1
|
Woźnicki P, Bartusik-Aebisher D, Przygórzewska A, Aebisher D. Molecular mechanisms of the effects of photodynamic therapy on the brain: A review of the literature. Photodiagnosis Photodyn Ther 2025; 52:104536. [PMID: 40023269 DOI: 10.1016/j.pdpdt.2025.104536] [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: 11/01/2024] [Revised: 02/07/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Malignant gliomas are the most common primary brain tumors in adults. These tumors have a diverse molecular origin and a very poor prognosis. There is a lack of effective treatment at WHO grade IV glioma, and all glioblastomas progress or recur. Current treatments including surgical intervention, radiation therapy, and chemotherapy are insufficient and can cause damage to healthy brain tissue and neurological deficits. The preservation of healthy brain tissue during therapeutic intervention is made extremely difficult by the ability of malignant gliomas to diffusely infiltrate the surrounding brain parenchyma. Photodynamic therapy (PDT) is a treatment modality for glioma that can possibly overcome the inherent shortcommings of traditional therapies. Photodynamic therapy involves the use of a photosensitizer (PS) which, upon absorption of light by photosensitized tissue, triggers photochemical reactions generating reactive oxygen species (ROS) leading to the killing of tumor cells. Research focusing on the effective use of PDT in the treatment of glioma is already underway with promising results. Clinical studies on PDT for the treatment of gliomas have shown it to be a safe therapeutic modality with acceptable levels of side effects. However, some adverse sequelae have been observed during PDT of these tumours, such as increased photosensitivity, increased intracranial pressure or transient aphasia and worsening of pre-existing neurological deficits. Although the clinical sequelae of PDT are well described, the molecular mechanisms of PDT's effects on the healthy brain have not yet been thoroughly characterized. In our work, we attempt to summarize the molecular mechanisms of the effects of photosensitization on neural tissue, brain vasculature and the blood-brain barrier (BBB). We also point to findings presenting molecular approaches to protect the healthy brain from the adverse effects of photodynamic damage.
Collapse
Affiliation(s)
- Paweł Woźnicki
- Doctoral School, Medical College of the University of Rzeszów, Rzeszów 35-310, Poland
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of The Rzeszów University, Rzeszów 35-310, Poland
| | - Agnieszka Przygórzewska
- English Division Science Club, Medical College of The Rzeszów University, Rzeszów 35-310, Poland
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of The Rzeszów University, Rzeszów 35-310, Poland.
| |
Collapse
|
2
|
Yang X, Sha X, Wang G, Xu D, Zhang J, Tang M, Shi J. CaMKIIγ advances chronic intermittent hypoxia-induced cardiomyocyte apoptosis via HIF-1 signaling pathway. Sleep Breath 2025; 29:85. [PMID: 39836257 PMCID: PMC11750943 DOI: 10.1007/s11325-024-03225-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/28/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Our previous study have demonstrated chronic intermittent hypoxia (CIH) induced cardiomyocyte apoptosis and cardiac dysfunction. However, the molecular mechanisms are complicated and varied. In this study, we first investigated the CaMKIIγ expression and signaling pathway in the pathogenesis of cardiomyocyte apoptosis after CIH. METHODS Rats were separated into CIH and Normoxia groups, and H9c2 cells were divided into Control and CIH + 8 h groups. Rat body weight (BW) was markedly gained from two to six weeks. Furthermore, CIH decreased cardiac dysfunction, damaged cellular structure, induced myocardial fibrosis, and promoted cardiomyocyte apoptosis by HE, masson, sirius-red, and TUNEL staining. Western blot, immunohistochemical, immunofluorescence, double immunofluorescence staining were performed to investigate CaMKIIγ, Bcl-2, Bax, Caspase 3, HIF-1 protein expression. RESULTS Heart weight (HW) and HW/BW ratio in CIH group was markedly gained compared with the Normoxia group. CaMKIIγ expression was notably increased after CIH, and mainly expressed in the cytoplasm in vivo and vitro. The results of HIF-1 expression have the same trend of CaMKIIγ expression and cardiomyocyte apoptosis. In addition, the co-localizations of CaMKIIγ with Caspase 3, and CaMKIIγ with HIF-1 were observed by double immunofluorescence staining. CONCLUSIONS These results indicated increased CaMKIIγ expression advances CIH-induced cardiomyocyte apoptosis via HIF-1 signaling pathway, which afford a new insight and provide a potential therapy for OSA patients.
Collapse
Affiliation(s)
- Xuechao Yang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, 20 Xisi Rd, Nantong, Jiangsu, 226001, China
| | - Xinyu Sha
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, 20 Xisi Rd, Nantong, Jiangsu, 226001, China
| | - Gang Wang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, 20 Xisi Rd, Nantong, Jiangsu, 226001, China
| | - Duo Xu
- Department of Medical College, Nantong University, Nantong, Jiangsu, 226001, China
| | - Jingji Zhang
- Department of Medical College, Nantong University, Nantong, Jiangsu, 226001, China
| | - Ming Tang
- Department of Medical College, Nantong University, Nantong, Jiangsu, 226001, China
| | - Jiahai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, 20 Xisi Rd, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
3
|
Wang Z, Castro N, Bernstein AM, Wolosin JM. TGFβ1-driven SMAD2/3 phosphorylation and myofibroblast emergence are fully dependent on the TGFβ1 pre-activation of MAPKs and controlled by maternal leucine zipper kinase. Cell Signal 2024; 113:110963. [PMID: 37931692 PMCID: PMC10959399 DOI: 10.1016/j.cellsig.2023.110963] [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: 05/28/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Following wounding, endogenously secreted TGFβs drive resident and bone marrow-derived cells to convert into α-smooth actin (SMA)-rich, contractile myofibroblasts. The TGFβ effect is initiated by the phosphorylation of SMADs 2 and 3 (SMAD2/3). This event has been referred to as the canonical response to TGFβ. TGFβ also elicits other responses viewed as parallel events not directly connected to the SMAD activation, and thus referred to as noncanonical. A recognized response is the phosphorylation of the -activated kinase (TAK1/MAP3K), an upstream component of the mitogen-activated protein kinase (MAPK) cascade. We have now examined the relationship between these two effects of TGFβ1 at their earliest stages. The bulk of the studies were carried out with primary fibroblasts derived from the human cornea. The results' widespread relevance was confirmed in critical experiments with dermal-, and Tenon's capsule-derived fibroblasts. Cells were treated with kinase inhibitors or targeting siRNAs followed by induction by 2 ng/ml TGFβ1, and/or 10 ng/ml TNF-α. Cells were collected after 1 to 30 min for Western blot analysis and assayed for the accumulation of phosphorylated TAK1, ASK1, JNK1/2, p38, HPS27, MELK, SMAD2/3, and GAPDH. The effect of the kinase inhibitors on α-SMA expression and α-SMA stress fiber organization was also tested. For the immediate response to TGFβ1 we found that a) activation of the MAPK pathway was completed within 1 min after the addition of TGFβ1; b) phosphorylation of JNK1/2 was fully dependent on TAK1 and ASK1 activity, c) phosphorylation of MELK was fully dependent on JNK1/2 activity; d) phosphorylation of ASK1 depends on MELK activity, indicating the existence of an ASK1-MELK positive activation feedback loop; e) phosphorylation of SMAD2/3 started only after a 5 min period and reached a nadir after 10-15 min, f) the latter phosphorylation was fully blocked by inhibition of TAK1, ASK1, JNK1/2, and MELK, and siRNA-driven MELK downregulation; g) the inhibitors equally blocked the α-SMA protein expression, stress fiber development, and cell morphology changes at 72 h. These results demonstrate that the activation of the canonical pathway is fully subordinate to the activity of the MAPK pathway, challenging the concept of canonical and noncanonical TGFβ pathways and that SMAD2/3 activation is mediated by MELK, a kinase not previously associated with rapid pharmacological responses.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Ophthalmology and Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nileyma Castro
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Audrey M Bernstein
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, USA; New York VA Health Care, Medical Center, Syracuse, VA, USA
| | - J Mario Wolosin
- Department of Ophthalmology and Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Crystallographic mining of ASK1 regulators to unravel the intricate PPI interfaces for the discovery of small molecule. Comput Struct Biotechnol J 2022; 20:3734-3754. [PMID: 35891784 PMCID: PMC9294202 DOI: 10.1016/j.csbj.2022.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 11/22/2022] Open
Abstract
Protein seldom performs biological activities in isolation. Understanding the protein–protein interactions’ physical rewiring in response to pathological conditions or pathogen infection can help advance our comprehension of disease etiology, progression, and pathogenesis, which allow us to explore the alternate route to control the regulation of key target interactions, timely and effectively. Nonalcoholic steatohepatitis (NASH) is now a global public health problem exacerbated due to the lack of appropriate treatments. The most advanced anti-NASH lead compound (selonsertib) is withdrawn, though it is able to inhibit its target Apoptosis signal-regulating kinase 1 (ASK1) completely, indicating the necessity to explore alternate routes rather than complete inhibition. Understanding the interaction fingerprints of endogenous regulators at the molecular level that underpin disease formation and progression may spur the rationale of designing therapeutic strategies. Based on our analysis and thorough literature survey of the various key regulators and PTMs, the current review emphasizes PPI-based drug discovery’s relevance for NASH conditions. The lack of structural detail (interface sites) of ASK1 and its regulators makes it challenging to characterize the PPI interfaces. This review summarizes key regulators interaction fingerprinting of ASK1, which can be explored further to restore the homeostasis from its hyperactive states for therapeutics intervention against NASH.
Collapse
Key Words
- ASK1
- ASK1, Apoptosis signal-regulating kinase 1
- CFLAR, CASP8 and FADD-like apoptosis regulator
- CREG, Cellular repressor of E1A-stimulated genes
- DKK3, Dickkopf-related protein 3
- Interaction fingerprint
- NAFLD, Non-alcoholic fatty liver disease
- NASH
- NASH, Nonalcoholic steatohepatitis
- PPI, Protein-protein interaction
- PTM, Post-trancriptional modification
- PTMs
- Protein-protein interaction
- TNFAIP3, TNF Alpha Induced Protein 3
- TRAF2/6, Tumor necrosis factor receptor (TNFR)-associated factor2/6
- TRIM48, Tripartite Motif Containing 48
- TRX, Thioredoxin
- USP9X, Ubiquitin Specific Peptidase 9 X-Linked
Collapse
|
5
|
Kong LY, Liang MY, Liu JP, Lai P, Ye JS, Zhang ZX, Du ZM, Yu JJ, Gu L, Xie FC, Tang ZX, Liu ZY. Mesenchymal Stem Cell-derived Exosomes Rescue Oxygen-Glucose Deprivation-induced Injury in Endothelial Cells. Curr Neurovasc Res 2021; 17:155-163. [PMID: 32056526 DOI: 10.2174/1567202617666200214103950] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/08/2020] [Accepted: 01/16/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The effects of mesenchymal stem cell (MSC)-derived exosomes on brain microvascular endothelial cells under oxygen-glucose deprivation (OGD), which mimic cells in deep hypothermic circulatory arrest (DHCA) in vitro, are yet to be studied. METHODS MSCs were co-cultured with primary rat brain endothelial cells, which were then exposed to OGD. Cell viability, apoptosis, the inflammatory factors (IL-1β, IL-6, and TNF-α), and the activation of inflammation-associated TLR4-mediated pyroptosis and the NF-κB signaling pathway were determined. Furthermore, exosomes derived from MSCs were isolated and incubated with endothelial cells to investigate whether the effect of MSCs is associated with MSCderived exosomes. Apoptosis, cell viability, and the inflammatory response were also analyzed in OGD-induced endothelial cells incubated with MSC-derived exosomes. RESULTS OGD treatment promoted endothelial cell apoptosis, induced the release of inflammatory factors IL-1β, IL-6, and TNF-α, and inhibited cell viability. Western blot analysis showed that OGD treatment-induced TLR4, and NF-κB p65 subunit phosphorylation and caspase-1 upregulation, while co-culture with MSCs could reduce the effect of OGD treatment on endothelial cells. As expected, the effect of MSC-derived exosomes on OGD-treated endothelial cells was similar to that of MSCs. MSC-derived exosomes alleviated the OGD-induced decrease in the viability of endothelial cells, and increased levels of apoptosis, inflammatory factors, and the activation of inflammatory and inflammatory focal pathways. CONCLUSION Both MSCs and MSC-derived exosomes attenuated OGD-induced rat primary brain endothelial cell injury. These findings suggest that MSC-derived exosomes mediate at least some of the protective effects of MSCs on endothelial cells.
Collapse
Affiliation(s)
- Li-Yun Kong
- Department of Operation Room, Heart Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Meng-Ya Liang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Jian-Ping Liu
- Department of Vascular Department, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Ping Lai
- Department of Cardiology, Heart Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Jun-Song Ye
- Department of Clinical Research Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zu-Xiong Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zhi-Ming Du
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Jun-Jian Yu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Liang Gu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Fa-Chun Xie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zhi-Xian Tang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zi-You Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
6
|
Cardiac CaMKII δ and Wenxin Keli Prevents Ang II-Induced Cardiomyocyte Hypertrophy by Modulating CnA-NFATc4 and Inflammatory Signaling Pathways in H9c2 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9502651. [PMID: 33149757 PMCID: PMC7603598 DOI: 10.1155/2020/9502651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/18/2020] [Accepted: 09/20/2020] [Indexed: 01/23/2023]
Abstract
Previous studies have demonstrated that calcium-/calmodulin-dependent protein kinase II (CaMKII) and calcineurin A-nuclear factor of activated T-cell (CnA-NFAT) signaling pathways play key roles in cardiac hypertrophy (CH). However, the interaction between CaMKII and CnA-NFAT signaling remains unclear. H9c2 cells were cultured and treated with angiotensin II (Ang II) with or without silenced CaMKIIδ (siCaMKII) and cyclosporine A (CsA, a calcineurin inhibitor) and subsequently treated with Wenxin Keli (WXKL). Patch clamp recording was conducted to assess L-type Ca2+ current (ICa-L), and the expression of proteins involved in signaling pathways was measured by western blotting. Myocardial cytoskeletal protein and nuclear translocation of target proteins were assessed by immunofluorescence. The results indicated that siCaMKII suppressed Ang II-induced CH, as evidenced by reduced cell surface area and ICa-L. Notably, siCaMKII inhibited Ang II-induced activation of CnA and NFATc4 nuclear transfer. Inflammatory signaling was inhibited by siCaMKII and WXKL. Interestingly, CsA inhibited CnA-NFAT pathway expression but activated CaMKII signaling. In conclusion, siCaMKII may improve CH, possibly by blocking CnA-NFAT and MyD88 signaling, and WXKL has a similar effect. These data suggest that inhibiting CaMKII, but not CnA, may be a promising approach to attenuate CH and arrhythmia progression.
Collapse
|
7
|
Calcium-induced dissociation of CIB1 from ASK1 regulates agonist-induced activation of the p38 MAPK pathway in platelets. Biochem J 2020; 476:2835-2850. [PMID: 31530712 DOI: 10.1042/bcj20190410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 12/14/2022]
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase (MAPKKK) that regulates activation of the c-Jun N-terminal kinase (JNK)- and p38-stress response pathways leading to apoptosis in nucleated cells. We have previously shown that ASK1 is expressed in platelets and regulates agonist-induced platelet activation and thrombosis. However, the mechanism by which platelet agonists cause activation of ASK1 is unknown. Here, we show that in platelets agonist-induced activation of p38 is exclusively dependent on ASK1. Both thrombin and collagen were able to activate ASK1/p38. Activation of ASK1/p38 was strongly dependent on thromboxane A2 (TxA2) and ADP. Agonist-induced ASK1 activation is blocked by inhibition of phospholipase C (PLC) β/γ activity or by chelating intracellular Ca2+. Furthermore, treatment of platelets with thapsigargin or Ca2+ ionophore robustly induced ASK1/p38 activation. In addition, calcium and integrin-binding protein 1 (CIB1), a Ca2+-dependent negative regulator of ASK1, associates with ASK1 in resting platelets and is dissociated upon platelet activation by thrombin. Dissociation of CIB1 corresponds with ASK1 binding to tumor necrosis factor (TNF) receptor associated factor 6 (TRAF6) and the autophosphorylation of ASK1 Thr838 within the catalytic domain results in full activation of ASK1. Furthermore, genetic ablation of Cib1 in mice augments agonist-induced Ask1/p38 activation. Together our results suggest that in resting platelets ASK1 is bound to CIB1 at low Ca2+ concentrations. Agonist-induced platelet activation causes an increase in intracellular Ca2+ concentration that leads to the dissociation of CIB1 from ASK1, allowing for proper dimerization through ASK1 N-terminal coiled-coil (NCC) domains.
Collapse
|
8
|
Song YJ, Shi Y, Cui MM, Li M, Wen XR, Zhou XY, Lou HQ, Wang YL, Qi DS, Tang M, Zhang XB. H 2S attenuates injury after ischemic stroke by diminishing the assembly of CaMKII with ASK1-MKK3-p38 signaling module. Behav Brain Res 2020; 384:112520. [PMID: 32006563 DOI: 10.1016/j.bbr.2020.112520] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 01/10/2020] [Accepted: 01/29/2020] [Indexed: 10/25/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury is a leading cause of learning and memory dysfunction. Hydrogen sulfide (H2S) has been shown to confer neuroprotection in various neurodegenerative diseases, including cerebral I/R-induced hippocampal CA1 injury. However, the underlying mechanisms have not been completely understood. In the present study, rats were pretreated with SAM/NaHS (SAM, an H2S agonist, and NaHS, an H2S donor) only or SAM/NaHS combined with CaM (an activator of CaMKII) prior to cerebral ischemia. The Morris water maze test demonstrated that SAM/NaHS could alleviate learning and memory impairment induced by cerebral I/R injury. Cresyl violet staining was used to show the survival of hippocampal CA1 pyramidal neurons. SAM/NaHS significantly increased the number of surviving cells, whereas CaM weakened the protection induced by SAM/NaHS. The immunohistochemistry results indicated that the number of Iba1-positive microglia significantly increased after cerebral I/R. Compared with the I/R group, the number of Iba1-positive microglia in the SAM/NaHS groups significantly decreased. Co-Immunoprecipitation and immunoblotting were conducted to demonstrate that SAM/NaHS suppressed the assembly of CaMKII with the ASK1-MKK3-p38 signal module after cerebral I/R, which decreased the phosphorylation of p38. In contrast, CaM significantly inhibited the effects of SAM/NaHS. Taken together, the results suggested that SAM/NaHS could suppress cerebral I/R injury by downregulating p38 phosphorylation via decreasing the assembly of CaMKII with the ASK1-MKK3-p38 signal module.
Collapse
Affiliation(s)
- Yuan-Jian Song
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Yue Shi
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Miao-Miao Cui
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Man Li
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Xiang-Ru Wen
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Xiao-Yan Zhou
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - He-Qing Lou
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Yu-Lan Wang
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Da-Shi Qi
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Man Tang
- School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China.
| | - Xun-Bao Zhang
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China; School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China.
| |
Collapse
|
9
|
Kumar P, Rawat K, Sharma T, Kumari S, Saxena R, Kumar B, Baghel T, Afshan T, Siddiqi MI, Nazir A, Ghosh JK, Tripathi RK. HIV-1 Nef physically associate with CAMKIIδ - ASK-1 complex to inhibit p38MAPK signalling and apoptosis in infected cells. Life Sci 2019; 224:263-273. [PMID: 30902545 DOI: 10.1016/j.lfs.2019.03.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 11/15/2022]
Abstract
Human immunodeficiency type 1 virus accessory protein Nef is a key modulator of AIDS pathogenesis. With no enzymatic activity, Nef regulated functions in host cells largely depends on its ability to form multi-protein complex with the cellular proteins. Here, we identified Calcium (Ca2+)/Calmodulin dependent protein kinase II subunit delta (CAMKIIδ) as novel Nef interacting host protein. Further, we confirmed that Nef mediated [Ca2+]I promote formation of Nef-CAMKIIδ - apoptosis signal-regulating kinase (ASK-1) heterotrimeric complex. The assembly of Nef with CAMKIIδ - ASK-1 inhibits the downstream p38MAPK phosphorylation resulting in abrogation of apoptosis. Further, using competitive peptide inhibitors against Nef binding domains to CAMKIIδ, identified in the present study and ASK-1, individually blocked physical interaction of Nef with CAMKIIδ-ASK-1 complex and restored p38MAPK phosphorylation and apoptosis. Altogether, our study indicates that HIV-Nef modulates cytosolic [Ca2+]I and blocks CAMKIIδ - ASK-1 kinase activity to inhibit apoptosis of infected cells.
Collapse
Affiliation(s)
- Pradeep Kumar
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Kavita Rawat
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Tanuj Sharma
- Division of Molecular and Structural Biology, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Sushila Kumari
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Reshu Saxena
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Balawant Kumar
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Tanvi Baghel
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Tayyaba Afshan
- Division of Molecular and Structural Biology, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Mohammad Imran Siddiqi
- Division of Molecular and Structural Biology, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Aamir Nazir
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Jimut Kanti Ghosh
- Division of Molecular and Structural Biology, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India
| | - Raj Kamal Tripathi
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Council of Scientific & Industrial Research, BS-10/1, Sector-10 Jankipuram Extension, Uttar Pradesh, India..
| |
Collapse
|
10
|
Cao Z, Chen L, Liu Y, Peng T. Oxysophoridine rescues spinal cord injury via anti‑inflammatory, anti‑oxidative stress and anti‑apoptosis effects. Mol Med Rep 2017; 17:2523-2528. [PMID: 29207118 DOI: 10.3892/mmr.2017.8170] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/11/2017] [Indexed: 11/05/2022] Open
Abstract
Oxysophoridine (OSR) is an alkaloid extracted from Sophora alopecuroides L and has various pharmacological activities. The present study aimed to investigate the protective effects and underlying mechanisms of OSR on spinal cord injury (SCI), a clinically common serious trauma, in a rat model. The results of the present study demonstrated that the anti‑inflammatory effect of OSR improved Basso, Beatie and Bresnahan Locomotor Rating Scale scores and reduced spinal cord tissue water contents in an SCI rat model. Inflammatory activation was measured by ELISA, and Prostaglandin E2 (PGE2), intercellular adhesion molecule‑1 (ICAM‑1), cyclooxygenase‑2 (COX‑2), nuclear factor‑κB (NF‑κB) and B‑cell lymphoma 2 (Bcl‑2)/Bcl‑2‑associated X (Bax) protein expression levels using western blotting. The results revealed that treatment with OSR reduced tumor necrosis factor‑α, interleukin (IL)‑1β, IL‑6, IL‑8 and malondialdehyde, and increased superoxide dismutase and glutathione peroxidase levels in the serum of an SCI rat model. OSR significantly reduced the protein expression of inflammation‑associated proteins PGE2, ICAM‑1, COX‑2, NF‑κB and Bcl‑2/Bax ratio in the spinal cord tissue of an SCI rat model. Furthermore, the results of the current study demonstrate that OSR ameliorates SCI via anti‑inflammatory, anti‑oxidative stress and anti‑apoptosis effects.
Collapse
Affiliation(s)
- Zhong Cao
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Liang Chen
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yanbin Liu
- Department of Orthopedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Tao Peng
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
11
|
Zhong P, Quan D, Peng J, Xiong X, Liu Y, Kong B, Huang H. Role of CaMKII in free fatty acid/hyperlipidemia-induced cardiac remodeling both in vitro and in vivo. J Mol Cell Cardiol 2017; 109:1-16. [DOI: 10.1016/j.yjmcc.2017.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 01/24/2023]
|
12
|
Nishida T, Hattori K, Watanabe K. The regulatory and signaling mechanisms of the ASK family. Adv Biol Regul 2017; 66:2-22. [PMID: 28669716 DOI: 10.1016/j.jbior.2017.05.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 05/17/2017] [Accepted: 05/17/2017] [Indexed: 01/05/2023]
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) was identified as a MAP3K that activates the JNK and p38 pathways, and subsequent studies have reported ASK2 and ASK3 as members of the ASK family. The ASK family is activated by various intrinsic and extrinsic stresses, including oxidative stress, ER stress and osmotic stress. Numerous lines of evidence have revealed that members of the ASK family are critical for signal transduction systems to control a wide range of stress responses such as cell death, differentiation and cytokine induction. In this review, we focus on the precise signaling mechanisms of the ASK family in response to diverse stressors.
Collapse
Affiliation(s)
- Takuto Nishida
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Kazuki Hattori
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| |
Collapse
|
13
|
Shao S, Xu M, Zhou J, Ge X, Chen G, Guo L, Luo L, Li K, Zhu Z, Zhang F. Atorvastatin Attenuates Ischemia/Reperfusion-Induced Hippocampal Neurons Injury Via Akt-nNOS-JNK Signaling Pathway. Cell Mol Neurobiol 2017; 37:753-762. [PMID: 27488855 PMCID: PMC11482104 DOI: 10.1007/s10571-016-0412-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/29/2016] [Indexed: 12/29/2022]
Abstract
Ischemia-induced brain damage leads to apoptosis like delayed neuronal death in selectively vulnerable regions, which could further result in irreversible damages. Previous studies have demonstrated that neurons in the CA1 area of hippocampus are particularly sensitive to ischemic damage. Atorvastatin (ATV) has been reported to attenuate cognitive deficits after stroke, but precise mechanism for neuroprotection remains unknown. Therefore, the aims of this study were to investigate the neuroprotective mechanisms of ATV against ischemic brain injury induced by cerebral ischemia reperfusion. In this study, four-vessel occlusion model was established in rats with cerebral ischemia. Rats were divided into five groups: sham group, I/R group, I/R+ATV group, I/R+ATV+LY, and I/R+SP600125 group. Cresyl violet staining was carried out to examine the neuronal death of hippocampal CA1 region. Immunoblotting was used to detect the expression of the related proteins. Results showed that ATV significantly protected hippocampal CA1 pyramidal neurons against cerebral I/R. ATV could increase the phosphorylation of protein kinase B (Akt1) and nNOS, diminished the phosphorylation of JNK3 and c-Jun, and further inhibited the activation of caspase-3. Whereas, all of the aforementioned effects of ATV were reversed by LY294002 (an inhibitor of Akt1). Furthermore, pretreatment with SP600125 (an inhibitor of JNK) diminished the phosphorylation of JNK3 and c-Jun, and further inhibited the activation of caspase-3 after cerebral I/R. Taken together, our results implied that Akt-mediated phosphorylation of nNOS is involved in the neuroprotection of ATV against ischemic brain injury via suppressing JNK3 signaling pathway that provide a new experimental foundation for stroke therapy.
Collapse
Affiliation(s)
- Sen Shao
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China.
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China.
| | - Mingwei Xu
- The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, 310058, People's Republic of China
| | - Jiajun Zhou
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China
| | - Xiaoling Ge
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China
| | - Guanfeng Chen
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China
| | - Lili Guo
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China
| | - Lian Luo
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China
| | - Kun Li
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China
| | - Zhou Zhu
- The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310023, People's Republic of China
| | - Fayong Zhang
- Department of Neurosurgery, Huashan Hospital Affiliated to Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
14
|
Tetrandrine protects against oxygen-glucose-serum deprivation/reoxygenation-induced injury via PI3K/AKT/NF-κB signaling pathway in rat spinal cord astrocytes. Biomed Pharmacother 2016; 84:925-930. [DOI: 10.1016/j.biopha.2016.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/20/2016] [Accepted: 10/01/2016] [Indexed: 12/25/2022] Open
|
15
|
Gonzalez P, Rodríguez FJ. Analysis of the expression of the Wnt family of proteins and its modulatory role on cytokine expression in non activated and activated astroglial cells. Neurosci Res 2016; 114:16-29. [PMID: 27562517 DOI: 10.1016/j.neures.2016.08.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 12/23/2022]
Abstract
Despite the essential functions of astrocytes and the emerging relevance of the Wnt family of proteins in the CNS under physiological and pathological conditions, the astroglial expression of this family of proteins and its potential modulatory role on astroglial activation is almost unknown. Thus, we have evaluated the expression of all Wnt ligands, receptors and regulators, and the activation state of Wnt-related signaling pathways in non-activated and differentially activated astroglial cultures. We found that numerous Wnt ligands, receptors and regulators were expressed in non-activated astrocytes, while the Wnt-dependent pathways were constitutively active. Moreover, the expression of most detectable Wnt-related molecules and the activity of the Wnt-dependent pathways suffered post-activation variations which frequently depended on the activation system. Finally, the analysis of the effects exerted by Wnt1 and 5a on the astroglial expression of prototypical genes related to astroglial activation showed that both Wnt ligands increased the astroglial expression of interleukin 1β depending on the experimental context, while did not modulate tumor necrosis factor α, interleukin 6, transforming growth factor β1 and glial fibrillary acidic protein expression. These results strongly suggest that the Wnt family of proteins is involved in how astrocytes modulate and respond to the physiological and pathological CNS.
Collapse
Affiliation(s)
- Pau Gonzalez
- Laboratory of Molecular Neurology, National Hospital for Paraplegics, Finca la Peraleda s/n, 45071 Toledo, Spain.
| | - Francisco Javier Rodríguez
- Laboratory of Molecular Neurology, National Hospital for Paraplegics, Finca la Peraleda s/n, 45071 Toledo, Spain.
| |
Collapse
|
16
|
Impact of Heat Shock Protein A 12B Overexpression on Spinal Astrocyte Survival Against Oxygen-Glucose-Serum Deprivation/Restoration in Primary Cultured Astrocytes. J Mol Neurosci 2016; 59:511-20. [PMID: 27179807 DOI: 10.1007/s12031-016-0768-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022]
Abstract
Heat shock protein A 12B (HSPA12B) is a newly discovered member of the heat shock protein 70 family. Preclinical evidence indicates that HSPA12B helps protect the brain from ischemic injury, although its specific function remains unclear. The aim of this study is to investigate whether HSPA12B overexpression can protect astrocytes from oxygen-glucose-serum deprivation/restoration (OGD/R) injury. We analyzed the effects of HSPA12B overexpression on spinal cord ischemia-reperfusion injury and spinal astrocyte survival. After ischemia-reperfusion injury, we found that HSPA12B overexpression decreased spinal cord water content and infarct volume. MTT assay showed that HSPA12B overexpression increased astrocyte survival after OGD/R treatment. Flow cytometry results showed a marked inhibition of OGD/R-induced astrocyte apoptosis. Western blot assay showed that HSPA12B overexpression significantly increased regulatory protein B-cell lymphocyte 2 (Bcl-2) levels, whereas it decreased expression of the Bax protein, which forms a heterodimer with Bcl-2. Measurements of the level of activation of caspase-3 by Caspase-Glo®3/7 Assay kit showed that HSPA12B overexpression markedly inhibited caspase-3 activation. Notably, we demonstrated that the effects of HSPA12B on spinal astrocyte survival depended on activation of the PI3K/Akt signal pathway. These findings indicate that HSPA12B protects against spinal cord ischemia-reperfusion injury and may represent a potential treatment target.
Collapse
|
17
|
Hao H, Li S, Tang H, Liu B, Cai Y, Shi C, Xiao X. NQDI-1, an inhibitor of ASK1 attenuates acute perinatal hypoxic-ischemic cerebral injury by modulating cell death. Mol Med Rep 2016; 13:4585-92. [PMID: 27081917 PMCID: PMC4878550 DOI: 10.3892/mmr.2016.5123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 03/29/2016] [Indexed: 12/31/2022] Open
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a ubiquitously expressed protein kinase, which regulates cell fate in numerous injury conditions. Therefore, ASK1 may be a promising novel therapeutic target for injury. However, the expression and distribution of ASK1 in the perinatal brain following hypoxia-ischemia (HI) remains to be elucidated. In the present study, western blotting and immunofluorescence were used to determine the expression and distribution of ASK1 and any associated downstream targets in the perinatal rat brain following HI. NQDI‑1, a specific inhibitor of ASK1 was intracerebroventricularly injected following neonatal rats brain insult for neuroprotection. The results revealed an increased expression of ASK1 and this expression was localized to the neurons and astrocytes, compared with the sham controls. Additionally, it was demonstrated that the ASK1/ c‑Jun N‑terminal kinases (JNK) pathway was involved in the brain damage following HI in neonatal rats. Notably, NQDI‑1 significantly inhibited the in vivo expression levels of ASK1, phosphorylated (p‑)JNK, p‑c‑Jun, p53 and caspase 3. Reduced acute hypoxic‑ischemic cerebral injury and cell apoptosis was observed following the injection of NQDI‑1. Collectively, NQDI-1 attenuated acute perinatal hypoxic‑ischemic cerebral injury by inhibiting the expression of ASK1 and cell apoptosis. This may be a promising novel neuroprotective inhibitor for perinatal cerebra injury.
Collapse
Affiliation(s)
- Hu Hao
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Sitao Li
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Hui Tang
- Central Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630 P.R. China
| | - Bingqing Liu
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Yao Cai
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Congcong Shi
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Xin Xiao
- Department of Pediatrics, The Sixth Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| |
Collapse
|
18
|
Zhang K, Zhao T, Huang X, He Y, Zhou Y, Wu L, Wu K, Fan M, Zhu L. Dissolved oxygen concentration in the medium during cell culture: Defects and improvements. Cell Biol Int 2016; 40:354-60. [PMID: 26648388 DOI: 10.1002/cbin.10570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022]
Abstract
In vitro cell culture has provided a useful model to study the effects of oxygen on cellular behavior. However, it remains unknown whether the in vitro operations themselves affect the medium oxygen levels and the living states of cells. In addition, a prevailing controversy is whether reactive oxygen species (ROS) production is induced by continuous hypoxia or reoxygenation. In this study, we have measured the effects of different types of cell culture containers and the oxygen environment where medium replacement takes place on the actual oxygen tension in the medium. We found that the deviations of oxygen concentrations in the medium are much greater in 25-cm(2) flasks than in 24-well plates and 35-mm dishes. The dissolved oxygen concentrations in the medium were increased after medium replacement in normoxia, but remained unchanged in glove boxes in which the oxygen tension remained at a low level (11.4, 5.7, and 0.5% O2 ). We also found that medium replacement in normoxia increased the number of ROS-positive cells and reduced the cell viability; meanwhile, medium replacement in a glove box did not produce the above effects. Therefore, we conclude that the use of 25-cm(2) flasks should be avoided and demonstrate that continuous hypoxia does not produce ROS, whereas the reoxygenation that occurs during the harvesting of cells leads to ROS and induces cell death.
Collapse
Affiliation(s)
- Kuan Zhang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China.,Brain Research Center, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China
| | - Tong Zhao
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xin Huang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yunlin He
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yanzhao Zhou
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Liying Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Kuiwu Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ming Fan
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| | - Lingling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| |
Collapse
|
19
|
Liu J, Du L. PERK pathway is involved in oxygen-glucose-serum deprivation-induced NF-kB activation via ROS generation in spinal cord astrocytes. Biochem Biophys Res Commun 2015; 467:197-203. [DOI: 10.1016/j.bbrc.2015.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022]
|
20
|
Cell Death Inducing Microbial Protein Phosphatase Inhibitors--Mechanisms of Action. Mar Drugs 2015; 13:6505-20. [PMID: 26506362 PMCID: PMC4626703 DOI: 10.3390/md13106505] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 02/07/2023] Open
Abstract
Okadaic acid (OA) and microcystin (MC) as well as several other microbial toxins like nodularin and calyculinA are known as tumor promoters as well as inducers of apoptotic cell death. Their intracellular targets are the major serine/threonine protein phosphatases. This review summarizes mechanisms believed to be responsible for the death induction and tumor promotion with focus on the interdependent production of reactive oxygen species (ROS) and activation of Ca2+/calmodulin kinase II (CaM-KII). New data are presented using inhibitors of specific ROS producing enzymes to curb nodularin/MC-induced liver cell (hepatocyte) death. They indicate that enzymes of the arachidonic acid pathway, notably phospholipase A2, 5-lipoxygenase, and cyclooxygenases, may be required for nodularin/MC-induced (and presumably OA-induced) cell death, suggesting new ways to overcome at least some aspects of OA and MC toxicity.
Collapse
|
21
|
Huang W, Lv B, Zeng H, Shi D, Liu Y, Chen F, Li F, Liu X, Zhu R, Yu L, Jiang X. Paracrine Factors Secreted by MSCs Promote Astrocyte Survival Associated With GFAP Downregulation After Ischemic Stroke via p38 MAPK and JNK. J Cell Physiol 2015; 230:2461-75. [DOI: 10.1002/jcp.24981] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 02/23/2015] [Accepted: 03/02/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Weiyi Huang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Bingke Lv
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Huijun Zeng
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Dandan Shi
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Yi Liu
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Fanfan Chen
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Feng Li
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Xinghui Liu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Rong Zhu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Lei Yu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Xiaodan Jiang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| |
Collapse
|
22
|
Toledo FD, Pérez LM, Basiglio CL, Ochoa JE, Sanchez Pozzi EJ, Roma MG. The Ca²⁺-calmodulin-Ca²⁺/calmodulin-dependent protein kinase II signaling pathway is involved in oxidative stress-induced mitochondrial permeability transition and apoptosis in isolated rat hepatocytes. Arch Toxicol 2014; 88:1695-709. [PMID: 24614978 DOI: 10.1007/s00204-014-1219-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/19/2014] [Indexed: 11/26/2022]
Abstract
Oxidative stress (OS) is a common event in most hepatopathies, leading to mitochondrial permeability transition pore (MPTP) formation and further exacerbation of both OS from mitochondrial origin and cell death. Intracellular Ca²⁺ increase plays a permissive role in these events, but the underlying mechanisms are poorly known. We examined in primary cultured rat hepatocytes whether the Ca²⁺/calmodulin (CaM)-dependent protein kinase II (CaMKII) signaling pathway is involved in this process, by using tert-butyl hydroperoxide (tBOOH) as a pro-oxidant, model compound. tBOOH (500 μM, 15 min) induced MPTP formation, as assessed by measuring mitochondrial membrane depolarization as a surrogate marker, and increased lipid peroxidation in a cyclosporin A (CsA)-sensitive manner, revealing the involvement of MPTPs in tBOOH-induced radical oxygen species (ROS) formation. Intracellular Ca²⁺ sequestration with BAPTA/AM, CaM blockage with W7 or trifluoperazine, and CaMKII inhibition with KN-62 all fully prevented tBOOH-induced MPTP opening and reduced tBOOH-induced lipid peroxidation to a similar extent to CsA, suggesting that Ca²⁺/CaM/CaMKII signaling pathway fully mediates MPTP-mediated mitochondrial ROS generation. tBOOH-induced apoptosis, as shown by flow cytometry of annexin V/propidium iodide, mitochondrial release of cytochrome c, activation of caspase-3 and increase in the Bax-to-Bcl-xL ratio, and the Ca²⁺/CaM/CaMKII signaling antagonists fully prevented these effects. Intramitochondrial CaM and CaMKII were partially involved in tBOOH-induced MPTP formation, since W7 and KN-62 both attenuated the tBOOH-induced, MPTP-mediated swelling of isolated mitochondria. We concluded that Ca²⁺/CaM/CaMKII signaling pathway is a key mediator of OS-induced MPTP formation and the subsequent exacerbation of OS from mitochondrial origin and apoptotic cell death.
Collapse
Affiliation(s)
- Flavia D Toledo
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET-Universidad Nacional de Rosario, Suipacha 570, 2000, Rosario, Argentina
| | | | | | | | | | | |
Collapse
|
23
|
Cardioprotective 3',4'-dihydroxyflavonol attenuation of JNK and p38(MAPK) signalling involves CaMKII inhibition. Biochem J 2014; 456:149-61. [PMID: 24032640 DOI: 10.1042/bj20121538] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
DiOHF (3',4'-dihydroxyflavonol) is cardioprotective against I/R (ischaemia/reperfusion) injury. The biological activities of flavonols are associated with kinase modulation to alter cell signalling. We thus investigated the effects of DiOHF on the activation of MAPKs (mitogen-activated protein kinases) that regulate the cardiac stress response. In an ovine model of I/R, JNK (c-Jun N-terminal kinase), p38(MAPK), ERK (extracellular-signal-regulated kinase) and Akt were activated, and NP202, a pro-drug of DiOHF, reduced infarct size and inhibited JNK and p38(MAPK) activation, whereas ERK and Akt phosphorylation were unaltered. Similarly, in cultured myoblasts, DiOHF pre-treatment preserved viability and inhibited activation of JNK and p38(MAPK), but not ERK in response to acute oxidative and chemotoxic stress. Furthermore, DiOHF prevented stress-activation of the direct upstream regulators MKK4/7 (MAPK kinase 4/7) and MKK3/6 respectively. We utilized small-molecule affinity purification and identified CaMKII (Ca(2+)/calmodulin-dependent protein kinase II) as a kinase targeted by DiOHF and demonstrated potent CaMKII inhibition by DiOHF in vitro. Moreover, the specific inhibition of CaMKII with KN-93, but not KN-92, prevented oxidative stress-induced activation of JNK and p38(MAPK). The present study indicates DiOHF inhibition of CaMKII and attenuation of MKK3/6→p38(MAPK) and MKK4/7→JNK signalling as a requirement for the protective effects of DiOHF against stress stimuli and myocardial I/R injury.
Collapse
|
24
|
Dziegielewska B, Brautigan DL, Larner JM, Dziegielewski J. T-type Ca2+ channel inhibition induces p53-dependent cell growth arrest and apoptosis through activation of p38-MAPK in colon cancer cells. Mol Cancer Res 2013; 12:348-58. [PMID: 24362252 DOI: 10.1158/1541-7786.mcr-13-0485] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Epithelial tumor cells express T-type Ca(2+) channels, which are thought to promote cell proliferation. This study investigated the cellular response to T-type Ca(2+) channel inhibition either by small-molecule antagonists or by RNAi-mediated knockdown. Selective T-type Ca(2+) channel antagonists caused growth inhibition and apoptosis more effectively in HCT116 cells expressing wild-type p53 (p53wt), than in HCT116 mutant p53(-/-) cells. These antagonists increased p53-dependent gene expression and increased genomic occupancy of p53 at specific target sequences. The knockdown of a single T-type Ca(2+) channel subunit (CACNA1G) reduced cell growth and induced caspase-3/7 activation in HCT116 p53wt cells as compared with HCT116 mutant p53(-/-) cells. Moreover, CaCo2 cells that do not express functional p53 were made more sensitive to CACNA1G knockdown when p53wt was stably expressed. Upon T-type Ca(2+) channel inhibition, p38-MAPK promoted phosphorylation at Ser392 of p53wt. Cells treated with the inhibitor SB203580 or specific RNAi targeting p38-MAPKα/β (MAPK14/MAPK11) showed resistance to T-type Ca(2+) channel inhibition. Finally, the decreased sensitivity to channel inhibition was associated with decreased accumulation of p53 and decreased expression of p53 target genes, p21Cip1 (CDKN1A) and BCL2-binding component 3 (BBC3/PUMA). IMPLICATIONS A novel pathway involving p53 and p38-MAPK is revealed and provides a rationale for antitumor therapies that target T-type Ca(2+) channels.
Collapse
Affiliation(s)
- Barbara Dziegielewska
- Department of Radiation Oncology, University of Virginia School of Medicine, P.O. Box 800383, Charlottesville, VA 22908.
| | | | | | | |
Collapse
|
25
|
Berchtold MW, Villalobo A. The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:398-435. [PMID: 24188867 DOI: 10.1016/j.bbamcr.2013.10.021] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/24/2013] [Accepted: 10/26/2013] [Indexed: 12/21/2022]
Abstract
Calmodulin (CaM) is a ubiquitous Ca(2+) receptor protein mediating a large number of signaling processes in all eukaryotic cells. CaM plays a central role in regulating a myriad of cellular functions via interaction with multiple target proteins. This review focuses on the action of CaM and CaM-dependent signaling systems in the control of vertebrate cell proliferation, programmed cell death and autophagy. The significance of CaM and interconnected CaM-regulated systems for the physiology of cancer cells including tumor stem cells, and processes required for tumor progression such as growth, tumor-associated angiogenesis and metastasis are highlighted. Furthermore, the potential targeting of CaM-dependent signaling processes for therapeutic use is discussed.
Collapse
Key Words
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-ethyl]-4,5-dihydro-pyrazol-1-yl]-benzoic acid
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-vinyl]-4,5-dihydro-pyrazol-1-yl]-phenyl)-(4-methyl-piperazin-1-yl)-methanone
- (−) enantiomer of dihydropyrine 3-methyl-5-3-(4,4-diphenyl-1-piperidinyl)-propyl-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-piridine-3,5-dicarboxylate-hydrochloride (niguldipine)
- 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine
- 12-O-tetradecanoyl-phorbol-13-acetate
- 2-chloro-(ε-amino-Lys(75))-[6-(4-(N,N′-diethylaminophenyl)-1,3,5-triazin-4-yl]-CaM adduct
- 3′-(β-chloroethyl)-2′,4′-dioxo-3,5′-spiro-oxazolidino-4-deacetoxy-vinblastine
- 7,12-dimethylbenz[a]anthracene
- Apoptosis
- Autophagy
- B859-35
- CAPP(1)-CaM
- Ca(2+) binding protein
- Calmodulin
- Cancer biology
- Cell proliferation
- DMBA
- EBB
- FL-CaM
- FPCE
- HBC
- HBCP
- J-8
- KAR-2
- KN-62
- KN-93
- N-(4-aminobutyl)-2-naphthalenesulfonamide
- N-(4-aminobutyl)-5-chloro-2-naphthalenesulfonamide
- N-(6-aminohexyl)-1-naphthalenesulfonamide
- N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide
- N-8-aminooctyl-5-iodo-naphthalenesulfonamide
- N-[2-[N-(4-chlorocinnamyl)-N-methylaminomethyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulfonamide
- O-(4-ethoxyl-butyl)-berbamine
- RITC-CaM
- TA-CaM
- TFP
- TPA
- W-12
- W-13
- W-5
- W-7
- fluorescein-CaM adduct
- fluphenazine-N-2-chloroethane
- norchlorpromazine-CaM adduct
- rhodamine isothiocyanate-CaM adduct
- trifluoperazine
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, Copenhagen Biocenter 4-2-09 Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark.
| | - Antonio Villalobo
- Instituto de Investigaciones Biomédicas, Department of Cancer Biology, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
26
|
Prell T, Lautenschläger J, Grosskreutz J. Calcium-dependent protein folding in amyotrophic lateral sclerosis. Cell Calcium 2013; 54:132-43. [DOI: 10.1016/j.ceca.2013.05.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 05/16/2013] [Accepted: 05/18/2013] [Indexed: 12/25/2022]
|