+ signalling is a master regulator required for all contributing cell behaviours; JNK signalling primarily drives cell shape changes and divisions, whereas signals from macrophages largely regulate cell migration and proliferation. Our studies show deep learning to be a valuable tool for unravelling complex signalling hierarchies underlying tissue repair.
Collapse
Affiliation(s)
- Jake Turley
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- School of Mathematics, University of Bristol, Bristol BS8 1UG, UK
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | | | | | - Tanniemola B Liverpool
- School of Mathematics, University of Bristol, Bristol BS8 1UG, UK
- Isaac Newton Institute for Mathematical Sciences, 20 Clarkson Rd, Cambridge CB3 0EH, UK
| | - Helen Weavers
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Paul Martin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
41
|
Liu X, Cheng LT, Ye QR, Gao HC, Zhu JW, Zhao K, Liu HM, Wang YJ, Alinejad T, Zhang XH, Chen GZ. Cyy-272, an indazole derivative, effectively mitigates obese cardiomyopathy as a JNK inhibitor. Biomed Pharmacother 2024; 178:117172. [PMID: 39128188 DOI: 10.1016/j.biopha.2024.117172] [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: 05/01/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
Obesity has shown a global epidemic trend. The high-lipid state caused by obesity can maintain the heart in a prolonged low-grade inflammatory state and cause ventricular remodeling, leading to a series of pathologies, such as hypertrophy, fibrosis, and apoptosis, which eventually develop into obese cardiomyopathy. Therefore, prolonged low-grade inflammation plays a crucial role in the progression of obese cardiomyopathy, making inflammation regulation an essential strategy for treating this disease. Cyy-272, an indazole derivative, is an anti-inflammatory compound independently synthesized by our laboratory. Our previous studies revealed that Cyy-272 can exert anti-inflammatory effects by inhibiting the phosphorylation and activation of C-Jun N-terminal kinase (JNK), thereby alleviating lipopolysaccharide (LPS)-induced acute lung injury (ALI). The current study aimed to evaluate the potential of Cyy-272 to mitigate the occurrence and progression of obese cardiomyopathy through the inhibition of the JNK signaling pathway. Our results indicate that the compound Cyy-272 has encouraging therapeutic effects on obesity-induced cardiac injury. It significantly inhibits inflammation in cardiomyocytes and heart tissues induced by high lipid concentrations, further alleviating the resulting hypertrophy, fibrosis, and apoptosis. Mechanistically, the protective effect of Cyy-272 on obese cardiomyopathy can be attributed to its direct inhibition of JNK protein phosphorylation. In conclusion, we identified a novel compound, Cyy-272, capable of alleviating obese cardiomyopathy and confirmed that its effect is achieved through direct inhibition of JNK.
Collapse
Affiliation(s)
- Xin Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Lin-Ting Cheng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qian-Ru Ye
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hao-Cheng Gao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jin-Wei Zhu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kai Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hua-Min Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yun-Jie Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Tahereh Alinejad
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiu-Hua Zhang
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Gao-Zhi Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
42
|
Wang B, Ma Y, Zhang Y, Yin X. Therapeutic potential of ASK1 activators in cancer treatment: Current insights and future directions. Biomed Pharmacother 2024; 178:117214. [PMID: 39079264 DOI: 10.1016/j.biopha.2024.117214] [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: 05/16/2024] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Apoptosis signal-regulated kinase 1 (ASK1) is a member of the mitogen-activated protein kinase kinase (MAP3K) family, whose activation and regulation are intricately associated with apoptosis. ASK1 is activated in response to oxidative stress, among other stimuli, subsequently triggering downstream JNK, p38 MAPK, and mitochondria-dependent apoptotic signaling, which participate in the initiation of tumor cell apoptosis induced by various stimuli. Research has shown that ASK1 plays a crucial role in the apoptosis of lung cancer, breast cancer, and liver cancer cells. Currently, the investigation of effective ASK1 activators is a hot topic in research on tumor cell apoptosis. Synthetic compounds such as human β-defensin, triazolothiazide derivatives and heat shock protein 27 inhibitors; natural compounds such as quercetin, Laminarina japonica polysaccharide-1 peptide and theabrownin; and nanomedicines such as cerium oxide nanoparticles, magnetite FeO nanoparticles and silver nanoparticles can activate ASK1 and induce apoptosis in various tumor cells. This review extensively investigates the roles and activation mechanisms of ASK1, explores its impact on a variety of apoptotic signaling pathways, and discusses the potential therapeutic applications of various ASK1 activators in cancer treatment. In addition, this paper provides an in-depth discussion of the future development of this field and proposes a promising method for further research and clinical progress.
Collapse
Affiliation(s)
- Bo Wang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China
| | - Ying Ma
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China
| | - Yue Zhang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China.
| | - Xunzhe Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| |
Collapse
|
43
|
Lu J, Yu M, Li J. PKC-δ Promotes IL-1β-Induced Apoptosis of Rat Chondrocytes and Via Activating JNK and P38 MAPK Pathways. Cartilage 2024; 15:315-327. [PMID: 37491820 PMCID: PMC11418514 DOI: 10.1177/19476035231181446] [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: 11/26/2022] [Revised: 03/17/2023] [Accepted: 05/26/2023] [Indexed: 07/27/2023] Open
Abstract
OBJECTIVE Protein kinase C-delta (PKC-δ) is involved in apoptosis. This study aimed to establish whether PKC-δ can further promote IL-1β-induced chondrocyte apoptosis by mediating the phosphorylation of the JNK and p38 mitogen-activated protein kinase (MAPK) signaling pathways In osteoarthritis (OA). METHODS We employed chondrocyte staining to determine the extent of cartilage degeneration. PKC-δ and p38 signal expressions were used in the immunohistochemical (IHC) test and apoptosis was assayed at the TUNEL test in human osteoarthritic and controls. We stimulated rat cartilage cells using IL-1β (10 ng/ml)/rottlerin (10 μM) or lentivirus. To determine the apoptosis rate, we employed flow cytometry. The mRNA of both BCL2-related X (BAX) and cysteine aspartate protease 3 (caspase-3) could be measured via qRT-PCR. Western blot measured the protein levels of BAX, caspase-3, PKC-δ, p-JNK/JNK and p-p38/p38. RESULTS The positive rate of PKC-δ and the apoptotic rate of chondrocytes in OA were higher than controls. The manifestation of PKC-δ was positively related to the degree of cartilage degeneration, p38 protein expression, and apoptosis rate. IL-1β exposure upregulated PKC-δ expression in chondrocytes in a dose-dependent manner. Decreasing PKC-δ expression and its phosphorylation in OA can inhibit MAPK signaling pathway activation (phosphorylation) by downregulating JNK and p38 protein phosphorylation and expression. This inhibition decreases caspase-3 and BAX levels, consequently lowering the apoptosis rate in chondrocytes. CONCLUSION PKC-δ activation by IL-1β in OA promotes chondrocyte apoptosis via activation of the JNK and p38 MAPK signal pathways, thereby promoting the OA progression.
Collapse
Affiliation(s)
- Jinfeng Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Miao Yu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jia Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
44
|
Zhang C, Singla RK, Tang M, Shen B. Natural products act as game-changer potentially in treatment and management of sepsis-mediated inflammation: A clinical perspective. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155710. [PMID: 38759311 DOI: 10.1016/j.phymed.2024.155710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Sepsis, a life-threatening condition resulting from uncontrolled host responses to infection, poses a global health challenge with limited therapeutic options. Due to high heterogeneity, sepsis lacks specific therapeutic drugs. Additionally, there remains a significant gap in the clinical management of sepsis regarding personalized and precise medicine. PURPOSE This review critically examines the scientific landscape surrounding natural products in sepsis and sepsis-mediated inflammation, highlighting their clinical potential. METHODS Following the PRISMA guidelines, we retrieved articles from PubMed to explore potential natural products with therapeutic effects in sepsis-mediated inflammation. RESULTS 434 relevant in vitro and in vivo studies were identified and screened. Ultimately, 55 studies were obtained as the supporting resources for the present review. We divided the 55 natural products into three categories: those influencing the synthesis of inflammatory factors, those affecting surface receptors and modulatory factors, and those influencing signaling pathways and the inflammatory cascade. CONCLUSION Natural products' potential as game-changers in sepsis-mediated inflammation management lies in their ability to modulate hallmarks in sepsis, including inflammation, immunity, and coagulopathy, which provides new therapeutic avenues that are readily accessible and capable of undergoing rapid clinical validation and deployment, offering a gift from nature to humanity. Innovative techniques like bioinformatics, metabolomics, and systems biology offer promising solutions to overcome these obstacles and facilitate the development of natural product-based therapeutics, holding promise for personalized and precise sepsis management and improving patient outcomes. However, standardization, bioavailability, and safety challenges arise during experimental validation and clinical trials of natural products.
Collapse
Affiliation(s)
- Chi Zhang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Min Tang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China; West China School of Nursing, Sichuan University, Chengdu, PR China
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China.
| |
Collapse
|
45
|
Harisa GI, Bakheit AH, Alshehri S, Attia SM, Attia MSM. Chitosan-enclosed SLN improved SV-induced hepatocellular cell carcinoma death by modulation of IQGAP gene expression, JNK, and HDAC activities. Mol Biol Rep 2024; 51:824. [PMID: 39023688 DOI: 10.1007/s11033-024-09757-2] [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: 04/18/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a global life-threatening problem and therapeutic interventions are still encountered. IQGAP genes are involved in HCC oncogenesis. The modulatory effect of statins on the expression of IQGAP genes is still unclear. This study aims to study the effect of free SV and chitosan (CS) decorated simvastatin (SV) loaded solid lipid nanoparticles (C-SV-SLNs) on HCC mortality. METHODS AND RESULTS Plain, SV-SLN, and C-SV- SLN were prepared and characterized in terms of particle size (PS), zeta potential (ZP), and polydispersity index (PDI). The biosafety of different SLN was investigated using fresh erythrocytes, moreover, cytotoxicity was investigated using HepG2 cell lines. The effect of SLNs on IQGAPs gene expression as well as JNK, HDAC6, and HDAC8 activity was investigated using PCR and MOE-docking. The current results displayed that SV-SLNs have nanosized, negative ZP and are homogenous, CS decoration shifts the ZP of SLN into cationic ZP. Furthermore, all SLNs exhibited desirable biosafety in terms of no deleterious effect on erythrocyte integrity. SV solution and SV-SLN significantly increase the mortality of HepG2 compared to undertreated cells, however, the effect of SV-SLN is more pronounced compared to free SV. Remarkably, C-SV-SLN elicits high HepG2 cell mortality compared to free SV and SV-SLN. The treatment of HepG2 cells with SV solution, SV-SLN, or C-SV-SLN significantly upregulates the IQGAP2 gene with repression of IQGAP1 and IQGAP3 genes. MOE-docking studies revealed both SV and tenivastatin exhibit interactions with the active sites of JNK, HDAC6, and HDAC8. Moreover, tenivastatin exhibited greater interactions with magnesium and zinc compared to SV. CONCLUSIONS This research provides novel insights into the therapeutic potential of SV, SV-SLN and C-SV-SLNs in HCC treatment, modulating critical signaling cascades involving IQGAPs, JNK, and HDAC. The development of C-SV-SLNs presents a promising strategy for effective HCC therapy.
Collapse
Affiliation(s)
- Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.
| | - Ahmed H Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Samiyah Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed S M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
46
|
Ye S, Hu X, Sun S, Su B, Cai J, Jiang J. Oridonin promotes RSL3-induced ferroptosis in breast cancer cells by regulating the oxidative stress signaling pathway JNK/Nrf2/HO-1. Eur J Pharmacol 2024; 974:176620. [PMID: 38685305 DOI: 10.1016/j.ejphar.2024.176620] [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: 11/22/2023] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
The incidence and mortality of breast cancer, the most common malignant tumor among women in the world, are increasing year by year, which greatly threatens women's health. Ferroptosis is an iron and lipid reactive oxygen species (ROS)-dependent process, a novel form of cell death that is distinct from apoptosis and is closely related to the progression of breast cancer. Inducing the occurrence of ferroptosis in tumor cells can effectively block its malignant progress in vivo. Oridonin (ORI), the primary active ingredient extracted from the Chinese herbal medicine Rabdosia rubescens, has been shown to cause glutathione depletion and directly inhibit glutathione peroxidase 4 induced cell death by ferroptosis, but its mechanism of action in breast cancer remains inadequately elucidated. Therefore, we further investigated whether ORI could promote RSL3-induced ferroptosis in breast cancer cells by regulating the oxidative stress pathway JNK/Nrf2/HO-1. In our study, we assessed cell survival of RSL3 and ORI treatment by MTT assay, and found that co-treatment with RSL3 and ORI inhibited cell proliferation, as evidenced by the cloning assay. To investigate the ability of ORI to promote RSL3-induced ferroptosis in breast cancer cells, we measured levels of ROS, malondialdehyde, glutathione, superoxide dismutase, and Fe2+ content. Lipid peroxidation, ROS, and mitochondrial membrane potential levels induced by co-treatment of ORI with RSL3 were reversed by ferrostatin-1, further confirming that the cell death induced by RSL3 and ORI was ferroptosis rather than other programmed cell death modes. Moreover, RSL3 and ORI co-treatment regulated the JNK/Nrf2/HO-1 axis, as demonstrated by western blotting and target activator validation. Our results showed that ORI could enhance the inhibitory effect of RSL3 on breast cancer cells viability via the induction of ferroptosis. Mechanistically, it potentiated RSL3-induced ferroptosis in breast cancer cells by activating the JNK/Nrf2/HO-1 axis. This study provides a theoretical basis for the application of ORI based on the mechanism of ferroptosis, and provides potential natural drug candidates for cancer prevention and treatment.
Collapse
Affiliation(s)
- Shiying Ye
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| | - Xiangyan Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Su
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiye Cai
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Jinhuan Jiang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
47
|
Emanuele S, Giuliano M. Riding the Wave of Ambivalence in Cell Biology. Int J Mol Sci 2024; 25:7348. [PMID: 39000455 PMCID: PMC11242416 DOI: 10.3390/ijms25137348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Increasing evidence clearly shows that most functional molecules in the cell exert a dual role depending on the specific interactive context, biochemical pathway, or subcellular localization [...].
Collapse
Affiliation(s)
- Sonia Emanuele
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Biochemistry Building, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| |
Collapse
|
48
|
Chen Y, Li H, Zhang XL, Wang W, Rashed MMA, Duan H, Li LL, Zhai KF. Exploring the anti-skin inflammation substances and mechanism of Paeonia lactiflora Pall. Flower via network pharmacology-HPLC integration. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155565. [PMID: 38579646 DOI: 10.1016/j.phymed.2024.155565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/04/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND Paeonia lactiflora Pall. (PL) is widely used in China as a homologous plant of medicine and food. PL flower is rich in bioactive substances with anti-inflammatory effects, while the pathogenesis of skin inflammation is complex and the specific mechanism is not clear, the current treatment of skin inflammation is mainly hormonal drugs, and hormonal drugs have obvious toxic side effects. The research on the treatment of skin inflammation by PL flowers is relatively small, so this study provides a basis for the development and utilisation of PL resources. OBJECTIVE Our study was to investigate the interventional effects of PL flower extracts on skin inflammation and thus to understand its functional role in the treatment of skin inflammation and its molecular mechanisms. METHODS The major active substances in PL flower extracts were investigated by the HPLC-DAD method, and the potential targets of action were predicted by network pharmacology, which was combined with in vitro experimental validation to explore the mechanism of PL flower extracts on the regulation of skin inflammation. The HPLC-DAD analysis identified seven major active components in PL flower extracts, and in response to the results, combined with the potential mechanism of network pharmacological prediction with skin inflammation, the PL flower extract is closely related to MAPK and NF-κB signaling pathways. In addition, we also investigated the interventional effects of PL flower extract on skin inflammation by western blot detection of MAPK signaling pathway and NF-κB signaling pathway proteins in cells. RESULT Seven active components were identified and quantified from the extract of PL flowers, including Gallic acid, 1,2,3,4,6-O-Pentagalloylglucose, Oxypaeoniflorin, Paeoniflorin, Albiflorin, Benzoyloxypeoniflorin, and Rutin. It was predicted targets for the treatment of skin inflammation, with PPI showing associations with targets such as TNF, MAPK1, and IL-2. KEGG enrichment analysis revealed that the main signaling pathways involved included MAPK and T cell receptor signaling pathways. Cell experiments showed that the peony flower extract could inhibit the release of NO and inflammatory factors, as well as reduce ROS levels and inhibit cell apoptosis. Furthermore, the extract was found to inhibit the activation of the MAPK and NF-κB signaling pathways in cells. CONCLUSIONS In this study, we found that PL flower extract can inhibit the production of cell inflammatory substances, suppress the release of inflammatory factors, and deactivate inflammatory signaling pathways, further inhibiting the production of cell inflammation. This indicates that PL flower extract has a therapeutic effect on skin inflammation.
Collapse
Affiliation(s)
- Yuan Chen
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
| | - Han Li
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
| | - Xin-Lian Zhang
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
| | - Wei Wang
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
| | - Marwan M A Rashed
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
| | - Hong Duan
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China.
| | - Li-Li Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou 234000, China.
| | - Ke-Feng Zhai
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China; School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China.
| |
Collapse
|
49
|
Acero-Riaguas L, Griso-Acevedo AB, SanLorenzo-Vaquero A, Ibáñez-Herrera B, Fernandez-Diaz SM, Mascaraque M, Sánchez-Siles R, López-García I, Benítez-Buelga C, Bravo-Burguillos ER, Castelo B, Cebrián-Carretero JL, Perona R, Sastre L, Sastre-Perona A. DUSP1 and SOX2 expression determine squamous cell carcinoma of the salivary gland progression. Sci Rep 2024; 14:15007. [PMID: 38951654 PMCID: PMC11217270 DOI: 10.1038/s41598-024-65945-x] [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: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Salivary gland squamous cell carcinomas (SG-SCCs) constitute a rare type of head and neck cancer which is linked to poor prognosis. Due to their low frequency, the molecular mechanisms responsible for their aggressiveness are poorly understood. In this work we studied the role of the phosphatase DUSP1, a negative regulator of MAPK activity, in controlling SG-SCC progression. We generated DUSP1 KO clones in A253 human cells. These clones showed a reduced ability to grow in 2D, self-renew in ECM matrices and to form tumors in immunodeficient mice. This was caused by an overactivation of the stress and apoptosis kinase JNK1/2 in DUSP1-/+ clones. Interestingly, RNAseq analysis revealed that the expression of SOX2, a well-known self-renewal gene was decreased at the mRNA and protein levels in DUSP1-/+ cells. Unexpectedly, CRISPR-KO of SOX2 did not recapitulate DUSP1-/+ phenotype, and SOX2-null cells had an enhanced ability to self-renew and to form tumors in mice. Gene expression analysis demonstrated that SOX2-null cells have a decreased squamous differentiation profile -losing TP63 expression- and an increased migratory phenotype, with an enhanced epithelial to mesenchymal transition signature. In summary, our data indicates that DUSP1 and SOX2 have opposite functions in SG-SCC, being DUSP1 necessary for tumor growth and SOX2 dispensable showing a tumor suppressor function. Our data suggest that the combined expression of SOX2 and DUSP1 could be a useful biomarker to predict progression in patients with SG-SCCs.
Collapse
Affiliation(s)
- Lucía Acero-Riaguas
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Ana Belén Griso-Acevedo
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Alejandro SanLorenzo-Vaquero
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Blanca Ibáñez-Herrera
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Sara María Fernandez-Diaz
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Marta Mascaraque
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Rocío Sánchez-Siles
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Iván López-García
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Carlos Benítez-Buelga
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Elena Ruiz Bravo-Burguillos
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
| | - Beatriz Castelo
- Medical Oncology Department, University Hospital La Paz, 28046, Madrid, Spain
| | - José Luis Cebrián-Carretero
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain
- Oral and Maxillofacial Surgery Department, University Hospital La Paz, 28046, Madrid, Spain
| | - Rosario Perona
- Instituto de Salud Carlos III and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Leandro Sastre
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Ana Sastre-Perona
- Laboratory of Translational Research in Maxillofacial Surgery and Head and Neck Cancer, IdiPAZ, 28046, Madrid, Spain.
- Instituto de Investigaciones Biomédicas CSIC/UAM and CIBER de Enfermedades Raras (CIBERER), 28029, Madrid, Spain.
| |
Collapse
|
50
|
Zavvarian MM, Modi AD, Sadat S, Hong J, Fehlings MG. Translational Relevance of Secondary Intracellular Signaling Cascades Following Traumatic Spinal Cord Injury. Int J Mol Sci 2024; 25:5708. [PMID: 38891894 PMCID: PMC11172219 DOI: 10.3390/ijms25115708] [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: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a life-threatening and life-altering condition that results in debilitating sensorimotor and autonomic impairments. Despite significant advances in the clinical management of traumatic SCI, many patients continue to suffer due to a lack of effective therapies. The initial mechanical injury to the spinal cord results in a series of secondary molecular processes and intracellular signaling cascades in immune, vascular, glial, and neuronal cell populations, which further damage the injured spinal cord. These intracellular cascades present promising translationally relevant targets for therapeutic intervention due to their high ubiquity and conservation across eukaryotic evolution. To date, many therapeutics have shown either direct or indirect involvement of these pathways in improving recovery after SCI. However, the complex, multifaceted, and heterogeneous nature of traumatic SCI requires better elucidation of the underlying secondary intracellular signaling cascades to minimize off-target effects and maximize effectiveness. Recent advances in transcriptional and molecular neuroscience provide a closer characterization of these pathways in the injured spinal cord. This narrative review article aims to survey the MAPK, PI3K-AKT-mTOR, Rho-ROCK, NF-κB, and JAK-STAT signaling cascades, in addition to providing a comprehensive overview of the involvement and therapeutic potential of these secondary intracellular pathways following traumatic SCI.
Collapse
Affiliation(s)
- Mohammad-Masoud Zavvarian
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Akshat D. Modi
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Department of Biological Sciences, University of Toronto, Scarborough, ON M1C 1A4, Canada
- Department of Human Biology, University of Toronto, Toronto, ON M5S 3J6, Canada
| | - Sarah Sadat
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - James Hong
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
| | - Michael G. Fehlings
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|