1
|
Jin T, Li F, Wei W, Li Q, Gao Y, Yuwen C, Sun Y, Li W, Zhu L, Hao J. SDF2L1 downregulation mediates high glucose-caused Schwann cell dysfunction by inhibiting nuclear import of TFEB and CREB via KPNA3. Exp Neurol 2025; 390:115273. [PMID: 40294738 DOI: 10.1016/j.expneurol.2025.115273] [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: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
Schwann cells dysfunction is a key contributor to diabetic peripheral neuropathy (DPN), affecting both neurons and blood vessels. However, the precise mechanisms underlying high glucose-induced Schwann cells dysfunction are still not fully elucidated. In the present study, we investigated the expression, function and molecular mechanisms of SDF2L1 in Schwann cells using diabetic mice, SDF2L1 KO mice, rat Schwann cell (RSC96) and primary rat Schwann cell (PRSC). The RNA-seq of high glucose-treated RSC96 cells revealed an evident downregulation of SDF2L1 at both 48 and 72 h. The inhibition of high glucose on SDF2L1 expression was further confirmed at the levels of mRNA and protein in RSC96 and PRSC cells. Again, reduced SDF2L1 expression was also observed in the sciatic nerves of both type 1 and 2 diabetic mice. Functional exploration revealed that SDF2L1 knockdown in RSC96 cells suppressed the expression of LC3-II, P62, BDNF, NGF and IGF. In vivo SDF2L1 KO also decreased these proteins expression in the sciatic nerve of C57BL/6 J mice, along with the reduced nerve conduction velocity and action potential amplitude. Then, proteomics analyses and biological experiments demonstrated that SDF2L1 knockdown significantly decreased KPNA3 expression in RSC96 cells. Overexpression of KPNA3 ameliorated the decreases in LC3-II, P62, BDNF, NGF and IGF caused by SDF2L1 downregulation in vitro. Moreover, KPNA3 affected the nuclear import of transcription factors TFEB and CREB in RSC96 cells. Next, KPNA3 overexpression reversed SDF2L1 KO-reduced the nuclear aggregation of TFEB and CREB, and the expression of LC3, P62, BDNF and NGF in vivo. Collectively, these findings suggest that decreased SDF2L1 inhibits cell autophagy and neurotrophin expression by impeding the nuclear import of TFEB and CREB via KPNA3 downregulation in high glucose-treated Schwann cells.
Collapse
Affiliation(s)
- Tingting Jin
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fan Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wandi Wei
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiuli Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yue Gao
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chao Yuwen
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuanyuan Sun
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wenhui Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lin Zhu
- Department of Electromyogram, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, Hebei, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Forensic Medicine, Shijiazhuang, Hebei, China.
| |
Collapse
|
2
|
Liang X, Xu B, Wang Q, Gong K, Han C, Sun B, Ma K, Wang L. RGS14 promotes the progression of hepatocellular carcinoma by activating the cAMP/PKA/CREB signaling pathway. J Cancer Res Clin Oncol 2025; 151:153. [PMID: 40312502 PMCID: PMC12045833 DOI: 10.1007/s00432-025-06212-y] [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: 03/22/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND G protein-coupled receptors (GPCRs) mediate the intracellular signals that drive tumor development. Regulator of G protein signaling 14 (RGS14), a key negative regulator of GPCR signaling, influences liver injury, fat metabolism, and inflammation. However, the role of RGS14 in hepatocellular carcinoma (HCC) progression and its underlying mechanisms remain unclear. METHODS In this study, we compared three pairs of HCC tissues and matched portal vein tumor thrombus (PVTT) samples using 4D-FastDIA proteomics to identify differentially expressed proteins. The clinical significance of RGS14 expression was further evaluated in HCC patient cohorts. Stable RGS14-overexpressing/knockdown cell models were established for functional assays (CCK-8, colony formation, Transwell, and wound healing assays). Additionally, tumor proliferation was evaluated through in vivo studies using a subcutaneous xenograft mouse model. RNA sequencing and western blot analysis were subsequently applied to validate the potential downstream signaling pathways. RESULTS The results revealed that RGS14 was overexpressed in HCC tissues, which was correlated with adverse clinical outcomes. We also confirmed that RGS14 increased the proliferation, colony formation, migration, and invasion and promoted the epithelial‒mesenchymal transition (EMT) of HCC cells both in vitro and in vivo. Mechanistically, RGS14 elevated intracellular cAMP levels, activating the PKA/CREB axis to drive HCC progression. CONCLUSION Our findings suggest that RGS14 plays a critical oncogenic role in HCC by regulating cAMP/PKA/CREB pathway activation, underscoring its potential as both a prognostic marker and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Xiangnan Liang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bin Xu
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiuxiang Wang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Kai Gong
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Chun Han
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, China
| | - Binwen Sun
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
- Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Kexin Ma
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, China.
| | - Liming Wang
- Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, Liaoning, 116023, China.
| |
Collapse
|
3
|
Fukuoka S, Adachi N, Ouchi E, Ikemoto H, Okumo T, Ishikawa F, Onda H, Sunagawa M. Mechanoreceptor Piezo1 channel-mediated interleukin expression in conjunctival epithelial cells: Linking mechanical stress to ocular inflammation. Ocul Surf 2025; 36:56-68. [PMID: 39778715 DOI: 10.1016/j.jtos.2025.01.001] [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: 06/26/2024] [Revised: 12/27/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
PURPOSE Mechanical stress on the ocular surface, such as from eye-rubbing, has been reported to lead to inflammation and various ocular conditions. We hypothesized that the mechanosensitive Piezo1 channel in the conjunctival epithelium contributes to the inflammatory response at the ocular surface after receiving mechanical stimuli. METHODS Human conjunctival epithelial cells (HConjECs) were treated with Yoda1, a Piezo1-specific agonist, and various allergens to measure cytokine expression levels using qRT-PCR. Piezo1 activation-induced intracellular signaling pathways were also investigated by Western blot. Mechanical stretching experiments were conducted to simulate Piezo1 activation in HConjECs. Specificity of Piezo1 was confirmed by PIEZO1 knockdown and GsMTx4. In in vivo studies, using immunohistochemistry, rats were administered Yoda1 eye drops to examine the inflammatory response in the conjunctiva and Piezo1-induced signaling activation. RESULTS HConjECs expressed functional Piezo1 channel which was the dominant mechanoreceptor among putative channels and whose activation significantly increased IL-6 and IL-8 expression through the p38 MAPK-CREB pathway. Piezo1-induced [Ca2+]i elevation was crucial for the production of IL-6. The Yoda1-induced inflammatory responses were blocked by PIEZO1 knockdown. Mechanical stretching mimicked these effects, which were suppressed by GsMTx4. In vivo, Yoda1 administration led to increased phospho-p38 MAPK, phospho-CREB, and IL-6 in the rat conjunctival epithelium, with significant neutrophil infiltration. CONCLUSION Mechanical stress-induced Piezo1 channel activation in conjunctival epithelial cells can cause ocular inflammation by upregulating pro-inflammatory cytokines via the p38 MAPK-CREB pathway and promoting neutrophil infiltration. These findings suggest that mechanical stimuli on ocular surface tissues are significant risk factors for ocular inflammation.
Collapse
Affiliation(s)
- Seiya Fukuoka
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Department of Ophthalmology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Naoki Adachi
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan.
| | - Erika Ouchi
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Hideshi Ikemoto
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Takayuki Okumo
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Fumihiro Ishikawa
- Center for Biotechnology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Hidetoshi Onda
- Department of Ophthalmology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Masataka Sunagawa
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| |
Collapse
|
4
|
Hong J, Wu Y, Li M, Man KF, Song D, Koh SB. cAMP response element-binding protein: A credible cancer drug target. J Pharmacol Exp Ther 2025; 392:103529. [PMID: 40157009 PMCID: PMC12060161 DOI: 10.1016/j.jpet.2025.103529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/22/2025] [Indexed: 04/01/2025] Open
Abstract
Despite advancements in radiotherapy, chemotherapy, endocrine therapy, targeted therapy, and immunotherapy, resistance to therapy remains a pervasive challenge in oncology, in part owing to tumor heterogeneity. Identifying new therapeutic targets is key to addressing this challenge because it can both diversify and enhance existing treatment options, particularly through combination regimens. The cAMP response element-binding protein (CREB) is a transcription factor involved in various biological processes. It is aberrantly activated in several aggressive cancer types, including breast cancer. Clinically, high CREB expression is associated with increased breast tumor aggressiveness and poor prognosis. Functionally, CREB promotes breast cancer cell proliferation, survival, invasion, metastasis, as well as therapy resistance by deregulating genes related to apoptosis, cell cycle, and metabolism. Targeting CREB with small molecule inhibitors has demonstrated promise in preclinical studies. This review summarizes the current understanding of CREB mechanisms and their potential as a therapeutic target. SIGNIFICANCE STATEMENT: cAMP response element-binding protein (CREB) is a master regulator of multiple biological processes, including neurodevelopment, metabolic regulation, and immune response. CREB is a putative proto-oncogene in breast cancer that regulates the cell cycle, apoptosis, and cellular migration. Preclinical development of CREB-targeting small molecules is underway.
Collapse
Affiliation(s)
- Jinghui Hong
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China; Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Yuheng Wu
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mengxin Li
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ki-Fong Man
- Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Dong Song
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Siang-Boon Koh
- Faculty of Health and Life Sciences, University of Bristol, Bristol, United Kingdom; University Hospitals Bristol and Weston, National Health Service (NHS) Foundation Trust, Bristol, United Kingdom.
| |
Collapse
|
5
|
Xu J, Wang B, Ao H. Corticosterone effects induced by stress and immunity and inflammation: mechanisms of communication. Front Endocrinol (Lausanne) 2025; 16:1448750. [PMID: 40182637 PMCID: PMC11965140 DOI: 10.3389/fendo.2025.1448750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The body instinctively responds to external stimuli by increasing energy metabolism and initiating immune responses upon receiving stress signals. Corticosterone (CORT), a glucocorticoid (GC) that regulates secretion along the hypothalamic-pituitary-adrenal (HPA) axis, mediates neurotransmission and humoral regulation. Due to the widespread expression of glucocorticoid receptors (GR), the effects of CORT are almost ubiquitous in various tissue cells. Therefore, on the one hand, CORT is a molecular signal that activates the body's immune system during stress and on the other hand, due to the chemical properties of GCs, the anti-inflammatory properties of CORT act as stabilizers to control the body's response to stress. Inflammation is a manifestation of immune activation. CORT plays dual roles in this process by both promoting inflammation and exerting anti-inflammatory effects in immune regulation. As a stress hormone, CORT levels fluctuate with the degree and duration of stress, determining its effects and the immune changes it induces. The immune system is essential for the body to resist diseases and maintain homeostasis, with immune imbalance being a key factor in the development of various diseases. Therefore, understanding the role of CORT and its mechanisms of action on immunity is crucial. This review addresses this important issue and summarizes the interactions between CORT and the immune system.
Collapse
Affiliation(s)
- Jingyu Xu
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baojuan Wang
- Department of Reproductive Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Haiqing Ao
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Bao Y, Sheng B, Lv P. Oroxylin A Attenuates Homocysteine-Induced Blood-Brain Barrier (BBB) Dysfunction by Reducing Endothelial Permeability and Activating the CREB/Claudin-5 Signaling Pathway. ACS Chem Neurosci 2025; 16:1079-1085. [PMID: 39998410 DOI: 10.1021/acschemneuro.4c00749] [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: 02/26/2025] Open
Abstract
Recent reports have indicated that elevated levels of homocysteine (Hcy) are closely linked to blood-brain barrier (BBB) dysfunction in neurological disorders. Oroxylin A (OA) is a key bioactive flavonoid that has been reported to regulate brain functions. However, the role of OA in Hcy-related BBB dysfunction is less reported. In this study, we aimed to elucidate the role and molecular mechanism of OA in Hcy-mediated BBB dysfunction using both in vivo and in vitro investigations. Our findings indicate that the expression of the tight junction (TJ) protein Claudin-5 declined, and the diffusion of sodium fluorescein elevated in brains of Hcy-challenged mice. These effects were notably rescued by administration of OA. In Hcy-challenged bEnd.3 brain microvascular endothelial cells, increased endothelial permeability, reduced trans-endothelial electrical resistance (TEER), and downregulated Claudin-5 were observed. These effects were significantly reversed by 25 and 50 μM OA. Interestingly, OA treatment restored the dephosphorylation of CREB at Ser133 induced by Hcy. However, the addition of the protein kinase A/cAMP-response element binding protein (PKA/CREB) inhibitor H89 counteracted the protective effects of OA on inhibiting endothelial permeability and promoting Claudin-5 expression. Together, we demonstrate that OA protects against Hcy-induced BBB dysfunction by maintaining the integrity of endothelial barriers. This protective effect is achieved through the activation of the CREB/Claudin-5 signaling pathway, highlighting the potential therapeutic value of OA in addressing BBB-related neurological disorders.
Collapse
Affiliation(s)
- Yilu Bao
- Department of Culinary, Shunde Polytechic, No.1 Desheng East Road, Daliang, Shunde District, Foshan City, Guangdong Province 528300, China
| | - Baiyang Sheng
- Calt (JiangSu) Biopharm Co., Ltd, Building A8, Huizhi Technology Park, Nanjing Economic and Technological Development Zone, Nanjing, Jiangsu Province 211899, China
| | - Ping Lv
- Calt (JiangSu) Biopharm Co., Ltd, Building A8, Huizhi Technology Park, Nanjing Economic and Technological Development Zone, Nanjing, Jiangsu Province 211899, China
| |
Collapse
|
7
|
Jafri S, Ghani M, Stickle N, Virtanen C, Hazrati LN, Visanji NP. Translational profiling reveals novel gene expression changes in the direct and indirect pathways in a mouse model of levodopa induced dyskinesia. Front Cell Neurosci 2025; 18:1477511. [PMID: 40144773 PMCID: PMC11936753 DOI: 10.3389/fncel.2024.1477511] [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: 08/07/2024] [Accepted: 11/18/2024] [Indexed: 03/28/2025] Open
Abstract
Introduction The molecular mechanisms underlying L-dihydroxyphenylalanine (LDOPA) induced dyskinesia in Parkinson's disease are poorly understood. Here we employ two transgenic mouse lines, combining translating ribosomal affinity purification (TRAP) with bacterial artificial chromosome expression (Bac), to selectively isolate RNA from either DRD1A expressing striatonigral, or DRD2 expressing striatopallidal medium spiny neurons (MSNs) of the direct and indirect pathways respectively, to study changes in translational gene expression following repeated LDOPA treatment. Methods 6-OHDA lesioned DRD1A and DRD2 BacTRAP mice were treated with either saline or LDOPA bi-daily for 21 days over which time they developed abnormal involuntary movements reminiscent of dyskinesia. On day 22, all animals received LDOPA 40min prior to sacrifice. The striatum of the lesioned hemisphere was dissected and subject to TRAP. Extracted ribosomal RNA was amplified, purified, and gene expression was quantified using microarray. Results One hundred ninety-five significantly varying transcripts were identified among the four treatment groups. Pathway analysis revealed an overrepresentation of calcium signaling and long-term potentiation in the DRD1A expressing MSNs of the direct pathway, with significant involvement of long-term depression in the DRD2 expressing MSNs of the indirect pathway following chronic treatment with LDOPA. Several MAPK associated genes (NR4A1, GADD45G, STMN1, FOS, and DUSP1) differentiated the direct and indirect pathways following both acute and chronic LDOPA treatment. However, the MAPK pathway activator PAK1 was downregulated in the indirect pathway and upregulated in the direct pathway, strongly suggesting a role for PAK1 in regulating the opposing effects of LDOPA on these two pathways in dyskinesia. Discussion Future studies will assess the potential of targeting these genes and pathways to prevent the development of LDOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Sabika Jafri
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Krembil Discovery Tower, Toronto, ON, Canada
| | - Natalie Stickle
- University Health Network Microarray Centre, Toronto Medical Discovery Tower, Toronto, ON, Canada
| | - Carl Virtanen
- University Health Network Microarray Centre, Toronto Medical Discovery Tower, Toronto, ON, Canada
| | - Lili-Naz Hazrati
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Naomi P. Visanji
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Krembil Discovery Tower, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Estevez I, Buckley BD, Lindman M, Panzera N, Chou TW, McCourt M, Vaglio BJ, Atkins C, Firestein BL, Daniels BP. The kinase RIPK3 promotes neuronal survival by suppressing excitatory neurotransmission during central nervous system viral infection. Immunity 2025; 58:666-682.e6. [PMID: 39999836 PMCID: PMC11903149 DOI: 10.1016/j.immuni.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/17/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
While recent work has identified roles for immune mediators in regulating neural activity, how innate immune signaling within neurons influences neurotransmission remains poorly understood. Emerging evidence suggests that the modulation of neurotransmission may serve important roles in host protection during infection of the central nervous system. Here, we showed that receptor-interacting protein kinase-3 (RIPK3) preserved neuronal survival during flavivirus infection through the suppression of excitatory neurotransmission. These effects occurred independently of the traditional functions of RIPK3 in promoting necroptosis and inflammatory transcription. Instead, RIPK3 promoted phosphorylation of the neuronal regulatory kinase calcium/calmodulin-dependent protein kinase II (CaMKII), which in turn activated the transcription factor cyclic AMP response element-binding protein (CREB) to drive a neuroprotective transcriptional program and suppress deleterious glutamatergic signaling. These findings identify an unexpected function for a canonical cell death protein in promoting neuronal survival during viral infection through the modulation of neuronal activity, highlighting mechanisms of neuroimmune crosstalk.
Collapse
Affiliation(s)
- Irving Estevez
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Benjamin D Buckley
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Marissa Lindman
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Nicholas Panzera
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Tsui-Wen Chou
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Micheal McCourt
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Brandon J Vaglio
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Colm Atkins
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Brian P Daniels
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
9
|
Chen D, Tang H, Liu J, Zhang H, Rao K, Teng X, Yang F, Liu H. Luteolin-mediated phosphoproteomic changes in chicken splenic lymphocytes: Unraveling the detoxification mechanisms against ammonia-induced stress. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136931. [PMID: 39709809 DOI: 10.1016/j.jhazmat.2024.136931] [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: 06/19/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Long-term exposure to high ammonia concentrations could severely impact chicken health. On the other hand, luteolin has been shown to protect against ammonia poisoning. Although phosphorylation is critically involved in toxicity induction, the specific role of phosphorylated proteins in ammonia poisoning remains unclear. Herein, we constructed an in vitro model to study chicken ammonia poisoning and also analyzed the protective effects of luteolin. Specifically, a combined series of organic techniques such as protein extraction, enzyme digestion, modified peptide enrichment, Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) analysis, and bioinformatics analysis were employed for a quantitative omics study of phosphorylation modification in three groups of samples. Our findings revealed thousands of Differentially Expressed Proteins (DEPs). The differentially expressed modified proteins were subjected to GO classification, KEGG pathway analysis, cluster analysis, and protein interaction analysis, revealing the detoxification mechanism encompassed mitochondrial maintenance, signal transduction, transcriptional regulation, and cytoskeleton regulation. In the process, mitochondria and Golgi apparatus were the key organelles. Furthermore, the AKT1/FOXO signaling pathway and Heat Shock Proteins (HSPs) were the key core modifiers of the proteins. We hope that our findings will provide a theoretical basis and experimental support for future research on luteolin's detoxification mechanism against ammonia poisoning.
Collapse
Affiliation(s)
- Dechun Chen
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Haojinming Tang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Jiahao Liu
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Huanrong Zhang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Kaijing Rao
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Falong Yang
- Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China.
| | - Haifeng Liu
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
10
|
Parrotta ME, Colangeli L, Scipione V, Vitale C, Sbraccia P, Guglielmi V. Time Restricted Eating: A Valuable Alternative to Calorie Restriction for Addressing Obesity? Curr Obes Rep 2025; 14:17. [PMID: 39899119 PMCID: PMC11790783 DOI: 10.1007/s13679-025-00609-z] [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] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE OF REVIEW In this review, we summarize the molecular effects of time-restricted eating (TRE) and its possible role in appetite regulation. We also discuss the potential clinical benefits of TRE in obesity. RECENT FINDINGS TRE is an emerging dietary approach consisting in limiting food intake to a specific window of time each day. The rationale behind this strategy is to restore the circadian misalignment, commonly seen in obesity. Preclinical studies have shown that restricting food intake only during the active phase of the day can positively influence several cellular functions including senescence, mitochondrial activity, inflammation, autophagy and nutrients' sensing pathways. Furthermore, TRE may play a role by modulating appetite and satiety hormones, though further research is needed to clarify its exact mechanisms. Clinical trials involving patients with obesity or type 2 diabetes suggest that TRE can be effective for weight loss, but its broader effects on improving other clinical outcomes, such as cardiovascular risk factors, remain less certain. The epidemic proportions of obesity cause urgency to find dietary, pharmacological and surgical interventions that can be effective in the medium and long term. According to its molecular effects, TRE can be an interesting alternative to caloric restriction in the treatment of obesity, but the considerable variability across clinical trials regarding population, intervention, and follow-up duration makes it difficult to reach definitive conclusions.
Collapse
Affiliation(s)
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Scipione
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carolina Vitale
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
11
|
Kang K, Zhou T, Gong J, Chen W, Yue X, Zhang D, Yue L. A bitter taste receptor liganded by oxalic acid inhibits brown planthopper feeding by promoting CREB phosphorylation via the PI3K-AKT signaling pathway. Int J Biol Macromol 2025; 290:138999. [PMID: 39708894 DOI: 10.1016/j.ijbiomac.2024.138999] [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: 08/19/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Insect gustatory receptors play a critical role in modulating feeding behaviors by detecting external nutritional cues through complex biochemical pathways. Bitter taste receptors are essential for insects to identify and avoid toxins. However, the detailed molecular and cellular mechanisms by which these receptors influence insect feeding behavior remain poorly understood. Our previous research identified the bitter taste receptor NlGr23a in the brown planthopper (BPH), which specifically binds to oxalic acid and elicits a significant feeding rejection response. In this study, using an Sf9 cell line stably expressing NlGr23a, we demonstrated that oxalic acid exposure significantly enhances phosphorylation of cyclic adenosine monophosphate response element-binding protein (CREB), a protein associated with BPH food consumption. Further analysis revealed the involvement of phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway in facilitating CREB phosphorylation upon activation by oxalic acid-NlGr23a binding. These in vitro findings were corroborated by in vivo experiments examining the expression profiles of relevant proteins and protein kinases in BPHs fed an oxalic acid-supplemented diet. Our results elucidate the biochemical cascades triggered by oxalic acid-NlGr23a interaction, advancing our understanding of insect gustatory receptor-mediated feeding behavior modulation and potentially informing novel strategies for integrated pest management.
Collapse
Affiliation(s)
- Kui Kang
- College of Biology and Agriculture, Zunyi Normal University, Zunyi 563006, China
| | - Ting Zhou
- College of Biology and Agriculture, Zunyi Normal University, Zunyi 563006, China
| | - Jun Gong
- College of Biology and Agriculture, Zunyi Normal University, Zunyi 563006, China
| | - Weiwen Chen
- College of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Xiangzhao Yue
- School of Life Sciences, Shangrao Normal University, Shangrao 334001, China
| | - Daowei Zhang
- College of Biology and Agriculture, Zunyi Normal University, Zunyi 563006, China.
| | - Lei Yue
- School of Life Sciences, Hebei University, Baoding 071002, China.
| |
Collapse
|
12
|
Pisani A, Rolesi R, Mohamed-Hizam V, Montuoro R, Paludetti G, Giorgio C, Cocchiaro P, Brandolini L, Detta N, Sirico A, Amendola PG, Novelli R, Aramini A, Allegretti M, Paciello F, Grassi C, Fetoni AR. Early transtympanic administration of rhBDNF exerts a multifaceted neuroprotective effect against cisplatin-induced hearing loss. Br J Pharmacol 2025; 182:546-563. [PMID: 39390645 DOI: 10.1111/bph.17359] [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: 04/30/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Cisplatin-induced sensorineural hearing loss is a significant clinical challenge. Although the potential effects of brain-derived neurotrophic factor (BDNF) have previously been investigated in some ototoxicity models, its efficacy in cisplatin-induced hearing loss remains uncertain. This study aimed to investigate the therapeutic potential of recombinant human BDNF (rhBDNF) in protecting cells against cisplatin-induced ototoxicity. EXPERIMENTAL APPROACH Using an in vivo model of cisplatin-induced hearing loss, we investigated the beneficial effects of transtympanic administration of rhBDNF in a thermogel solution on hearing function and cochlear injury, using electrophysiological, morphological, immunofluorescence and molecular analyses. KEY RESULTS Our data showed that local rhBDNF treatment counteracted hearing loss in rats receiving cisplatin by preserving synaptic connections in the cochlear epithelium and protecting hair cells (HCs) and spiral ganglion neurons (SGNs) against cisplatin-induced cell death. Specifically, rhBDNF maintains the balance of its receptor levels (pTrkB and p75), boosting TrkB-CREB pro-survival signalling and reducing caspase 3-dependent apoptosis in the cochlea. Additionally, it activates antioxidant mechanisms while inhibiting inflammation and promoting vascular repair. CONCLUSION AND IMPLICATIONS Collectively, we demonstrated that early transtympanic treatment with rhBDNF plays a multifaceted protective role against cisplatin-induced ototoxicity, thus holding promise as a novel potential approach to preserve hearing in adult and paediatric patients undergoing cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Anna Pisani
- Department of Neuroscience, Unit of Audiology, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Rolando Rolesi
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Raffaele Montuoro
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gaetano Paludetti
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cristina Giorgio
- Research & Early Development, Dompé Farmaceutici S.p.A., L'Aquila, Italy
| | - Pasquale Cocchiaro
- Research & Early Development, Dompé Farmaceutici S.p.A., L'Aquila, Italy
| | - Laura Brandolini
- Research & Early Development, Dompé Farmaceutici S.p.A., L'Aquila, Italy
| | | | - Anna Sirico
- Research & Early Development, Dompé Farmaceutici S.p.A., L'Aquila, Italy
| | | | - Rubina Novelli
- Research & Early Development, Dompé Farmaceutici S.p.A., L'Aquila, Italy
| | - Andrea Aramini
- Research & Early Development, Dompé Farmaceutici S.p.A., L'Aquila, Italy
| | | | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Rita Fetoni
- Department of Neuroscience, Unit of Audiology, Università degli Studi di Napoli Federico II, Naples, Italy
| |
Collapse
|
13
|
Spirrison AN, Lannigan DA. RSK1 and RSK2 as therapeutic targets: an up-to-date snapshot of emerging data. Expert Opin Ther Targets 2024; 28:1047-1059. [PMID: 39632509 PMCID: PMC11801519 DOI: 10.1080/14728222.2024.2433123] [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: 07/16/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION The four members of the p90 ribosomal S6 kinase (RSK) family are serine/threonine protein kinases, which are phosphorylated and activated by ERK1/2. RSK1/2/3 are further phosphorylated by PDK1. Receiving inputs from two major signaling pathways places RSK as a key signaling node in numerous pathologies. A plethora of RSK1/2 substrates have been identified, and in the majority of cases the causative roles these RSK substrates play in the pathology are unknown. AREAS COVERED The majority of studies have focused on RSK1/2 and their functions in a diverse group of cancers. However, RSK1/2 are known to have important functions in cardiovascular disease and neurobiological disorders. Based on the literature, we identified substrates that are common in these pathologies with the goal of identifying fundamental physiological responses to RSK1/2. EXPERT OPINION The core group of targets in pathologies driven by RSK1/2 are associated with the immune response. However, there is a paucity of the literature addressing RSK function in inflammation, which is critical to know as the pan RSK inhibitor, PMD-026, is entering phase II clinical trials for metastatic breast cancer. A RSK inhibitor has the potential to be used in numerous diverse diseases and disorders.
Collapse
Affiliation(s)
| | - Deborah A. Lannigan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
14
|
Liu Z, Lin Z, Chen Y, Lu M, Hong W, Yu B, Liu G. Lipoteichoic Acid Rescued Age-Related Bone Loss by Enhancing Neuroendocrine and Growth Hormone Secretion Through TLR2/COX2/PGE2 Signalling Pathway. J Cell Mol Med 2024; 28:e70247. [PMID: 39622781 PMCID: PMC11611525 DOI: 10.1111/jcmm.70247] [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: 05/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
The phenomenon of brain-bone crosstalk pertains to the intricate interaction and communication pathways between the central nervous system and the skeletal system. Disruption in brain-bone crosstalk, particularly in disorders such as osteoporosis, can result in skeletal irregularities. Consequently, investigating and comprehending this communication network holds paramount importance in the realm of bone disease prevention and management. In this study, we found that Staphylococcus aureus lipoteichoic acid promoted the conversion of arachidonic acid to PGE2 by interacting with TLR2 receptors acting on the surface of microglial cells in the pituitary gland, leading to the upregulation of COX-2 expression. Subsequently, PGE2 bound to the EP4 receptor of growth hormone-secreting cells and activated the intracellular CREB signalling pathway, promoting GH secretion and ameliorating age-related bone loss.
Collapse
Affiliation(s)
- Zixian Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- The Second Hospital and Clinical Medical SchoolLanzhou UniversityLanzhouChina
| | - Zexin Lin
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yingqi Chen
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Mincheng Lu
- Department of Orthopedic, Shenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Weisheng Hong
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Bin Yu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guanqiao Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
15
|
Rinne A, Pluteanu F. Ca 2+ Signaling in Cardiovascular Fibroblasts. Biomolecules 2024; 14:1365. [PMID: 39595542 PMCID: PMC11592142 DOI: 10.3390/biom14111365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Fibrogenesis is a physiological process required for wound healing and tissue repair. It is induced by activation of quiescent fibroblasts, which first proliferate and then change their phenotype into migratory, contractile myofibroblasts. Myofibroblasts secrete extracellular matrix proteins, such as collagen, to form a scar. Once the healing process is terminated, most myofibroblasts undergo apoptosis. However, in some tissues, such as the heart, myofibroblasts remain active and sensitive to neurohumoral factors and inflammatory mediators, which lead eventually to excessive organ fibrosis. Many cellular processes involved in fibroblast activation, including cell proliferation, protein secretion and cell contraction, are highly regulated by intracellular Ca2+ signals. This review summarizes current research on Ca2+ signaling pathways underlying fibroblast activation. We present receptor- and ion channel-mediated Ca2+ signaling pathways, discuss how localized Ca2+ signals of the cell nucleus may be involved in fibroblast activation and present Ca2+-sensitive transcription pathways relevant for fibroblast biology. When investigated, we highlight how the function of Ca2+-handling proteins changes during cardiac and pulmonary fibrosis. Many aspects of Ca2+ signaling remain unexplored in different types of cardiovascular fibroblasts in relation to pathologies, and a better understanding of Ca2+ signaling in fibroblasts will help to design targeted therapies against fibrosis.
Collapse
Affiliation(s)
- Andreas Rinne
- Department of Biophysics and Cellular Biotechnology, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania;
| | - Florentina Pluteanu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| |
Collapse
|
16
|
Makovka YV, Oshchepkov DY, Fedoseeva LA, Markel AL, Redina OE. Effect of Short-Term Restraint Stress on the Expression of Genes Associated with the Response to Oxidative Stress in the Hypothalamus of Hypertensive ISIAH and Normotensive WAG Rats. Antioxidants (Basel) 2024; 13:1302. [PMID: 39594444 PMCID: PMC11590967 DOI: 10.3390/antiox13111302] [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: 09/28/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Normotensive and hypertensive organisms respond differently to stress factors; however, the features of the central molecular genetic mechanisms underlying the reaction of the hypertensive organism to stress have not been fully established. In this study, we examined the transcriptome profiles of the hypothalamus of hypertensive ISIAH rats, modeling a stress-sensitive form of arterial hypertension, and normotensive WAG rats at rest and after exposure to a single short-term restraint stress. It was shown that oxidative phosphorylation is the most significantly enriched process among metabolic changes in the hypothalamus of rats of both strains when exposed to a single short-term restraint stress. The analysis revealed DEGs representing both a common response to oxidative stress for both rat strains and a strain-specific response to oxidative stress for hypertensive ISIAH rats. Among the genes of the common response to oxidative stress, the most significant changes in the transcription level were observed in Nos1, Ppargc1a, Abcc1, Srxn1, Cryab, Hspb1, and Fosl1, among which Abcc1 and Nos1 are associated with hypertension, and Fosl1 and Ppargc1a encode transcription factors. The response to oxidative stress specific to hypertensive rats is associated with the activation of the Fos gene. The DEG's promoter region enrichment analysis allowed us to hypothesize that the response to oxidative stress may be mediated by the participation of the transcription factor CREB1 (Cyclic AMP-responsive element-binding protein 1) and the glucocorticoid receptor (NR3C1) under restraint stress in the hypothalamus of both rat strains. The results of the study revealed common and strain-specific features in the molecular mechanisms associated with oxidative phosphorylation and oxidative stress response in the hypothalamus of hypertensive ISIAH and normotensive WAG rats following a single short-term restraint stress. The obtained results expand the understanding of the most significant molecular targets for further research aimed at developing new therapeutic strategies for the prevention of the consequences of acute emotional stress, taking into account the hypertensive state of the patient.
Collapse
Affiliation(s)
- Yulia V. Makovka
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (Y.V.M.); (D.Y.O.); (L.A.F.); (A.L.M.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Dmitry Yu. Oshchepkov
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (Y.V.M.); (D.Y.O.); (L.A.F.); (A.L.M.)
- Kurchatov Genomic Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Larisa A. Fedoseeva
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (Y.V.M.); (D.Y.O.); (L.A.F.); (A.L.M.)
| | - Arcady L. Markel
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (Y.V.M.); (D.Y.O.); (L.A.F.); (A.L.M.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Olga E. Redina
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (Y.V.M.); (D.Y.O.); (L.A.F.); (A.L.M.)
| |
Collapse
|
17
|
Li Z, Schneikert J, Tripathi SR, Jin M, Bal G, Zuberbier T, Babina M. CREB Is Critically Implicated in Skin Mast Cell Degranulation Elicited via FcεRI and MRGPRX2. Cells 2024; 13:1681. [PMID: 39451199 PMCID: PMC11506305 DOI: 10.3390/cells13201681] [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: 07/06/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Skin mast cells (MCs) mediate acute allergic reactions in the cutaneous environment and contribute to chronic dermatoses, including urticaria, and atopic or contact dermatitis. The cAMP response element binding protein (CREB), an evolutionarily well conserved transcription factor (TF) with over 4,000 binding sites in the genome, was recently found to form a feedforward loop with KIT, maintaining MC survival. The most selective MC function is degranulation with its acute release of prestored mediators. Herein, we asked whether CREB contributes to the expression and function of the degranulation-competent receptors FcεRI and MRGPRX2. Interference with CREB by pharmacological inhibition (CREBi, 666-15) or RNA interference only slightly affected the expression of these receptors, while KIT was strongly attenuated. Interestingly, MRGPRX2 surface expression moderately increased following CREB-knockdown, whereas MRGPRX2-dependent exocytosis simultaneously decreased. FcεRI expression and function were regulated consistently, although the effect was stronger at the functional level. Preformed MC mediators (tryptase, histamine, β-hexosaminidase) remained comparable following CREB attenuation, suggesting that granule synthesis did not rely on CREB function. Collectively, in contrast to KIT, FcεRI and MRGPRX2 moderately depend on unperturbed CREB function. Nevertheless, CREB is required to maintain MC releasability irrespective of stimulus, insinuating that CREB may operate by safeguarding the degranulation machinery. To our knowledge, CREB is the first factor identified to regulate MRGPRX2 expression and function in opposite direction. Overall, the ancient TF is an indispensable component of skin MCs, orchestrating not only survival and proliferation but also their secretory competence.
Collapse
Affiliation(s)
- Zhuoran Li
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (Z.L.); (J.S.); (S.R.T.); (M.J.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Jean Schneikert
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (Z.L.); (J.S.); (S.R.T.); (M.J.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Shiva Raj Tripathi
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (Z.L.); (J.S.); (S.R.T.); (M.J.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Manqiu Jin
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (Z.L.); (J.S.); (S.R.T.); (M.J.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (Z.L.); (J.S.); (S.R.T.); (M.J.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (Z.L.); (J.S.); (S.R.T.); (M.J.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (Z.L.); (J.S.); (S.R.T.); (M.J.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
18
|
Wani SN, Grewal AK, Khan H, Singh TG. Elucidating the molecular symphony: unweaving the transcriptional & epigenetic pathways underlying neuroplasticity in opioid dependence and withdrawal. Psychopharmacology (Berl) 2024; 241:1955-1981. [PMID: 39254835 DOI: 10.1007/s00213-024-06684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
The persistent use of opioids leads to profound changes in neuroplasticity of the brain, contributing to the emergence and persistence of addiction. However, chronic opioid use disrupts the delicate balance of the reward system in the brain, leading to neuroadaptations that underlie addiction. Chronic cocaine usage leads to synchronized alterations in gene expression, causing modifications in the Nucleus Accumbens (NAc), a vital part of the reward system of the brain. These modifications assist in the development of maladaptive behaviors that resemble addiction. Neuroplasticity in the context of addiction involves changes in synaptic connectivity, neuronal morphology, and molecular signaling pathways. Drug-evoked neuroplasticity in opioid addiction and withdrawal represents a complicated interaction between environmental, genetic, and epigenetic factors. Identifying specific transcriptional and epigenetic targets that can be modulated to restore normal neuroplasticity without disrupting essential physiological processes is a critical consideration. The discussion in this article focuses on the transcriptional aspects of drug-evoked neuroplasticity, emphasizing the role of key transcription factors, including cAMP response element-binding protein (CREB), ΔFosB, NF-kB, Myocyte-enhancing factor 2 (MEF2), Methyl-CpG binding protein 2 (MeCP2), E2F3a, and FOXO3a. These factors regulate gene expression and lead to the neuroadaptive changes observed in addiction and withdrawal. Epigenetic regulation, which involves modifying gene accessibility by controlling these structures, has been identified as a critical component of addiction development. By unraveling these complex molecular processes, this study provides valuable insights that may pave the way for future therapeutic interventions targeting the mechanisms underlying addiction and withdrawal.
Collapse
Affiliation(s)
- Shahid Nazir Wani
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Aman Pharmacy College, Dholakhera, Udaipurwati, Jhunjhunu, Rajasthan, 333307, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
19
|
Gil-Martí B, Isidro-Mézcua J, Poza-Rodriguez A, Asti Tello GS, Treves G, Turiégano E, Beckwith EJ, Martin FA. Socialization causes long-lasting behavioral changes. Sci Rep 2024; 14:22302. [PMID: 39333212 PMCID: PMC11436997 DOI: 10.1038/s41598-024-73218-w] [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: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
In modern human societies, social isolation acts as a negative factor for health and life quality. On the other hand, social interaction also has profound effects on animal and human, impacting aggressiveness, feeding and sleep, among many other behaviors. Here, we observe that in the fly Drosophila melanogaster these behavioral changes long-last even after social interaction has ceased, suggesting that the socialization experience triggers behavioral plasticity. These modified behaviors maintain similar levels for 24 h and persist up to 72 h, although showing a progressive decay. We also find that impairing long-term memory mechanisms either genetically or by anesthesia abolishes the expected behavioral changes in response to social interaction. Furthermore, we show that socialization increases CREB-dependent neuronal activity and synaptic plasticity in the mushroom body, the main insect memory center analogous to mammalian hippocampus. We propose that social interaction triggers socialization awareness, understood as long-lasting changes in behavior caused by experience with mechanistic similarities to long-term memory formation.
Collapse
Affiliation(s)
- Beatriz Gil-Martí
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Julia Isidro-Mézcua
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
| | - Adriana Poza-Rodriguez
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
| | - Gerson S Asti Tello
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), UBA-CONICET, Buenos Aires, Argentina
| | - Gaia Treves
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
| | - Enrique Turiégano
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Esteban J Beckwith
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), UBA-CONICET, Buenos Aires, Argentina.
| | - Francisco A Martin
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain.
| |
Collapse
|
20
|
Chowdhury MAR, Haq MM, Lee JH, Jeong S. Multi-faceted regulation of CREB family transcription factors. Front Mol Neurosci 2024; 17:1408949. [PMID: 39165717 PMCID: PMC11333461 DOI: 10.3389/fnmol.2024.1408949] [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: 04/04/2024] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
cAMP response element-binding protein (CREB) is a ubiquitously expressed nuclear transcription factor, which can be constitutively activated regardless of external stimuli or be inducibly activated by external factors such as stressors, hormones, neurotransmitters, and growth factors. However, CREB controls diverse biological processes including cell growth, differentiation, proliferation, survival, apoptosis in a cell-type-specific manner. The diverse functions of CREB appear to be due to CREB-mediated differential gene expression that depends on cAMP response elements and multi-faceted regulation of CREB activity. Indeed, the transcriptional activity of CREB is controlled at several levels including alternative splicing, post-translational modification, dimerization, specific transcriptional co-activators, non-coding small RNAs, and epigenetic regulation. In this review, we present versatile regulatory modes of CREB family transcription factors and discuss their functional consequences.
Collapse
Affiliation(s)
- Md Arifur Rahman Chowdhury
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Md Mazedul Haq
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jeong Hwan Lee
- Division of Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Sangyun Jeong
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Molecular Biology, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
21
|
Kaur G, Devi S, Sharma A, Sood P. Pharmacological insights and role of bufalin (bufadienolides) in inflammation modulation: a narrative review. Inflammopharmacology 2024:10.1007/s10787-024-01517-9. [PMID: 39012431 DOI: 10.1007/s10787-024-01517-9] [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: 04/23/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Bufadienolides, specifically bufalin, have garnered attention for their potential therapeutic application in modulating inflammatory pathways. Bufalin is derived from toad venom and exhibits promising anti-inflammatory properties. Its anti-inflammatory effects have been demonstrated by influencing crucial signaling pathways like NF-B, MAPK, and JAK-STAT, resulting in the inhibition of pro-inflammatory substances like cytokines, chemokines, and adhesion molecules. Bufalin blocks inflammasome activation and reduces oxidative stress, hence increasing its anti-inflammatory properties. Bufalin has shown effectiveness in reducing inflammation-related diseases such as cancer, cardiovascular problems, and autoimmune ailments in preclinical investigations. Furthermore, producing new approaches of medication delivery and combining therapies with bufalin shows potential for improving its effectiveness and reducing adverse effects. This review explores the pharmacological effects and mechanistic approaches of bufalin as an anti-inflammatory agent, which further highlights its potential for therapy and offers the basis for further study on its therapeutic application in inflammation-related disorders.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Chitkara University School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| | - Sushma Devi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Akhil Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Parul Sood
- Chitkara University School of Pharmacy, Chitkara University, Baddi, Himachal Pradesh, India
| |
Collapse
|
22
|
Ho DM, Shaban M, Mahmood F, Ganguly P, Todeschini L, Van Vactor D, Artavanis-Tsakonas S. cAMP/PKA signaling regulates TDP-43 aggregation and mislocalization. Proc Natl Acad Sci U S A 2024; 121:e2400732121. [PMID: 38838021 PMCID: PMC11181030 DOI: 10.1073/pnas.2400732121] [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: 01/12/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Cytoplasmic mislocalization and aggregation of TDP-43 protein are hallmarks of amyotrophic lateral sclerosis (ALS) and are observed in the vast majority of both familial and sporadic cases. How these two interconnected processes are regulated on a molecular level, however, remains enigmatic. Genome-wide screens for modifiers of the ALS-associated genes TDP-43 and FUS have identified the phospholipase D (Pld) pathway as a key regulator of ALS-related phenotypes in the fruit fly Drosophila melanogaster [M. W. Kankel et al., Genetics 215, 747-766 (2020)]. Here, we report the results of our search for downstream targets of the enzymatic product of Pld, phosphatidic acid. We identify two conserved negative regulators of the cAMP/PKA signaling pathway, the phosphodiesterase dunce and the inhibitory subunit PKA-R2, as modifiers of pathogenic phenotypes resulting from overexpression of the Drosophila TDP-43 ortholog TBPH. We show that knockdown of either of these genes results in a mitigation of both TBPH aggregation and mislocalization in larval motor neuron cell bodies, as well as an amelioration of adult-onset motor defects and shortened lifespan induced by TBPH. We determine that PKA kinase activity is downstream of both TBPH and Pld and that overexpression of the PKA target CrebA can rescue TBPH mislocalization. These findings suggest a model whereby increasing cAMP/PKA signaling can ameliorate the molecular and functional effects of pathological TDP-43.
Collapse
Affiliation(s)
- Diana M. Ho
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
| | - Muhammad Shaban
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA02115
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA02142
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA02115
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA02142
| | - Payel Ganguly
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
| | | | - David Van Vactor
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
| | | |
Collapse
|
23
|
Hu Y, Liu J, Pan Q, Shi X, Wu X. Effects of Artificial Sugar Supplementation on the Composition and Nutritional Potency of Honey from Apis cerana. INSECTS 2024; 15:344. [PMID: 38786900 PMCID: PMC11121883 DOI: 10.3390/insects15050344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
In the global apiculture industry, reward feeding and supplementary feeding are essential for maintaining bee colonies. Beekeepers provide artificial supplements to their colonies, typically in the form of either a honey-water solution or sugar syrup. Owing to cost considerations associated with beekeeping, most beekeepers opt for sugar syrup. However, the effects of different types of artificial sugar supplements on bee colonies and their subsequent impact on honey composition remain unclear. To address this gap, this study compared the chemical composition, antioxidant capacity, and nutritional potency of three types of honey: honey derived from colonies fed sugar syrup (sugar-based product, SP) or a honey-water solution (honey-sourced honey, HH) and naturally sourced honey (flower-sourced honey, FH), which served as the control. The results revealed that FH outperformed HH and SP in terms of total acidity, sugar content, total protein content, and antioxidant capacity, and HH outperformed SP. Regarding nutritional efficacy, including the lifespan and learning and memory capabilities of worker bees, FH exhibited the best outcomes, with no significant differences observed between HH and SP. This study underscores the importance of sugar source selection in influencing honey quality and emphasizes the potential consequences of substituting honey with sugar syrup in traditional apiculture practices.
Collapse
Affiliation(s)
- Yueyang Hu
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, China; (Y.H.); (J.L.); (X.S.)
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, China
- Jiangxi Anyuan Honeybee Science and Technology Backyard, Anyuan 342100, China;
| | - Jianhui Liu
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, China; (Y.H.); (J.L.); (X.S.)
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, China
| | - Qizhong Pan
- Jiangxi Anyuan Honeybee Science and Technology Backyard, Anyuan 342100, China;
- Jiangxi Ganzhou Agricultural College, Ganzhou 341199, China
| | - Xinxin Shi
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, China; (Y.H.); (J.L.); (X.S.)
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, China
| | - Xiaobo Wu
- Honeybee Research Institute, Jiangxi Agricultural University, Nanchang 330045, China; (Y.H.); (J.L.); (X.S.)
- Jiangxi Province Key Laboratory of Honeybee Biology and Beekeeping, Nanchang 330045, China
- Jiangxi Anyuan Honeybee Science and Technology Backyard, Anyuan 342100, China;
| |
Collapse
|
24
|
Li L, Chen Z, Hao C. Neuroprotective effects of polyphyllin VI against rotenone-induced toxicity in SH-SY5Y cells. Brain Res 2024; 1830:148824. [PMID: 38417654 DOI: 10.1016/j.brainres.2024.148824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND A substantial body of evidence is drawing connections between Parkinson's disease (PD) and the phenomena of oxidative stress and mitochondrial dysfunction. Polyphyllin VI (PPVI), an active compound found in Rhizoma Paridis-commonly known as Chonglou (CL) in China, has been identified for its various pharmacological properties, including anti-tumor and anti-inflammatory effects. OBJECTIVE In the present study, an in vitro model of PD was established by treating SH-SY5Y cells with rotenone (ROT), to evaluate the potential neuroprotective effects of polyphyllin VI and its underlying mechanism. METHODS SH-SY5Y cells were treated with ROT to establish an in vitro model of PD. The effects of polyphyllin VI on cell viability were assessed using the resazurin assay. Cell morphology was examined using a microscope. The YO-PRO-1/PI was used to detect apoptosis. Mito-Tracker Red CMXRos, Mito-Tracker Green, and JC-1 were used to detect the effects of polyphyllin Ⅵ on mitochondrial viability, morphology, and function. Oxidative stress-related marker detection kits were used to identify the effects of polyphyllin VI on oxidative stress. Western blot analysis was employed to investigate the signaling pathways associated with neuroprotection. RESULTS PPVI increased ROT-induced SH-SY5Y cell viability and improved ROT-induced cellular morphological changes. PPVI ameliorated ROT-induced oxidative stress status, and attenuated mitochondrial function and morphological changes. PPVI may exert neuroprotective effects through FOXO3α/CREB1/DJ-1-related signaling pathways. CONCLUSION These preliminary findings suggested that PPVI possesses neuroprotective attributes in vitro, and it may be a potential candidate for PD treatment. However, extensive research is necessary to fully understand the mechanisms of PPVI and its effectiveness both in vitro and in vivo.
Collapse
Affiliation(s)
- Lanxin Li
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhengqian Chen
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Cui Hao
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
25
|
Estevez I, Buckley BD, Panzera N, Lindman M, Chou TW, McCourt M, Vaglio BJ, Atkins C, Firestein BL, Daniels BP. RIPK3 promotes neuronal survival by suppressing excitatory neurotransmission during CNS viral infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591333. [PMID: 38712188 PMCID: PMC11071512 DOI: 10.1101/2024.04.26.591333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
While recent work has identified roles for immune mediators in the regulation of neural activity, the capacity for cell intrinsic innate immune signaling within neurons to influence neurotransmission remains poorly understood. However, the existing evidence linking immune signaling with neuronal function suggests that modulation of neurotransmission may serve previously undefined roles in host protection during infection of the central nervous system. Here, we identify a specialized function for RIPK3, a kinase traditionally associated with necroptotic cell death, in preserving neuronal survival during neurotropic flavivirus infection through the suppression of excitatory neurotransmission. We show that RIPK3 coordinates transcriptomic changes in neurons that suppress neuronal glutamate signaling, thereby desensitizing neurons to excitotoxic cell death. These effects occur independently of the traditional functions of RIPK3 in promoting necroptosis and inflammatory transcription. Instead, RIPK3 promotes phosphorylation of the key neuronal regulatory kinase CaMKII, which in turn activates the transcription factor CREB to drive a neuroprotective transcriptional program and suppress deleterious glutamatergic signaling. These findings identify an unexpected function for a canonical cell death protein in promoting neuronal survival during viral infection through the modulation of neuronal activity, highlighting new mechanisms of neuroimmune crosstalk.
Collapse
Affiliation(s)
- Irving Estevez
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Benjamin D. Buckley
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Nicholas Panzera
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Marissa Lindman
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Tsui-Wen Chou
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Micheal McCourt
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Brandon J. Vaglio
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Colm Atkins
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Brian P. Daniels
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Lead Contact
| |
Collapse
|
26
|
Lee A, Lim J, Lim JS. Emerging roles of MITF as a crucial regulator of immunity. Exp Mol Med 2024; 56:311-318. [PMID: 38351314 PMCID: PMC10907664 DOI: 10.1038/s12276-024-01175-5] [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: 07/31/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 02/19/2024] Open
Abstract
Microphthalmia-associated transcription factor (MITF), a basic helix-loop-helix leucine zipper transcription factor (bHLH-Zip), has been identified as a melanocyte-specific transcription factor and plays a critical role in melanocyte survival, differentiation, function, proliferation and pigmentation. Although numerous studies have explained the roles of MITF in melanocytes and in melanoma development, the function of MITF in the hematopoietic or immune system-beyond its function in melanin-producing cells-is not yet fully understood. However, there is convincing and increasing evidence suggesting that MITF may play multiple important roles in immune-related cells. Therefore, this review is focused on recent advances in elucidating novel functions of MITF in cancer progression and immune responses to cancer. In particular, we highlight the role of MITF as a central modulator in the regulation of immune responses, as elucidated in recent studies.
Collapse
Affiliation(s)
- Aram Lee
- Department of Biological Science and the Cellular Heterogeneity Research Center, Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jihyun Lim
- Department of Biological Science and the Cellular Heterogeneity Research Center, Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jong-Seok Lim
- Department of Biological Science and the Cellular Heterogeneity Research Center, Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
27
|
Zhang H, Liu Y, Liu J, Chen J, Wang J, Hua H, Jiang Y. cAMP-PKA/EPAC signaling and cancer: the interplay in tumor microenvironment. J Hematol Oncol 2024; 17:5. [PMID: 38233872 PMCID: PMC10792844 DOI: 10.1186/s13045-024-01524-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Cancer is a complex disease resulting from abnormal cell growth that is induced by a number of genetic and environmental factors. The tumor microenvironment (TME), which involves extracellular matrix, cancer-associated fibroblasts (CAF), tumor-infiltrating immune cells and angiogenesis, plays a critical role in tumor progression. Cyclic adenosine monophosphate (cAMP) is a second messenger that has pleiotropic effects on the TME. The downstream effectors of cAMP include cAMP-dependent protein kinase (PKA), exchange protein activated by cAMP (EPAC) and ion channels. While cAMP can activate PKA or EPAC and promote cancer cell growth, it can also inhibit cell proliferation and survival in context- and cancer type-dependent manner. Tumor-associated stromal cells, such as CAF and immune cells, can release cytokines and growth factors that either stimulate or inhibit cAMP production within the TME. Recent studies have shown that targeting cAMP signaling in the TME has therapeutic benefits in cancer. Small-molecule agents that inhibit adenylate cyclase and PKA have been shown to inhibit tumor growth. In addition, cAMP-elevating agents, such as forskolin, can not only induce cancer cell death, but also directly inhibit cell proliferation in some cancer types. In this review, we summarize current understanding of cAMP signaling in cancer biology and immunology and discuss the basis for its context-dependent dual role in oncogenesis. Understanding the precise mechanisms by which cAMP and the TME interact in cancer will be critical for the development of effective therapies. Future studies aimed at investigating the cAMP-cancer axis and its regulation in the TME may provide new insights into the underlying mechanisms of tumorigenesis and lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Hongying Zhang
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongliang Liu
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jieya Liu
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinzhu Chen
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yangfu Jiang
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
Jeong S. Function and regulation of nitric oxide signaling in Drosophila. Mol Cells 2024; 47:100006. [PMID: 38218653 PMCID: PMC10880079 DOI: 10.1016/j.mocell.2023.12.004] [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: 11/08/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/15/2024] Open
Abstract
Nitric oxide (NO) serves as an evolutionarily conserved signaling molecule that plays an important role in a wide variety of cellular processes. Extensive studies in Drosophila melanogaster have revealed that NO signaling is required for development, physiology, and stress responses in many different types of cells. In neuronal cells, multiple NO signaling pathways appear to operate in different combinations to regulate learning and memory formation, synaptic transmission, selective synaptic connections, axon degeneration, and axon regrowth. During organ development, elevated NO signaling suppresses cell cycle progression, whereas downregulated NO leads to an increase in larval body size via modulation of hormone signaling. The most striking feature of the Drosophila NO synthase is that various stressors, such as neuropeptides, aberrant proteins, hypoxia, bacterial infection, and mechanical injury, can activate Drosophila NO synthase, initially regulating cellular physiology to enable cells to survive. However, under severe stress or pathophysiological conditions, high levels of NO promote regulated cell death and the development of neurodegenerative diseases. In this review, I highlight and discuss the current understanding of molecular mechanisms by which NO signaling regulates distinct cellular functions and behaviors.
Collapse
Affiliation(s)
- Sangyun Jeong
- Department of Molecular Biology, Department of Bioactive Material Sciences, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeollabukdo 54896, Republic of Korea.
| |
Collapse
|
29
|
Bal G, Schneikert J, Li Z, Franke K, Tripathi SR, Zuberbier T, Babina M. CREB Is Indispensable to KIT Function in Human Skin Mast Cells-A Positive Feedback Loop between CREB and KIT Orchestrates Skin Mast Cell Fate. Cells 2023; 13:42. [PMID: 38201246 PMCID: PMC10778115 DOI: 10.3390/cells13010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Skin mast cells (MCs) are critical effector cells in acute allergic reactions, and they contribute to chronic dermatoses like urticaria and atopic and contact dermatitis. KIT represents the cells' crucial receptor tyrosine kinase, which orchestrates proliferation, survival, and functional programs throughout the lifespan. cAMP response element binding protein (CREB), an evolutionarily well-conserved transcription factor (TF), regulates multiple cellular programs, but its function in MCs is poorly understood. We recently reported that CREB is an effector of the SCF (Stem Cell Factor)/KIT axis. Here, we ask whether CREB may also act upstream of KIT to orchestrate its functioning. Primary human MCs were isolated from skin and cultured in SCF+IL-4 (Interleukin-4). Pharmacological inhibition (666-15) and RNA interference served to manipulate CREB function. We studied KIT expression using flow cytometry and RT-qPCR, KIT-mediated signaling using immunoblotting, and cell survival using scatterplot and caspase-3 activity. The proliferation and cycle phases were quantified following BrdU incorporation. Transient CREB perturbation resulted in reduced KIT expression. Conversely, microphthalmia transcription factor (MITF) was unnecessary for KIT maintenance. KIT attenuation secondary to CREB was associated with heavily impaired KIT functional outputs, like anti-apoptosis and cell cycle progression. Likewise, KIT-elicited phosphorylation of ERK1/2 (Extracellular Signal-Regulated Kinase 1/2), AKT, and STAT5 (Signal Transducer and Activator of Transcription) was substantially diminished upon CREB inhibition. Surprisingly, the longer-term interference of CREB led to complete cell elimination, in a way surpassing KIT inhibition. Collectively, we reveal CREB as non-redundant in MCs, with its absence being incompatible with skin MCs' existence. Since SCF/KIT regulates CREB activity and, vice versa, CREB is required for KIT function, a positive feedforward loop between these elements dictates skin MCs' fate.
Collapse
Affiliation(s)
- Gürkan Bal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (G.B.); (J.S.); (Z.L.); (K.F.); (S.R.T.); (T.Z.)
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Jean Schneikert
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (G.B.); (J.S.); (Z.L.); (K.F.); (S.R.T.); (T.Z.)
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Zhuoran Li
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (G.B.); (J.S.); (Z.L.); (K.F.); (S.R.T.); (T.Z.)
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (G.B.); (J.S.); (Z.L.); (K.F.); (S.R.T.); (T.Z.)
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Shiva Raj Tripathi
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (G.B.); (J.S.); (Z.L.); (K.F.); (S.R.T.); (T.Z.)
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (G.B.); (J.S.); (Z.L.); (K.F.); (S.R.T.); (T.Z.)
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (G.B.); (J.S.); (Z.L.); (K.F.); (S.R.T.); (T.Z.)
- Institute of Allergology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
30
|
Chiricosta L, D’Angiolini S, Gugliandolo A, Salamone S, Pollastro F, Mazzon E. Transcriptomic Profiling after In Vitro Δ 8-THC Exposure Shows Cytoskeletal Remodeling in Trauma-Injured NSC-34 Cell Line. Pharmaceuticals (Basel) 2023; 16:1268. [PMID: 37765076 PMCID: PMC10535185 DOI: 10.3390/ph16091268] [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: 08/09/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Neuronal cell death is a physiological process that, when uncontrollable, leads to neurodegenerative disorders like spinal cord injury (SCI). SCI represents one of the major causes of trauma and disabilities worldwide for which no effective pharmacological intervention exists. Herein, we observed the beneficial effects of Δ8-Tetrahydrocannabinol (Δ8-THC) during neuronal cell death recovery. We cultured NSC-34 motoneuron cell line performing three different experiments. A traumatic scratch injury was caused in two experiments. One of the scratched was pretreated with Δ8-THC to observe the role of the cannabinoid following the trauma. An experimental control group was neither scratched nor pretreated. All the experiments underwent RNA-seq analysis. The effects of traumatic injury were observed in scratch against control comparison. Comparison of scratch models with or without pretreatment highlighted how Δ8-THC counteracts the traumatic event. Our results shown that Δ8-THC triggers the cytoskeletal remodeling probably due to the activation of the Janus Kinase Signal Transducer and Activator of Transcription (JAK/STAT) signaling pathway and the signaling cascade operated by the Mitogen-Activated Protein (MAP) Kinase signaling pathway. In light of this evidence, Δ8-THC could be a valid pharmacological approach in the treatment of abnormal neuronal cell death occurring in motoneuron cells.
Collapse
Affiliation(s)
- Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Simone D’Angiolini
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|