1
|
Liu Y, Hong J, Wang G, Mei Z. An emerging role of SNAREs in ischemic stroke: From pre-to post-diseases. Biochem Pharmacol 2025; 236:116907. [PMID: 40158821 DOI: 10.1016/j.bcp.2025.116907] [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: 12/18/2024] [Revised: 03/04/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Ischemic stroke is a debilitating condition characterized by high morbidity, disability, recurrence, and mortality rates on a global scale, posing a significant threat to public health and economic stability. Extensive research has thoroughly explored the molecular mechanisms underlying ischemic stroke, elucidating a strong association between soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor proteins (SNAREs) and the pathogenesis of this condition. SNAREs, a class of highly conserved proteins involved in membrane fusion, play a crucial role in modulating neuronal information transmission and promoting myelin formation in the central nervous system (CNS). Preventing the SNARE complex formation, malfunctions in SNARE-dependent exocytosis, and altered regulation of SNARE-mediated vesicle fusion are linked to excitotoxicity, endoplasmic reticulum (ER) stress, and programmed cell death (PCD) in ischemic stroke. However, its underlying mechanisms remain unclear. This study conducts a comprehensive review of the existing literature on SNARE proteins, encompassing the structure, classification, and expression of the SNARE protein family, as well as the assembly - disassembly cycle of SNARE complexes and their physiological roles in the CNS. We thoroughly examine the mechanisms by which SNAREs contribute to the pathological progression and associated risk factors of ischemic stroke (hypertension, hyperglycemia, dyslipidemia, and atherosclerosis). Furthermore, our findings highlight the promise of SNAREs as a viable target for pharmacological interventions in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yaxin Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jingyan Hong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, China.
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
2
|
Hristova SH, Zhivkov AM. Intermolecular Electrostatic Interactions in Cytochrome c Protein Monolayer on Montmorillonite Alumosilicate Surface: A Positive Cooperative Effect. Int J Mol Sci 2024; 25:6834. [PMID: 38999945 PMCID: PMC11241403 DOI: 10.3390/ijms25136834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024] Open
Abstract
Montmorillonite (MM) crystal nanoplates acquire anticancer properties when coated with the mitochondrial protein cytochrome c (cytC) due to the cancer cells' capability to phagocytize cytC-MM colloid particles. The introduced exogenous cytC initiates apoptosis: an irreversible cascade of biochemical reactions leading to cell death. In the present research, we investigate the organization of the cytC layer on the MM surface by employing physicochemical and computer methods-microelectrophoresis, static, and electric light scattering-to study cytC adsorption on the MM surface, and protein electrostatics and docking to calculate the local electric potential and Gibbs free energy of interacting protein globules. The found protein concentration dependence of the adsorbed cytC quantity is nonlinear, manifesting a positive cooperative effect that emerges when the adsorbed cytC globules occupy more than one-third of the MM surface. Computer analysis reveals that the cooperative effect is caused by the formation of protein associates in which the cytC globules are oriented with oppositely charged surfaces. The formation of dimers and trimers is accompanied by a strong reduction in the electrostatic component of the Gibbs free energy of protein association, while the van der Waals component plays a secondary role.
Collapse
Affiliation(s)
- Svetlana H Hristova
- Department of Medical Physics and Biophysics, Medical Faculty, Medical University-Sofia, Zdrave Str. 2, 1431 Sofia, Bulgaria
| | - Alexandar M Zhivkov
- Scientific Research Center, "St. Kliment Ohridski" Sofia University, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria
| |
Collapse
|
3
|
Richartz N, Pietka W, Yadav A, Bostad M, Bhagwat S, Naderi S, Naderi EH, Stokke T, Ruud E, Blomhoff HK. N-acetyl cysteine turns EPAC activators into potent killers of acute lymphoblastic leukemia cells. J Biol Chem 2024; 300:105509. [PMID: 38042493 PMCID: PMC10772734 DOI: 10.1016/j.jbc.2023.105509] [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: 08/21/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023] Open
Abstract
Today, the majority of patients with pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL) survive their disease, but many of the survivors suffer from life-limiting late effects of the treatment. ALL develops in the bone marrow, where the cells are exposed to cAMP-generating prostaglandin E2. We have previously identified the cAMP signaling pathway as a putative target for improved efficacy of ALL treatment, based on the ability of cAMP signaling to reduce apoptosis induced by DNA damaging agents. In the present study, we have identified the antioxidant N-acetyl cysteine (NAC) as a powerful modifier of critical events downstream of the cell-permeable cAMP analog 8-(4-chlorophenylthio) adenosine-3', 5'- cyclic monophosphate (8-CPT). Accordingly, we found NAC to turn 8-CPT into a potent killer of ALL cells in vitro both in the presence and absence of DNA damaging treatment. Furthermore, we revealed that NAC in combination with 8-CPT is able to delay the progression of ALL in a xenograft model in NOD-scid IL2Rγnull mice. NAC was shown to rely on the ability of 8-CPT to activate the guanine-nucleotide exchange factor EPAC, and we demonstrated that the ALL cells are killed by apoptosis involving sustained elevated levels of calcium imposed by the combination of the two drugs. Taken together, we propose that 8-CPT in the presence of NAC might be utilized as a novel strategy for treating pediatric ALL patients, and that this powerful combination might be exploited to enhance the therapeutic index of current ALL targeting therapies.
Collapse
Affiliation(s)
- Nina Richartz
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Wojciech Pietka
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ajay Yadav
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Monica Bostad
- Department of Core Facilities, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sampada Bhagwat
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Soheil Naderi
- Division of Laboratory Medicine, Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | - Elin Hallan Naderi
- Section of Head and Neck Oncology, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Trond Stokke
- Department of Core Facilities, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ellen Ruud
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Heidi Kiil Blomhoff
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
4
|
Protein-Mineral Composite Particles with Logarithmic Dependence of Anticancer Cytotoxicity on Concentration of Montmorillonite Nanoplates with Adsorbed Cytochrome c. Pharmaceutics 2023; 15:pharmaceutics15020386. [PMID: 36839708 PMCID: PMC9965668 DOI: 10.3390/pharmaceutics15020386] [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: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Montmorillonite (MM) colloid nanoplates have high adsorption capacity due to their large size/thickness ratio, which allows them to be used as carriers for drug delivery. Upon adsorption of the mitochondrial protein cytochrome c (cytC) onto MM plates, the composite cytC-MM particles acquire anticancer properties because of the ability of cancer cells to phagocytize submicron particles (in contrast to the normal cells). In this way, exogenous cytC can be introduced into tumor cells, thereby triggering apoptosis-an irreversible cascade of biochemical reactions leading to cell death. In the present study, we investigated the physicochemical properties of cytC-MM particles as a function of the cytC concentration in the suspension, namely, the electrophoretic mobility, the mass increment of MM monoplates upon cytC adsorption, the ratio of the adsorbed to the free cytC in the bulk, the protein density on the MM's surface, the number of cytC globules adsorbed on an MM monoplate, the concentration of cytC-MM composite particles in the suspension, and the dependence of cytotoxicity on the cytC-MM particle concentration. For this purpose, we used microelectrophoresis, static and electric light scattering, and a colon cancer cell culture to test the cytotoxic effects of the cytC-MM suspensions. The results show that the cytotoxicity depends linearly on the logarithm of the particle concentration in the cytC-MM suspension reaching 97%.
Collapse
|
5
|
Cancer as a Channelopathy—Appreciation of Complimentary Pathways Provides a Different Perspective for Developing Treatments. Cancers (Basel) 2022; 14:cancers14194627. [PMID: 36230549 PMCID: PMC9562872 DOI: 10.3390/cancers14194627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary While improvements in technology have improved our ability to treat many forms of cancer when diagnosed at an early stage of the disease, the ability to improve survival and quality of life for patients with late stage disease has been limited, largely due to the ability of cancer cells to evade destruction when treatments block preferred paths for survival. Here, we review the role that ions and ion channels play in normal cell function, the development of disease and their role in the life and death of a cell. It is hoped that viewing cancer from the perspective of altered ion channel expression and ion balance may provide a novel approach for developing more effective treatments for this devastating disease. Abstract Life depends upon the ability of cells to evaluate and adapt to a constantly changing environment and to maintain internal stability to allow essential biochemical reactions to occur. Ions and ion channels play a crucial role in this process and are essential for survival. Alterations in the expression of the transmembrane proteins responsible for maintaining ion balance that occur as a result of mutations in the genetic code or in response to iatrogenically induced changes in the extracellular environment is a characteristic feature of oncogenesis and identifies cancer as one of a constellation of diseases known as channelopathies. The classification of cancer as a channelopathy provides a different perspective for viewing the disease. Potentially, it may expand opportunities for developing novel ways to affect or reverse the deleterious changes that underlie establishing and sustaining disease and developing tolerance to therapeutic attempts at treatment. The role of ions and ion channels and their interactions in the cell’s ability to maintain ionic balance, homeostasis, and survival are reviewed and possible approaches that mitigate gain or loss of ion channel function to contribute to new or enhance existing cancer therapies are discussed.
Collapse
|
6
|
Hassan WM, Al-Dbass A, Al-Ayadhi L, Bhat RS, El-Ansary A. Discriminant analysis and binary logistic regression enable more accurate prediction of autism spectrum disorder than principal component analysis. Sci Rep 2022; 12:3764. [PMID: 35260688 PMCID: PMC8904630 DOI: 10.1038/s41598-022-07829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/31/2022] [Indexed: 12/04/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social interaction and restricted, repetitive behavior. Multiple studies have suggested mitochondrial dysfunction, glutamate excitotoxicity, and impaired detoxification mechanism as accepted etiological mechanisms of ASD that can be targeted for therapeutic intervention. In the current study, blood samples were collected from 40 people with autism and 40 control participants after informed consent and full approval from the Institutional Review Board of King Saud University. Sodium (Na+), Potassium (K+), lactate dehydrogenase (LDH), glutathione-s-transferase (GST), and mitochondrial respiratory chain complex I (MRC1) were measured in plasma of both groups. Predictive models were established to discriminate individuals with ASD from controls. The predictive power of these five variables, individually and in combination, was compared using the area under a ROC curve (AUC). We compared the performance of principal component analysis (PCA), discriminant analysis (DA), and binary logistic regression (BLR) as ways to combine single variables and create the predictive models. K+ had the highest AUC (0.801) of any single variable, followed by GST, LDH, Na+, and MRC1, respectively. Combining the five variables resulted in higher AUCs than those obtained using single variables across all models. Both DA and BLR were superior to PCA and comparable to each other. In our study, the combination of Na+, K+, LDH, GST, and MRC1 showed the highest promise in discriminating individuals with autism from controls. These results provide a platform that can potentially be used to verify the efficacy of our models with a larger sample size or evaluate other biomarkers.
Collapse
Affiliation(s)
- Wail M Hassan
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Abeer Al-Dbass
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.,Autism Research and Treatment Center, Riyadh, Saudi Arabia
| | - Ramesa Shafi Bhat
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Autism Research and Treatment Center, Riyadh, Saudi Arabia. .,Central Research Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
7
|
Park SH, Hwang I, McNaughton DA, Kinross AJ, Howe EN, He Q, Xiong S, Kilde MD, Lynch VM, Gale PA, Sessler JL, Shin I. Synthetic Na +/K + exchangers promote apoptosis by disturbing cellular cation homeostasis. Chem 2021; 7:3325-3339. [PMID: 38239771 PMCID: PMC10795848 DOI: 10.1016/j.chempr.2021.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A number of artificial cation ionophores (or transporters) have been developed for basic research and biomedical applications. However, their mechanisms of action and the putative correlations between changes in intracellular cation concentrations and induced cell death remain poorly understood. Here, we show that three hemispherand-strapped calix[4]pyrrole-based ion-pair receptors act as efficient Na+/K+ exchangers in the presence of Cl- in liposomal models and promote Na+ influx and K+ efflux (Na+/K+ exchange) in cancer cells to induce apoptosis. Mechanistic studies reveal that these cation exchangers induce endoplasmic reticulum (ER) stress in cancer cells by perturbing intracellular cation homeostasis, promote generation of reactive oxygen species, and eventually enhance mitochondria-mediated apoptosis. However, they neither induce osmotic stress nor affect autophagy. This study provides support for the notion that synthetic receptors, which perturb cellular cation homeostasis, may provide new small molecules with potentially useful apoptotic activity.
Collapse
Affiliation(s)
- Sang-Hyun Park
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
- These authors contributed equally
| | - Inhong Hwang
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
- These authors contributed equally
| | - Daniel A. McNaughton
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
- These authors contributed equally
| | - Airlie J. Kinross
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
| | - Ethan N.W. Howe
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
- Present address: GlaxoSmithKline, GSK Jurong, 1 Pioneer Sector 1, Singapore 628413
| | - Qing He
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P.R. China
| | - Shenglun Xiong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P.R. China
| | - Martin Drøhse Kilde
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
- Present address: Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen Ø, Denmark
| | - Vincent M. Lynch
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
| | - Philip A. Gale
- School of Chemistry (F11), The University of Sydney, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute (SydneyNano), The University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathan L. Sessler
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street, Stop A5300, Austin, TX 78712, USA
- Lead contact
| | - Injae Shin
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
8
|
Hristova SH, Zhivkov AM. Montmorillonite colloid plates with adsorbed cytochrome c: in vitro cytotoxic effect on colon cancer cell culture. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00095-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The apoptosis (a cascade of biochemical reactions leading to suicide of damaged biological cells) is blocked in the cancer cells because of impossibility of cytochrome c (cytC) go out from the mitochondria. However, the apoptosis can be started by introducing of exogenous cytC into cytoplasm using colloid particles as a protein carrier due to ability of the cancer cells to phagocytize extracellular particles with submicron size.
Results
The clay mineral montmorillonite (MM) were used to prepare aqueous suspension of protein/mineral composite particles by electrostatic adsorption of the positively charged cytC globules on the negatively charged MM colloid plates, and then added to colon cancel culture. The results shows out that separately cytC and MM have no effect but the composite cytC-MM particles kill 95% of the cancer cells after 96 h treatment using equine cytC which is 97% structurally identical with the human cytC. To reach this high cytotoxicity we have formulated requirements to: (a) bare colloid particles (electric charge, form and size), (b) conditions for protein adsorption (concentrations, pH, ionic strength), and (c) suspension with the composite particles (positive total charge and optimal concentration). Due to satisfying these requirements we have reached cytotoxicity which is 1/3 higher than the reached by other authors using different artificial particles. The cytotoxicity rapidly increases with concentration of the cytC-MM particles but further it shows tendency to saturation.
Methods
The optimal pH 6.5 and the 10:3 mg/mg cytC/MM concentration ratio at adsorption were found out by employing computer (protein electrostatics) and physicochemical methods (microelectrophoresis and colloid electrooptics) to prepare cytC-MM suspension. The anticancer capability of cytC-MM nanoplates were investigated using cell culture of metastasizing colon cancer.
Conclusion
The in vitro experiments with colon cancer cell culture disclose that cytC-MM composite particles have potential for application in anticancer therapy of superficial neoplasms of the skin and the alimentary system (mouth cavity, esophagus, stomach, jejunum and colon).
Graphic abstract
Collapse
|
9
|
Li Y, Zhang Y, Lu J, Yin Y, Xie J, Xu B. Anti-inflammatory mechanisms and research progress of colchicine in atherosclerotic therapy. J Cell Mol Med 2021; 25:8087-8094. [PMID: 34312998 PMCID: PMC8419170 DOI: 10.1111/jcmm.16798] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/17/2021] [Accepted: 07/10/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory responses play a vital role in the onset and development of atherosclerosis, and throughout the entire process of the chronic disease. The inflammatory responses in atherosclerosis are mainly mediated by the NLRP3 inflammasome and its downstream inflammatory factors. As a powerful anti‐inflammatory medicine, colchicine has a history of more than 200 years in clinical application and is the first‐choice treatment for immune diseases such as gout and familial Mediterranean fever. In atherosclerosis, colchicine can inhibit the assembly and activation of NLRP3 inflammasome via various mechanisms to effectively reduce the expression of inflammatory factors, thereby reducing the inflammation. Recent clinical trials show that a low dose of colchicine (0.5 mg per day) has a certain protective effect in stable angina patients or those with acute myocardial infarction after PCI. This article summarizes and discusses the mechanisms of colchicine in the treatment of atherosclerosis and the latest research progress.
Collapse
Affiliation(s)
- Yuyu Li
- Department of Cardiology, Nanjing Drum Tower Hospital, MOE Key Laboratory of Model Animal for Disease Study, School of Medicine, Nanjing University, Nanjing, China
| | - Yuxin Zhang
- Institution of Translational Medicine, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianrong Lu
- Department of Cardiology, Nanjing Drum Tower Hospital, MOE Key Laboratory of Model Animal for Disease Study, School of Medicine, Nanjing University, Nanjing, China
| | - Yong Yin
- Department of Cardiology, Nanjing Drum Tower Hospital, MOE Key Laboratory of Model Animal for Disease Study, School of Medicine, Nanjing University, Nanjing, China
| | - Jun Xie
- Department of Cardiology, Nanjing Drum Tower Hospital, MOE Key Laboratory of Model Animal for Disease Study, School of Medicine, Nanjing University, Nanjing, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, MOE Key Laboratory of Model Animal for Disease Study, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
10
|
Contribution of Apaf-1 to the pathogenesis of cancer and neurodegenerative diseases. Biochimie 2021; 190:91-110. [PMID: 34298080 DOI: 10.1016/j.biochi.2021.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/24/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
Deregulation of apoptosis is associated with various pathologies, such as neurodegenerative disorders at one end of the spectrum and cancer at the other end. Generally speaking, differentiated cells like cardiomyocytes, skeletal myocytes and neurons exhibit low levels of Apaf-1 (Apoptotic protease activating factor 1) protein suggesting that down-regulation of Apaf-1 is an important event contributing to the resistance of these cells to apoptosis. Nonetheless, upregulation of Apaf-1 has not emerged as a common phenomenon in pathologies associated with enhanced neuronal cell death, i.e., neurodegenerative diseases. In cancer, on the other hand, Apaf-1 downregulation is a common phenomenon, which occurs through various mechanisms including mRNA hyper-methylation, gene methylation, Apaf-1 localization in lipid rafts, inhibition by microRNAs, phosphorylation, and interaction with specific inhibitors. Due to the diversity of these mechanisms and involvement of other factors, defining the exact contribution of Apaf-1 to the development of cancer in general and neurodegenerative disorders, in particular, is complicated. The current review is an attempt to provide a comprehensive image of Apaf-1's contribution to the pathologies observed in cancer and neurodegenerative diseases with the emphasis on the therapeutic aspects of Apaf-1 as an important target in these pathologies.
Collapse
|
11
|
Quo vadis Cardiac Glycoside Research? Toxins (Basel) 2021; 13:toxins13050344. [PMID: 34064873 PMCID: PMC8151307 DOI: 10.3390/toxins13050344] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
Cardiac glycosides (CGs), toxins well-known for numerous human and cattle poisoning, are natural compounds, the biosynthesis of which occurs in various plants and animals as a self-protective mechanism to prevent grazing and predation. Interestingly, some insect species can take advantage of the CG’s toxicity and by absorbing them, they are also protected from predation. The mechanism of action of CG’s toxicity is inhibition of Na+/K+-ATPase (the sodium-potassium pump, NKA), which disrupts the ionic homeostasis leading to elevated Ca2+ concentration resulting in cell death. Thus, NKA serves as a molecular target for CGs (although it is not the only one) and even though CGs are toxic for humans and some animals, they can also be used as remedies for various diseases, such as cardiovascular ones, and possibly cancer. Although the anticancer mechanism of CGs has not been fully elucidated, yet, it is thought to be connected with the second role of NKA being a receptor that can induce several cell signaling cascades and even serve as a growth factor and, thus, inhibit cancer cell proliferation at low nontoxic concentrations. These growth inhibitory effects are often observed only in cancer cells, thereby, offering a possibility for CGs to be repositioned for cancer treatment serving not only as chemotherapeutic agents but also as immunogenic cell death triggers. Therefore, here, we report on CG’s chemical structures, production optimization, and biological activity with possible use in cancer therapy, as well as, discuss their antiviral potential which was discovered quite recently. Special attention has been devoted to digitoxin, digoxin, and ouabain.
Collapse
|
12
|
Hu S, Han R, Chen L, Qin W, Xu X, Shi J, Zhu X, Zhang M, Zeng C, Tang Z, Bao H, Liu Z. Upregulated LRRC55 promotes BK channel activation and aggravates cell injury in podocytes. J Exp Med 2021; 218:e20192373. [PMID: 33346797 PMCID: PMC7756252 DOI: 10.1084/jem.20192373] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/27/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Podocyte injury is a common hallmark in various glomerular diseases. The level of LRRC55 was increased in podocytes of patients with focal segmental glomerulosclerosis (FSGS), diabetic nephropathy (DN), and membranous nephropathy (MN). Upregulated LRRC55 and increased intracellular Ca2+ led to BK channel activation and the loss of intracellular potassium, resulting in apoptosome formation and caspase-3 activation in angiotensin II (Ang II)-treated podocytes. Knockout of Lrrc55 or the BK channel prevented the BK current and ameliorated podocyte injury in Ang II-treated mice. Upstream, NFATc3 regulated the expression of LRRC55. Increased LRRC55 expression in podocytes was also evident in animal models of FSGS, DN, and MN. Treatment with losartan or LRRC55 siRNA suppressed LRRC55 expression, prevented BK channel activation, and attenuated podocyte injury in animal models of FSGS, DN, and MN. In conclusion, upregulated LRRC55 promotes BK channel activation and aggravates cell injury in podocytes in FSGS, DN, and MN. LRRC55 inhibition may represent a new therapeutic approach for podocyte injury.
Collapse
Affiliation(s)
- Shuai Hu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Runhong Han
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Long Chen
- National Standard Laboratory of Pharmacology for Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weisong Qin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jingsong Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Zhu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zheng Tang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hao Bao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
13
|
Bortner CD, Cidlowski JA. Ions, the Movement of Water and the Apoptotic Volume Decrease. Front Cell Dev Biol 2020; 8:611211. [PMID: 33324655 PMCID: PMC7723978 DOI: 10.3389/fcell.2020.611211] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022] Open
Abstract
The movement of water across the cell membrane is a natural biological process that occurs during growth, cell division, and cell death. Many cells are known to regulate changes in their cell volume through inherent compensatory regulatory mechanisms. Cells can sense an increase or decrease in their cell volume, and compensate through mechanisms known as a regulatory volume increase (RVI) or decrease (RVD) response, respectively. The transport of sodium, potassium along with other ions and osmolytes allows the movement of water in and out of the cell. These compensatory volume regulatory mechanisms maintain a cell at near constant volume. A hallmark of the physiological cell death process known as apoptosis is the loss of cell volume or cell shrinkage. This loss of cell volume is in stark contrast to what occurs during the accidental cell death process known as necrosis. During necrosis, cells swell or gain water, eventually resulting in cell lysis. Thus, whether a cell gains or loses water after injury is a defining feature of the specific mode of cell death. Cell shrinkage or the loss of cell volume during apoptosis has been termed apoptotic volume decrease or AVD. Over the years, this distinguishing feature of apoptosis has been largely ignored and thought to be a passive occurrence or simply a consequence of the cell death process. However, studies on AVD have defined an underlying movement of ions that result in not only the loss of cell volume, but also the activation and execution of the apoptotic process. This review explores the role ions play in controlling not only the movement of water, but the regulation of apoptosis. We will focus on what is known about specific ion channels and transporters identified to be involved in AVD, and how the movement of ions and water change the intracellular environment leading to stages of cell shrinkage and associated apoptotic characteristics. Finally, we will discuss these concepts as they apply to different cell types such as neurons, cardiomyocytes, and corneal epithelial cells.
Collapse
Affiliation(s)
- Carl D. Bortner
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John A. Cidlowski
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
14
|
Barbirou M, Woldu HG, Sghaier I, Bedoui SA, Mokrani A, Aami R, Mezlini A, Yacoubi-Loueslati B, Tonellato PJ, Bouhaouala-Zahar B. Western influenced lifestyle and Kv2.1 association as predicted biomarkers for Tunisian colorectal cancer. BMC Cancer 2020; 20:1086. [PMID: 33172410 PMCID: PMC7656678 DOI: 10.1186/s12885-020-07605-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most diagnosed malignancy worldwide. The global burden is expected to increase along with ongoing westernized behaviors and lifestyle. The etiology of CRC remains elusive and most likely combines environmental and genetic factors. The Kv2.1 potassium channel encoded by KCNB1 plays a collection of roles in malignancy of cancer and may be a key factor of CRC susceptibility. Our study provides baseline association between Tunisian CRC and interactions between KCNB1 variants and lifestyle factors. METHODS A case-control study involving 300 CRC patients, and 300 controls was conducted Patients were carefully phenotyped and followed till the end of study. KCNB1 genotyping was confirmed by Sanger sequencing. Bivariate and multivariable logistic regression analyses were used to assess the clinical status, lifestyle and study polymorphisms association with CRC. RESULTS We noted significant gender association with CRC occurrence. Moreover, CRC risk increases with high meat and fat consumption, alcohol use and physical activity (PA). Carriage of rs1051296 A/G and both rs11468831 ins/del and del/del genotypes (p < 0.001) were significantly associated with CRC risk. Analysis according to gender reveals correlation of rs1051295 A/G, rs11468831 non ins/ins (p = 0.01) with CRC susceptibility regardless of patients' gender while rs3331 T/C (p = 0.012) was associated with females. Stratification study according to malignancy site; Rectal Cancer (RC) and Colon Cancer (CC), reveals increasing RC risk by gender and high meat and fat consumption, alcohol use and PA. However, additional association with high brine consumption was noted for CC. The rs1051295 A/G (p = 0.01) was associated with RC risk. Increased CC risk was associated with carriage of rs1051295 A/G, rs11168831 (del/del) and (ins/del) genotypes. CONCLUSION The risk of CRC increases with modifiable factors by Western influences on Tunisian lifestyle such as alcohol use, high fat consumption and possibly inadequate intake of vegetables. In addition, KCNB1 polymorphisms also markedly influence CRC susceptibility. Our study establishes key elements of a baseline characterization of clinical state, Western influenced lifestyle and KCNB1 variants associated with Tunisian CRC.
Collapse
Affiliation(s)
- Mouadh Barbirou
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur of Tunis, University of Tunis El Manar, 13 Place Pasteur, BP74, 1002, Tunis, Belvédère, Tunisia.,Center for Biomedical Informatics, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Henok G Woldu
- Department of Health Management and Informatics, Biostatistics & Research Design Unit School of Medicine, University of Missouri-Columbia, Columbia, MO, USA
| | - Ikram Sghaier
- Center for Biomedical Informatics, School of Medicine, University of Missouri, Columbia, MO, USA.,University of Tunis El Manar, Tunis, Tunisia
| | - Sinda A Bedoui
- Laboratory of Mycology Pathologies and Biomarkers Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Medical Oncology Division, Salah Azeiz Oncology Institute, University of Tunis El Manar, Tunis, Tunisia
| | - Amina Mokrani
- Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Radhia Aami
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur of Tunis, University of Tunis El Manar, 13 Place Pasteur, BP74, 1002, Tunis, Belvédère, Tunisia
| | - Amel Mezlini
- Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Besma Yacoubi-Loueslati
- Laboratory of Mycology Pathologies and Biomarkers Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Medical Oncology Division, Salah Azeiz Oncology Institute, University of Tunis El Manar, Tunis, Tunisia
| | - Peter J Tonellato
- Center for Biomedical Informatics, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur of Tunis, University of Tunis El Manar, 13 Place Pasteur, BP74, 1002, Tunis, Belvédère, Tunisia. .,Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
15
|
O'Malley J, Kumar R, Inigo J, Yadava N, Chandra D. Mitochondrial Stress Response and Cancer. Trends Cancer 2020; 6:688-701. [PMID: 32451306 DOI: 10.1016/j.trecan.2020.04.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/20/2022]
Abstract
Cancer cells survive and adapt to many types of stress including hypoxia, nutrient deprivation, metabolic, and oxidative stress. These stresses are sensed by diverse cellular signaling processes, leading to either degradation of mitochondria or alleviation of mitochondrial stress. This review discusses signaling during sensing and mitigation of stress involving mitochondrial communication with the endoplasmic reticulum, and how retrograde signaling upregulates the mitochondrial stress response to maintain mitochondrial integrity. The importance of the mitochondrial unfolded protein response, an emerging pathway that alleviates cellular stress, will be elaborated with respect to cancer. Detailed understanding of cellular pathways will establish mitochondrial stress response as a key mechanism for cancer cell survival leading to cancer progression and resistance, and provide a potential therapeutic target in cancer.
Collapse
Affiliation(s)
- Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Joseph Inigo
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nagendra Yadava
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
16
|
Yadav N, Gogada R, O'Malley J, Gundampati RK, Jayanthi S, Hashmi S, Lella R, Zhang D, Wang J, Kumar R, Suresh Kumar TK, Chandra D. Molecular insights on cytochrome c and nucleotide regulation of apoptosome function and its implication in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118573. [PMID: 31678591 PMCID: PMC7733678 DOI: 10.1016/j.bbamcr.2019.118573] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022]
Abstract
Cytochrome c (Cyt c) released from mitochondria interacts with Apaf-1 to form the heptameric apoptosome, which initiates the caspase cascade to execute apoptosis. Although lysine residue at 72 (K72) of Cyt c plays an important role in the Cyt c-Apaf-1 interaction, the underlying mechanism of interaction between Cyt c and Apaf-1 is still not clearly defined. Here we identified multiple lysine residues including K72, which are also known to interact with ATP, to play a key role in Cyt c-Apaf-1 interaction. Mutation of these lysine residues abrogates the apoptosome formation causing inhibition of caspase activation. Using in-silico molecular docking, we have identified Cyt c-binding interface on Apaf-1. Although mutant Cyt c shows higher affinity for Apaf-1, the presence of Cyt c-WT restores the apoptosome activity. ATP addition modulates only mutant Cyt c binding to Apaf-1 but not WT Cyt c binding to Apaf-1. Using TCGA and cBioPortal, we identified multiple mutations in both Apaf-1 and Cyt c that are predicted to interfere with apoptosome assembly. We also demonstrate that transcript levels of various enzymes involved with dATP or ATP synthesis are increased in various cancers. Silencing of nucleotide metabolizing enzymes such as ribonucleotide reductase subunit M1 (RRM1) and ATP-producing glycolytic enzymes PKM2 attenuated ATP production and enhanced caspase activation. These findings suggest important role for lysine residues of Cyt c and nucleotides in the regulation of apoptosome-dependent apoptotic cell death as well as demonstrate how these mutations and nucleotides may have a pivotal role in human diseases such as cancer.
Collapse
Affiliation(s)
- Neelu Yadav
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Raghu Gogada
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Jordan O'Malley
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Ravi Kumar Gundampati
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, United States of America
| | - Srinivas Jayanthi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, United States of America
| | - Sana Hashmi
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Ravi Lella
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Dianmu Zhang
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Jianmin Wang
- The Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | - Rahul Kumar
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America
| | | | - Dhyan Chandra
- The Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States of America.
| |
Collapse
|
17
|
Chea EE, Deredge DJ, Jones LM. Insights on the Conformational Ensemble of Cyt C Reveal a Compact State during Peroxidase Activity. Biophys J 2019; 118:128-137. [PMID: 31810655 DOI: 10.1016/j.bpj.2019.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/29/2019] [Accepted: 11/11/2019] [Indexed: 12/19/2022] Open
Abstract
Cytochrome c (cyt c) is known for its role in the electron transport chain but transitions to a peroxidase-active state upon exposure to oxidative species. The peroxidase activity ultimately results in the release of cyt c into the cytosol for the engagement of apoptosis. The accumulation of oxidative modifications that accompany the onset of the peroxidase function are well-characterized. However, the concurrent structural and conformational transitions of cyt c remain undercharacterized. Fast photochemical oxidation of proteins (FPOP) coupled with mass spectrometry is a protein footprinting technique used to structurally characterize proteins. FPOP coupled with native ion mobility separation shows that exposure to H2O2 results in the accumulation of a compact state of cyt c. Subsequent top-down fragmentation to localize FPOP modifications reveals changes in heme coordination between conformers. A time-resolved functional assay suggests that this compact conformer is peroxidase active. Altogether, combining FPOP, ion mobility separation, and top-down and bottom-up mass spectrometry allows us to discern individual conformations in solution and obtain a better understanding of the conformational ensemble and structural transitions of cyt c as it transitions from a respiratory role to a proapoptotic role.
Collapse
Affiliation(s)
- Emily E Chea
- Department of Pharmaceutical Sciences, University of Maryland Baltimore, Baltimore, Maryland
| | - Daniel J Deredge
- Department of Pharmaceutical Sciences, University of Maryland Baltimore, Baltimore, Maryland
| | - Lisa M Jones
- Department of Pharmaceutical Sciences, University of Maryland Baltimore, Baltimore, Maryland.
| |
Collapse
|
18
|
Yin S, Liu J, Kang Y, Lin Y, Li D, Shao L. Interactions of nanomaterials with ion channels and related mechanisms. Br J Pharmacol 2019; 176:3754-3774. [PMID: 31290152 DOI: 10.1111/bph.14792] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 06/10/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
The pharmacological potential of nanotechnology, especially in drug delivery and bioengineering, has developed rapidly in recent decades. Ion channels, which are easily targeted by external agents, such as nanomaterials (NMs) and synthetic drugs, due to their unique structures, have attracted increasing attention in the fields of nanotechnology and pharmacology for the treatment of ion channel-related diseases. NMs have significant effects on ion channels, and these effects are manifested in many ways, including changes in ion currents, kinetic characteristics and channel distribution. Subsequently, intracellular ion homeostasis, signalling pathways, and intracellular ion stores are affected, leading to the initiation of a range of biological processes. However, the effect of the interactions of NMs with ion channels is an interesting topic that remains obscure. In this review, we have summarized the recent research progress on the direct and indirect interactions between NMs and ion channels and discussed the related molecular mechanisms, which are crucial to the further development of ion channel-related nanotechnological applications.
Collapse
Affiliation(s)
- Suhan Yin
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jia Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuqing Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongjian Li
- Liwan District Stomatology Hospital, Guangzhou, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
19
|
Mitochondrial Entry of Cytotoxic Proteases: A New Insight into the Granzyme B Cell Death Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9165214. [PMID: 31249651 PMCID: PMC6556269 DOI: 10.1155/2019/9165214] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/08/2019] [Indexed: 02/03/2023]
Abstract
The mitochondria represent an integration and amplification hub for various death pathways including that mediated by granzyme B (GB), a granule enzyme expressed by cytotoxic lymphocytes. GB activates the proapoptotic B cell CLL/lymphoma 2 (Bcl-2) family member BH3-interacting domain death agonist (BID) to switch on the intrinsic mitochondrial death pathway, leading to Bcl-2-associated X protein (Bax)/Bcl-2 homologous antagonist/killer- (Bak-) dependent mitochondrial outer membrane permeabilization (MOMP), the dissipation of mitochondrial transmembrane potential (ΔΨm), and the production of reactive oxygen species (ROS). GB can also induce mitochondrial damage in the absence of BID, Bax, and Bak, critical for MOMP, indicating that GB targets the mitochondria in other ways. Interestingly, granzyme A (GA), GB, and caspase 3 can all directly target the mitochondrial respiratory chain complex I for ROS-dependent cell death. Studies of ROS biogenesis have revealed that GB must enter the mitochondria for ROS production, making the mitochondrial entry of cytotoxic proteases (MECP) an unexpected critical step in the granzyme death pathway. MECP requires an intact ΔΨm and is mediated though Sam50 and Tim22 channels in a mtHSP70-dependent manner. Preventing MECP severely compromises GB cytotoxicity. In this review, we provide a brief overview of the canonical mitochondrial death pathway in order to put into perspective this new insight into the GB action on the mitochondria to trigger ROS-dependent cell death.
Collapse
|
20
|
Roles of volume-regulatory anion channels, VSOR and Maxi-Cl, in apoptosis, cisplatin resistance, necrosis, ischemic cell death, stroke and myocardial infarction. CURRENT TOPICS IN MEMBRANES 2019; 83:205-283. [PMID: 31196606 DOI: 10.1016/bs.ctm.2019.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
21
|
Yu F, Tian T, Deng B, Wang T, Qi Q, Zhu M, Yan C, Ding H, Wang J, Dai J, Ma H, Ding Y, Jin G. Multi-marker analysis of genomic annotation on gastric cancer GWAS data from Chinese populations. Gastric Cancer 2019; 22:60-68. [PMID: 29859005 DOI: 10.1007/s10120-018-0841-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/24/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one of the high-incidence and high-mortality cancers all over the world. Though genome-wide association studies (GWASs) have found some genetic loci related to GC, they could only explain a small fraction of the potential pathogenesis for GC. METHODS We used multi-marker analysis of genomic annotation (MAGMA) to analyze pathways from four public pathway databases based on Chinese GWAS data including 2631 GC cases and 4373 controls. The differential expressions of selected genes in certain pathways were assessed on the basis of The Cancer Genome Atlas database. Immunohistochemistry was also conducted on 55 GC and paired normal tissues of Chinese patients to localize the expression of genes and further validate the differential expression. RESULTS We identified three pathways including chemokine signaling pathway, potassium ion import pathway, and interleukin-7 (IL7) pathway, all of which were associated with GC risk. NMI in IL7 pathway and RAC1 in chemokine signaling pathway might be two new candidate genes involved in GC pathogenesis. Additionally, NMI and RAC1 were overexpressed in GC tissues than normal tissues. CONCLUSION Immune and inflammatory associated processes and potassium transporting might participate in the development of GC. Besides, NMI and RAC1 might represent two new key genes related to GC. Our findings might give new insight into the biological mechanism and immunotherapy for GC.
Collapse
Affiliation(s)
- Fei Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Centre For Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Tian Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Nantong University, Nantong, 226019, China
| | - Bin Deng
- Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou, 225001, China
| | - Tianpei Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qi Qi
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Meng Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Caiwang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hui Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jinchen Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Centre For Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongxia Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Centre For Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Yanbing Ding
- Department of Gastroenterology, the Affiliated Hospital of Yangzhou University, Yangzhou, 225001, China.
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Centre For Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
22
|
Ramachandran RA, Lupfer C, Zaki H. The Inflammasome: Regulation of Nitric Oxide and Antimicrobial Host Defence. Adv Microb Physiol 2018; 72:65-115. [PMID: 29778217 DOI: 10.1016/bs.ampbs.2018.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is a gaseous signalling molecule that plays diverse physiological functions including antimicrobial host defence. During microbial infection, NO is synthesized by inducible NO synthase (iNOS), which is expressed by host immune cells through the recognition of microbial pattern molecules. Therefore, sensing pathogens or their pattern molecules by pattern recognition receptors (PRRs), which are located at the cell surface, endosomal and phagosomal compartment, or in the cytosol, is key in inducing iNOS and eliciting antimicrobial host defence. A group of cytosolic PRRs is involved in inducing NO and other antimicrobial molecules by forming a molecular complex called the inflammasome. Assembled inflammasomes activate inflammatory caspases, such as caspase-1 and caspase-11, which in turn process proinflammatory cytokines IL-1β and IL-18 into their mature forms and induce pyroptotic cell death. IL-1β and IL-18 play a central role in immunity against microbial infection through activation and recruitment of immune cells, induction of inflammatory molecules, and regulation of antimicrobial mediators including NO. Interestingly, NO can also regulate inflammasome activity in an autocrine and paracrine manner. Here, we discuss molecular mechanisms of inflammasome formation and the inflammasome-mediated regulation of host defence responses during microbial infections.
Collapse
Affiliation(s)
| | | | - Hasan Zaki
- UT Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
23
|
Martínez GJ, Celermajer DS, Patel S. The NLRP3 inflammasome and the emerging role of colchicine to inhibit atherosclerosis-associated inflammation. Atherosclerosis 2018; 269:262-271. [DOI: 10.1016/j.atherosclerosis.2017.12.027] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/22/2022]
|
24
|
Model MA, Petruccelli JC. Intracellular Macromolecules in Cell Volume Control and Methods of Their Quantification. CURRENT TOPICS IN MEMBRANES 2018; 81:237-289. [DOI: 10.1016/bs.ctm.2018.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
25
|
Zhang ZX, Gan I, Pavlosky A, Huang X, Fuhrmann B, Jevnikar AM. Intracellular pH Regulates TRAIL-Induced Apoptosis and Necroptosis in Endothelial Cells. J Immunol Res 2017; 2017:1503960. [PMID: 28884134 PMCID: PMC5572609 DOI: 10.1155/2017/1503960] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/26/2017] [Accepted: 07/05/2017] [Indexed: 12/22/2022] Open
Abstract
During ischemia or inflammation of organs, intracellular pH can decrease if acid production exceeds buffering capacity. Thus, the microenvironment can expose parenchymal cells to a reduced extracellular pH which can alter pH-dependent intracellular functions. We have previously shown that while silencing caspase-8 in an in vivo ischemia reperfusion injury (IRI) model results in improved organ function and survival, removal of caspase-8 function in a donor organ can paradoxically result in enhanced receptor-interacting protein kinase 1/3- (RIPK1/3-) regulated necroptosis and accelerated graft loss following transplantation. In our current study, TRAIL- (TNF-related apoptosis-inducing ligand-) induced cell death in vitro at neutral pH and caspase-8 inhibition-enhanced RIPK1-dependent necroptotic death were confirmed. In contrast, both caspase-8 inhibition and RIPK1 inhibition attenuated cell death at a cell pH of 6.7. Cell death was attenuated with mixed lineage kinase domain-like (MLKL) silencing, indicating that MLKL membrane rupture, a distinctive feature of necroptosis, occurs regardless of pH. In summary, there is a distinct regulatory control of apoptosis and necroptosis in endothelial cells at different intracellular pH. These results highlight the complexity of modulating cell death and therapeutic strategies that may need to consider different consequences on cell death dependent on the model.
Collapse
Affiliation(s)
- Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON, Canada
- Department of Pathology, Western University, London, ON, Canada
| | - Ingrid Gan
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON, Canada
- Department of Pathology, Western University, London, ON, Canada
| | - Alexander Pavlosky
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON, Canada
- Department of Pathology, Western University, London, ON, Canada
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON, Canada
| | - Benjamin Fuhrmann
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON, Canada
- Department of Microbiology & Immunology, Western University, London, ON, Canada
| | - Anthony M. Jevnikar
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, ON, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON, Canada
- Department of Microbiology & Immunology, Western University, London, ON, Canada
| |
Collapse
|
26
|
May LM, Anggono V, Gooch HM, Jang SE, Matusica D, Kerbler GM, Meunier FA, Sah P, Coulson EJ. G-Protein-Coupled Inwardly Rectifying Potassium (GIRK) Channel Activation by the p75 Neurotrophin Receptor Is Required for Amyloid β Toxicity. Front Neurosci 2017; 11:455. [PMID: 28848381 PMCID: PMC5550722 DOI: 10.3389/fnins.2017.00455] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/26/2017] [Indexed: 02/05/2023] Open
Abstract
Alzheimer's disease is characterized by cognitive decline, neuronal degeneration, and the accumulation of amyloid-beta (Aβ). Although, the neurotoxic Aβ peptide is widely believed to trigger neuronal dysfunction and degeneration in Alzheimer's disease, the mechanism by which this occurs is poorly defined. Here we describe a novel, Aβ-triggered apoptotic pathway in which Aβ treatment leads to the upregulation of G-protein activated inwardly rectifying potassium (GIRK/Kir3) channels, causing potassium efflux from neurons and Aβ-mediated apoptosis. Although, GIRK channel activity is required for Aβ-induced neuronal degeneration, we show that it is not sufficient, with coincident signaling by the p75 neurotrophin receptor (p75NTR) also required for potassium efflux and cell death. Our results identify a novel role for GIRK channels in mediating apoptosis, and provide a previously missing mechanistic link between the excitotoxicity of Aβ and its ability to trigger cell death pathways, such as that mediated by p75NTR. We propose that this death-signaling pathway contributes to the dysfunction of neurons in Alzheimer's disease and is responsible for their eventual degeneration.
Collapse
Affiliation(s)
- Linda M May
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Victor Anggono
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia
| | - Helen M Gooch
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Se E Jang
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia
| | - Dusan Matusica
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Centre for Neuroscience, College of Medicine and Public Health, Flinders UniversityAdelaide, SA, Australia
| | - Georg M Kerbler
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Frederic A Meunier
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia
| | - Pankaj Sah
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Elizabeth J Coulson
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia.,School of Biomedical Sciences, University of QueenslandBrisbane, QLD, Australia
| |
Collapse
|
27
|
Greve AS, Skals M, Fagerberg SK, Tonnus W, Ellermann-Eriksen S, Evans RJ, Linkermann A, Praetorius HA. P2X 1, P2X 4, and P2X 7 Receptor Knock Out Mice Expose Differential Outcome of Sepsis Induced by α-Haemolysin Producing Escherichia coli. Front Cell Infect Microbiol 2017; 7:113. [PMID: 28428949 PMCID: PMC5382212 DOI: 10.3389/fcimb.2017.00113] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/21/2017] [Indexed: 11/13/2022] Open
Abstract
α-haemolysin (HlyA)-producing Escherichia coli commonly inflict severe urinary tract infections, including pyelonephritis, which comprises substantial risk for sepsis. In vitro, the cytolytic effect of HlyA is mainly mediated by ATP release through the HlyA pore and subsequent P2X1/P2X7 receptor activation. This amplification of the lytic process is not unique to HlyA but is observed by many other pore-forming proteins including complement-induced haemolysis. Since free hemoglobin in the blood is known to be associated with a worse outcome in sepsis one could speculate that inhibition of P2X receptors would ameliorate the course of sepsis. Surprisingly, this study demonstrates that [Formula: see text] and [Formula: see text] mice are exceedingly sensitive to sepsis with uropathogenic E. coli. These mice have markedly lower survival, higher cytokine levels and activated intravascular coagulation. Quite the reverse is seen in [Formula: see text] mice, which had markedly lower cytokine levels and less coagulation activation compared to controls after exposure to uropathogenic E. coli. The high cytokine levels in the [Formula: see text] mouse are unexpected, since P2X7 is implicated in caspase-1-dependent IL-1β production. Here, we demonstrate that IL-1β production during sepsis with uropathogenic E. coli is mediated by caspase-8, since caspase-8 and RIPK3 double knock out mice show substantially lower cytokine during sepsis and increased survival after injection of TNFα. These data support that P2X7 and P2X4 receptor activation has a protective effect during severe E. coli infection.
Collapse
Affiliation(s)
| | - Marianne Skals
- Department of Biomedicine, Aarhus UniversityAarhus, Denmark.,Department of Clinical Microbiology, Aarhus University HospitalAarhus, Denmark
| | | | - Wulf Tonnus
- Division of Nephrology, Medical Clinic III, University Hospital Carl Gustav Carus DresdenDresden, Germany
| | | | - Richard J Evans
- Department of Molecular and Cell Biology, University of LeicesterLeicester, UK
| | - Andreas Linkermann
- Division of Nephrology, Medical Clinic III, University Hospital Carl Gustav Carus DresdenDresden, Germany
| | | |
Collapse
|
28
|
Shakeri R, Kheirollahi A, Davoodi J. Apaf-1: Regulation and function in cell death. Biochimie 2017; 135:111-125. [PMID: 28192157 DOI: 10.1016/j.biochi.2017.02.001] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 01/08/2023]
Abstract
Apoptosis, a form of programmed cell death, is responsible for eliminating damaged or unnecessary cells in multicellular organisms. Various types of intracellular stress trigger apoptosis by induction of cytochrome c release from mitochondria into the cytosol. Apoptotic protease activating factor-1 (Apaf-1) is a key molecule in the intrinsic or mitochondrial pathway of apoptosis, which oligomerizes in response to cytochrome c release and forms a large complex known as apoptosome. Procaspase-9, an initiator caspase in the mitochondrial pathway, is recruited and activated by the apoptosome leading to downstream caspase-3 processing. Various cellular proteins and small molecules can modulate apoptosome formation and function directly or indirectly. Despite recent progress in understanding the mitochondrial pathway of apoptosis, numerous questions such as the molecular mechanism of Apaf-1 oligomerization and caspase-9 activation remain poorly understood. In addition, reports have emerged showing non-apoptotic functions for Apaf-1. The current review summarizes the latest findings regarding structure-function relationship of Apaf-1 as well as its modifiers.
Collapse
Affiliation(s)
- Raheleh Shakeri
- Department of Biological Science and Biotechnology, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Asma Kheirollahi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Jamshid Davoodi
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran.
| |
Collapse
|
29
|
Multi-parametric imaging of cell heterogeneity in apoptosis analysis. Methods 2017; 112:105-123. [DOI: 10.1016/j.ymeth.2016.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/14/2016] [Accepted: 07/05/2016] [Indexed: 12/13/2022] Open
|
30
|
Guignabert C, Phan C, Seferian A, Huertas A, Tu L, Thuillet R, Sattler C, Le Hiress M, Tamura Y, Jutant EM, Chaumais MC, Bouchet S, Manéglier B, Molimard M, Rousselot P, Sitbon O, Simonneau G, Montani D, Humbert M. Dasatinib induces lung vascular toxicity and predisposes to pulmonary hypertension. J Clin Invest 2016; 126:3207-18. [PMID: 27482885 DOI: 10.1172/jci86249] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 05/26/2016] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease that can be induced by dasatinib, a dual Src and BCR-ABL tyrosine kinase inhibitor that is used to treat chronic myelogenous leukemia (CML). Today, key questions remain regarding the mechanisms involved in the long-term development of dasatinib-induced PAH. Here, we demonstrated that chronic dasatinib therapy causes pulmonary endothelial damage in humans and rodents. We found that dasatinib treatment attenuated hypoxic pulmonary vasoconstriction responses and increased susceptibility to experimental pulmonary hypertension (PH) in rats, but these effects were absent in rats treated with imatinib, another BCR-ABL tyrosine kinase inhibitor. Furthermore, dasatinib treatment induced pulmonary endothelial cell apoptosis in a dose-dependent manner, while imatinib did not. Dasatinib treatment mediated endothelial cell dysfunction via increased production of ROS that was independent of Src family kinases. Consistent with these findings, we observed elevations in markers of endothelial dysfunction and vascular damage in the serum of CML patients who were treated with dasatinib, compared with CML patients treated with imatinib. Taken together, our findings indicate that dasatinib causes pulmonary vascular damage, induction of ER stress, and mitochondrial ROS production, which leads to increased susceptibility to PH development.
Collapse
|
31
|
Jagot-Lacoussiere L, Kotula E, Villoutreix BO, Bruzzoni-Giovanelli H, Poyet JL. A Cell-Penetrating Peptide Targeting AAC-11 Specifically Induces Cancer Cells Death. Cancer Res 2016; 76:5479-90. [DOI: 10.1158/0008-5472.can-16-0302] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/24/2016] [Indexed: 11/16/2022]
|
32
|
Abstract
Inflammasome biology is one of the most exciting and rapidly growing areas in immunology. Over the past 10 years, inflammasomes have been recognized for their roles in the host defense against invading pathogens and in the development of cancer, auto-inflammatory, metabolic, and neurodegenerative diseases. Assembly of an inflammasome complex requires cytosolic sensing of pathogen-associated molecular patterns or danger-associated molecular patterns by a nucleotide-binding domain and leucine-rich repeat receptor (NLR) or absent in melanoma 2 (AIM2)-like receptors (ALR). NLRs and ALRs engage caspase-1, in most cases requiring the adapter protein apoptosis-associated speck-like protein containing a CARD (ASC), to catalyze proteolytic cleavage of pro-interleukin-1β (pro-IL-1β) and pro-IL-18 and drive pyroptosis. Recent studies indicate that caspase-8, caspase-11, IL-1R-associated kinases (IRAK), and receptor-interacting protein (RIP) kinases contribute to inflammasome functions. In addition, post-translational modifications, including ubiquitination, deubiquitination, phosphorylation, and degradation control almost every aspect of inflammasome activities. Genetic studies indicate that mutations in NLRP1, NLRP3, NLRC4, and AIM2 are linked with the development of auto-inflammatory diseases, enterocolitis, and cancer. Overall, these findings transform our understanding of the basic biology and clinical relevance of inflammasomes. In this review, we provide an overview of the latest development of inflammasome research and discuss how inflammasome activities govern health and disease.
Collapse
Affiliation(s)
- Si Ming Man
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
33
|
Langlais C, Hughes MA, Cain K, MacFarlane M. In Vitro Assembly and Analysis of the Apoptosome Complex. Cold Spring Harb Protoc 2015; 2015:pdb.prot087080. [PMID: 26631121 DOI: 10.1101/pdb.prot087080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
This protocol describes an in vitro model for studying the mechanisms of caspase activation and native apoptosome complex assembly in cell-free extracts. Active caspases in dATP-activated lysates are detected by fluorimetry using a tetrapeptide substrate (DEVD) tagged with a fluorophore (AFC), which, when released, produces a real-time readout for caspase-3 and -7 (DEVDase) activity. Gel filtration is used to isolate the apoptosome complex from the activated lysates, and assembly of Apaf-1 and caspase-9 from their monomeric forms into the multiprotein apoptosome can be confirmed via western blot. Apoptosome complex activity can be shown by incubation with exogenous procaspase-3 and -7 followed by fluorimetric bioassay (to confirm functionality of the processed effector caspases) and/or western blotting (for detection of cleaved caspase-3 and -7). A method for preparation of free procaspases for the bioassay is also described.
Collapse
Affiliation(s)
- Claudia Langlais
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| | - Michelle A Hughes
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| | - Kelvin Cain
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| | - Marion MacFarlane
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| |
Collapse
|
34
|
Langlais C, Hughes MA, Cain K, MacFarlane M. Biochemical Analysis of Initiator Caspase-Activating Complexes: The Apoptosome and the Death-Inducing Signaling Complex. Cold Spring Harb Protoc 2015; 2015:pdb.top070326. [PMID: 26631130 DOI: 10.1101/pdb.top070326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Apoptosis is a highly regulated process that can be initiated by activation of death receptors or perturbation of mitochondria causing the release of apoptogenic proteins. This results in the activation of caspases, which are responsible for many of the biochemical and morphological changes associated with apoptosis. Caspases are normally inactive and require activation in a cascade emanating from an "initiator" or activating caspase, which in turn activates a downstream or "effector" caspase. Activation of initiator caspases is tightly regulated and requires the assembly of caspase-9 (via mitochondrial perturbation) or caspase-8/10 (via death receptor ligation) activating complexes, which are termed the apoptosome and the death-inducing signaling complex (DISC), respectively. These large multiprotein complexes can initially be separated according to size by gel filtration chromatography and subsequently analyzed by affinity purification or immunoprecipitation. The advantage of combining these techniques is one can first assess the assembly of individual components into a multiprotein complex, and then assess the size and composition of the native functional signaling platform within a particular cell type alongside a biochemical analysis of the enriched/purified complex. Here, we describe various methods currently used for characterization of the apoptosome and DISC.
Collapse
Affiliation(s)
- Claudia Langlais
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| | - Michelle A Hughes
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| | - Kelvin Cain
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| | - Marion MacFarlane
- MRC Toxicology Unit, Hodgkin Building, Leicester LE1 9HN, United Kingdom
| |
Collapse
|
35
|
Ubels JL, Glupker CD, Schotanus MP, Haarsma LD. Involvement of the extrinsic and intrinsic pathways in ultraviolet B-induced apoptosis of corneal epithelial cells. Exp Eye Res 2015; 145:26-35. [PMID: 26559338 DOI: 10.1016/j.exer.2015.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/26/2015] [Accepted: 11/03/2015] [Indexed: 01/19/2023]
Abstract
The goal of this study was to elucidate the pathway by which UVB initiates efflux of K(+) and subsequently apoptosis in human corneal limbal epithelial (HCLE) cells. The initial focus of the study was on the extrinsic pathway involving Fas. HCLE cells transfected with Fas siRNA were exposed to 80-150 mJ/cm(2) UVB and incubated in culture medium with 5.5 mM K(+). Knockdown of Fas resulted in limited reduction in UVB-induced caspase-8 and -3 activity. Patch-clamp recordings showed no difference in UVB-induced normalized K(+) currents between Fas transfected and control cells. Knockdown of caspase-8 had no effect on the activation of caspase-3 following UVB exposure, while a caspase-8 inhibitor completely eliminated UVB activation of caspase-3. This suggests that caspase-8 is a robust enzyme, able to activate caspase-3 via residual caspase-8 present after knockdown, and that caspase-8 is directly involved in the UVB activation of caspase-3. Inhibition of caspase-9 significantly decreased the activation of caspases-8 and -3 in response to UVB. Knockdown of Apaf-1, required for activation of caspase-9, resulted in a significant reduction in UVB-induced activation of caspases-9, -8, and -3. Knockdown of Apaf-1 also inhibited intrinsic and UVB-induced levels of apoptosis, as determined by DNA fragmentation measured by TUNEL assay. In UVB exposed cultures treated with caspase-3 inhibitor, the percentage of apoptotic cells was reduced to control levels, confirming the necessity of caspase-3 activation in DNA fragmentation. The lack of effect of Fas knockdown on K(+) channel activation, as well as the limited effect on activation of caspases-8 and -3, strongly suggest that Fas and the extrinsic pathway is not of primary importance in the initiation of apoptosis in response to UVB in HCLE cells. Inhibition of caspase-8 and -3 activation following inhibition of caspase-9, as well as reduction in activation of caspases-9, -8, and -3 and DNA fragmentation in response to Apaf-1 knockdown support the conclusion that the intrinsic pathway is more important in UVB-induced apoptosis in HCLE cells.
Collapse
Affiliation(s)
- John L Ubels
- Department of Biology, Calvin College, Grand Rapids, MI, USA.
| | | | | | - Loren D Haarsma
- Department of Physics and Astronomy, Calvin College, Grand Rapids, MI, USA
| |
Collapse
|
36
|
Transcriptomic changes in Ca²⁺-depleted cells: Role of elevated intracellular [Na⁺]/[K⁺] ratio. Cell Calcium 2015; 58:317-24. [PMID: 26183762 DOI: 10.1016/j.ceca.2015.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/01/2015] [Accepted: 06/26/2015] [Indexed: 11/20/2022]
Abstract
Previously, we reported that Ca(2+) depletion increased permeability of the plasma membrane for Na(+). This study examined the relative impact of [Na(+)]i/[K(+)]i-mediated signaling on transcriptomic changes in cultured vascular smooth muscle cells from rat aorta (VSMC) subjected to Ca(2+)-depletion by extra-(EGTA) and intracellular (BAPTA-AM) Ca(2+) chelators. Na(+),K(+)-ATPase inhibition in K(+)-free medium during 3 h led to elevation of [Na(+)]i and attenuation of [K(+)]i by ∼7- and 10-fold, whereas Ca(2+)-depletion resulted in alteration of these parameters by ∼3- and 2-fold, respectively. Augmented VSMC permeability for Na(+) and elevation of the [Na(+)]i/[K(+)]i ratio was triggered by addition to Ca(2+)-free medium 50 μM EGTA and was not affected by 10 μM BAPTA-AM. Na(+),K(+)-ATPase inhibition and Ca(2+)-depletion changed expression of 3677 and 4610 mRNA transcripts, respectively. We found highly significant (p<10(-12)) positive (R(2)>0.51) correlation between levels of expression of 2071 transcripts whose expression was affected by both stimuli. Among genes whose expression in Ca(2+)-depleted cells was augmented by more than 7-fold we noted cyclic AMP-dependent transcription factor Atf3, early growth response protein Egr1 and nuclear receptor subfamily 4, group A member Nr4a1. Dissipation of transmembrane gradients of monovalent cations in high-K(+), low-Na(+)-medium abolished the increments of the [Na(+)]i/[K(+)]i ratio as well as the augmented expression of these genes triggered by incubation of VSMC in EGTA containing medium. Thus, our results demonstrate, for the first time, that robust transcriptomic changes triggered by Ca(2+)-depletion in the presence of extracellular Ca(2+)-chelators are at least partially mediated by elevation of the [Na(+)]i/[K(+)]i ratio and activation of Ca(2+)i-independent, [Na(+)]i/[K(+)]i-mediated mechanism of excitation-transcription coupling. These results shad a new light on analysis of data obtained in cells subjected to long-term exposure to Ca(2+) chelators.
Collapse
|
37
|
Chen J, Chen AY, Huang H, Ye X, Rollyson WD, Perry HE, Brown KC, Rojanasakul Y, Rankin GO, Dasgupta P, Chen YC. The flavonoid nobiletin inhibits tumor growth and angiogenesis of ovarian cancers via the Akt pathway. Int J Oncol 2015; 46:2629-38. [PMID: 25845666 PMCID: PMC4441297 DOI: 10.3892/ijo.2015.2946] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/19/2015] [Indexed: 11/24/2022] Open
Abstract
Despite its importance, the death rate of ovarian cancer has remained unchanged over the past five decades, demanding an improvement in prevention and treatment of this malignancy. With no known carcinogens, targeted prevention is currently unavailable, and efforts in early detection of this malignancy by screening biomarkers have failed. The inhibition of angiogenesis, also known as angioprevention, is a promising strategy to limit the growth of solid tumors, including ovarian cancers. Nobiletin, a polymethoxy flavonoid compound isolated from the tiansheng plant, has been shown to inhibit the growth of multiple types of human cancers. However, there are no reports involving the effect on nobiletin on human ovarian cancer. The present report shows that nobiletin potently decreases the viability of ovarian cancer cells in vitro. However, nobiletin does not affect the viability of normal ovarian epithelial cells at <40 μM. The antitumor activity of nobiletin was also observed in athymic mouse models and in chicken chorioallantoic membrane (CAM) models. The anti-neoplastic activity of nobiletin was due to its ability to inhibit angiogenesis. We also studied the molecular mechanisms by which nobiletin suppresses angiogenesis. We observed that nobiletin inhibits secretion of the key angiogenesis mediators, Akt, HIF-1α, NF-κB and vascular epithelial growth factor (VEGF) by ovarian cancer cells. Transient transfection experiments showed that nobiletin inhibits production of HIF-1α by downregulation of Akt. Such decreased levels of HIF-1α were responsible for nobiletin-induced suppression of VEGF. Our data suggest that nobiletin may be a promising anti-angiogenic agent relevant for therapy of ovarian cancers.
Collapse
Affiliation(s)
- Jianchu Chen
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, P.R. China
| | - Allen Y Chen
- Department of Pharmaceutical Science, West Virginia University, Morgantown, WV 26506, USA
| | - Haizhi Huang
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, P.R. China
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, P.R. China
| | - William D Rollyson
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Haley E Perry
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Kathleen C Brown
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Science, West Virginia University, Morgantown, WV 26506, USA
| | - Gary O Rankin
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Piyali Dasgupta
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Yi Charlie Chen
- College of Science, Technology and Mathematics, Alderson Broaddus University, Philippi, WV 26416, USA
| |
Collapse
|
38
|
Barghouth PG, Thiruvalluvan M, Oviedo NJ. Bioelectrical regulation of cell cycle and the planarian model system. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2629-37. [PMID: 25749155 DOI: 10.1016/j.bbamem.2015.02.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/13/2015] [Accepted: 02/23/2015] [Indexed: 12/27/2022]
Abstract
Cell cycle regulation through the manipulation of endogenous membrane potentials offers tremendous opportunities to control cellular processes during tissue repair and cancer formation. However, the molecular mechanisms by which biophysical signals modulate the cell cycle remain underappreciated and poorly understood. Cells in complex organisms generate and maintain a constant voltage gradient across the plasma membrane known as the transmembrane potential. This potential, generated through the combined efforts of various ion transporters, pumps and channels, is known to drive a wide range of cellular processes such as cellular proliferation, migration and tissue regeneration while its deregulation can lead to tumorigenesis. These cellular regulatory events, coordinated by ionic flow, correspond to a new and exciting field termed molecular bioelectricity. We aim to present a brief discussion on the biophysical machinery involving membrane potential and the mechanisms mediating cell cycle progression and cancer transformation. Furthermore, we present the planarian Schmidtea mediterranea as a tractable model system for understanding principles behind molecular bioelectricity at both the cellular and organismal level. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Paul G Barghouth
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California at Merced, 5200 North Lake Road, Merced, CA 95343, USA; Quantitative and Systems Biology Graduate Program, University of California at Merced, 5200 North Lake Road, Merced, CA 95343, USA
| | - Manish Thiruvalluvan
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California at Merced, 5200 North Lake Road, Merced, CA 95343, USA; Quantitative and Systems Biology Graduate Program, University of California at Merced, 5200 North Lake Road, Merced, CA 95343, USA
| | - Néstor J Oviedo
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California at Merced, 5200 North Lake Road, Merced, CA 95343, USA; Quantitative and Systems Biology Graduate Program, University of California at Merced, 5200 North Lake Road, Merced, CA 95343, USA; Health Sciences Research Institute, University of California at Merced, 5200 North Lake Road, Merced, CA 95343, USA.
| |
Collapse
|
39
|
Activation of NLRP3 inflammasome by crystalline structures via cell surface contact. Sci Rep 2014; 4:7281. [PMID: 25445147 PMCID: PMC4250918 DOI: 10.1038/srep07281] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/13/2014] [Indexed: 12/28/2022] Open
Abstract
Crystalline structures activate the NLRP3 inflammasome, leading to the production of IL-1β, however, the molecular interactions responsible for NLRP3 activation are not fully understood. Cathepsin B release from the ruptured phagolysosome and potassium ion efflux have been suggested to be critical for this activation. Here, we report that Cathepsin B redistribution was not a crucial event in crystal-induced IL-1β production. Silica and monosodium urate crystal-treated macrophages with undisturbed lysosomes demonstrated strong co-localization of ASC and Caspase-1, indicative of NLRP3 inflammasome activation. Importantly, we provided evidence to suggest that macrophage cell membrane binding to immobilized crystals was sufficient to induce IL-1β release, and this activation of the NLRP3 inflammasome was inhibited by blocking potassium efflux. Therefore, this work reveals additional complexity in crystalline structure-mediated NLRP3 inflammasome regulations.
Collapse
|
40
|
Ryland KE, Svoboda LK, Vesely ED, McIntyre JC, Zhang L, Martens JR, Lawlor ER. Polycomb-dependent repression of the potassium channel-encoding gene KCNA5 promotes cancer cell survival under conditions of stress. Oncogene 2014; 34:4591-600. [PMID: 25435365 PMCID: PMC4451446 DOI: 10.1038/onc.2014.384] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/23/2014] [Accepted: 10/10/2014] [Indexed: 02/08/2023]
Abstract
Relapse after clinical remission remains a leading cause of cancer-associated death. Although the mechanisms of tumor relapse are complex, the ability of cancer cells to survive physiological stress is a prerequisite for recurrence. Ewing sarcoma (ES) and neuroblastoma (NB) are aggressive cancers that frequently relapse after initial remission. In addition, both tumors overexpress the polycomb group (PcG) proteins BMI-1 and EZH2, which contribute to tumorigenicity. We have discovered that ES and NB resist hypoxic stress-induced death and that survival depends on PcG function. Epigenetic repression of developmental programs is the most well-established cancer-associated function of PcG proteins. However, we noted that voltage-gated potassium (Kv) channel genes are also targets of PcG regulation in stem cells. Given the role of potassium in regulating apoptosis, we reasoned that repression of Kv channel genes might have a role in cancer cell survival. Here we describe our novel finding that PcG-dependent repression of the Kv1.5 channel gene KCNA5 contributes to cancer cell survival under conditions of stress. We show that survival of cancer cells in stress is dependent upon suppression of Kv1.5 channel function. The KCNA5 promoter is marked in cancer cells with PcG-dependent chromatin repressive modifications that increase in hypoxia. Genetic and pharmacological inhibition of BMI-1 and EZH2, respectively, restore KCNA5 expression, which sensitizes cells to stress-induced death. In addition, ectopic expression of the Kv1.5 channel induces apoptotic cell death under conditions of hypoxia. These findings identify a novel role for PcG proteins in promoting cancer cell survival via repression of KCNA5.
Collapse
Affiliation(s)
- K E Ryland
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA.,Translational Oncology Program, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - L K Svoboda
- Translational Oncology Program, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - E D Vesely
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - J C McIntyre
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - L Zhang
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - J R Martens
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - E R Lawlor
- Translational Oncology Program, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA.,Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
41
|
Yee AG, Lee SM, Hunter MR, Glass M, Freestone PS, Lipski J. Effects of the Parkinsonian toxin MPP+ on electrophysiological properties of nigral dopaminergic neurons. Neurotoxicology 2014; 45:1-11. [DOI: 10.1016/j.neuro.2014.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/22/2014] [Accepted: 08/25/2014] [Indexed: 10/24/2022]
|
42
|
Caspase-1 autoproteolysis is differentially required for NLRP1b and NLRP3 inflammasome function. Proc Natl Acad Sci U S A 2014; 111:17254-9. [PMID: 25404286 DOI: 10.1073/pnas.1415756111] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Inflammasomes are caspase-1-activating multiprotein complexes. The mouse nucleotide-binding domain and leucine rich repeat pyrin containing 1b (NLRP1b) inflammasome was identified as the sensor of Bacillus anthracis lethal toxin (LT) in mouse macrophages from sensitive strains such as BALB/c. Upon exposure to LT, the NLRP1b inflammasome activates caspase-1 to produce mature IL-1β and induce pyroptosis. Both processes are believed to depend on autoproteolysed caspase-1. In contrast to human NLRP1, mouse NLRP1b lacks an N-terminal pyrin domain (PYD), indicating that the assembly of the NLRP1b inflammasome does not require the adaptor apoptosis-associated speck-like protein containing a CARD (ASC). LT-induced NLRP1b inflammasome activation was shown to be impaired upon inhibition of potassium efflux, which is known to play a major role in NLRP3 inflammasome formation and ASC dimerization. We investigated whether NLRP3 and/or ASC were required for caspase-1 activation upon LT stimulation in the BALB/c background. The NLRP1b inflammasome activation was assessed in both macrophages and dendritic cells lacking either ASC or NLRP3. Upon LT treatment, the absence of NLRP3 did not alter the NLRP1b inflammasome activity. Surprisingly, the absence of ASC resulted in IL-1β cleavage and pyroptosis, despite the absence of caspase-1 autoprocessing activity. By reconstituting caspase-1/caspase-11(-/-) cells with a noncleavable or catalytically inactive mutant version of caspase-1, we directly demonstrated that noncleavable caspase-1 is fully active in response to the NLRP1b activator LT, whereas it is nonfunctional in response to the NLRP3 activator nigericin. Taken together, these results establish variable requirements for caspase-1 cleavage depending on the pathogen and the responding NLR.
Collapse
|
43
|
Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N. Ion channels in the regulation of apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2532-46. [PMID: 25450339 DOI: 10.1016/j.bbamem.2014.10.030] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/08/2014] [Accepted: 10/20/2014] [Indexed: 02/07/2023]
Abstract
Apoptosis, a type of genetically controlled cell death, is a fundamental cellular mechanism utilized by multicellular organisms for disposal of cells that are no longer needed or potentially detrimental. Given the crucial role of apoptosis in physiology, deregulation of apoptotic machinery is associated with various diseases as well as abnormalities in development. Acquired resistance to apoptosis represents the common feature of most and perhaps all types of cancer. Therefore, repairing and reactivating apoptosis represents a promising strategy to fight cancer. Accumulated evidence identifies ion channels as essential regulators of apoptosis. However, the contribution of specific ion channels to apoptosis varies greatly depending on cell type, ion channel type and intracellular localization, pathology as well as intracellular signaling pathways involved. Here we discuss the involvement of major types of ion channels in apoptosis regulation. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Artem Kondratskyi
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Kateryna Kondratska
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Roman Skryma
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France.
| |
Collapse
|
44
|
Gama V, Swahari V, Schafer J, Kole AJ, Evans A, Huang Y, Cliffe A, Golitz B, Sciaky N, Pei XH, Xiong Y, Deshmukh M. The E3 ligase PARC mediates the degradation of cytosolic cytochrome c to promote survival in neurons and cancer cells. Sci Signal 2014; 7:ra67. [PMID: 25028717 DOI: 10.1126/scisignal.2005309] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The ability to withstand mitochondrial damage is especially critical for the survival of postmitotic cells, such as neurons. Likewise, cancer cells can also survive mitochondrial stress. We found that cytochrome c (Cyt c), which induces apoptosis upon its release from damaged mitochondria, is targeted for proteasome-mediated degradation in mouse neurons, cardiomyocytes, and myotubes and in human glioma and neuroblastoma cells, but not in proliferating human fibroblasts. In mouse neurons, apoptotic protease-activating factor 1 (Apaf-1) prevented the proteasome-dependent degradation of Cyt c in response to induced mitochondrial stress. An RNA interference screen in U-87 MG glioma cells identified p53-associated Parkin-like cytoplasmic protein (PARC, also known as CUL9) as an E3 ligase that targets Cyt c for degradation. The abundance of PARC positively correlated with differentiation in mouse neurons, and overexpression of PARC reduced the abundance of mitochondrially-released cytosolic Cyt c in various cancer cell lines and in mouse embryonic fibroblasts. Conversely, neurons from Parc-deficient mice had increased sensitivity to mitochondrial damage, and neuroblastoma or glioma cells in which PARC or ubiquitin was knocked down had increased abundance of mitochondrially-released cytosolic Cyt c and decreased viability in response to stress. These findings suggest that PARC-mediated ubiquitination and degradation of Cyt c is a strategy engaged by both neurons and cancer cells to prevent apoptosis during conditions of mitochondrial stress.
Collapse
Affiliation(s)
- Vivian Gama
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Vijay Swahari
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Johanna Schafer
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Adam J Kole
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Allyson Evans
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yolanda Huang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Anna Cliffe
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian Golitz
- UNC RNAi Screening Facility, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Noah Sciaky
- UNC RNAi Screening Facility, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xin-Hai Pei
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yue Xiong
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mohanish Deshmukh
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA. Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA. Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
45
|
McStay GP, Green DR. Preparation of cytosolic extracts and activation of caspases by cytochrome c. Cold Spring Harb Protoc 2014; 2014:778-82. [PMID: 24987139 DOI: 10.1101/pdb.prot080275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
It can be useful to explore the caspase activation process in an in vitro setting. In this protocol, cytosolic extracts prepared from cell culture are incubated with cytochrome c and adenosine triphosphate (dATP), leading to the oligomerization of apoptotic protease activating factor-1 (APAF-1) and the formation of the apoptosome. The apoptosome serves as an activation platform for caspase-9, which binds to the apoptosome through heterodimeric caspase recruitment domain (CARD) interactions and then dimerizes. This leads to cleavage of the executioner, caspase-3. These extracts contain highly active caspases that can be analyzed using a variety of biochemical assays.
Collapse
Affiliation(s)
- Gavin P McStay
- Department of Life Sciences, New York Institute of Technology, Old Westbury, New York 11568
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| |
Collapse
|
46
|
Abstract
Monitoring the activity of a caspase, either as an isolated protein or in a complex mixture (e.g., a cytosolic extract), can be achieved by measuring substrate cleavage. Chromogenic or fluorogenic substrates are available for many caspases. These substrates usually consist of the four-amino-acid motif that is optimal for each caspase and a moiety that, when cleaved, generates either a chromophore or a fluorophore that can be detected using spectrophotometric or fluorimetric means. In this protocol, we describe how to use these substrates to monitor caspase activity in samples containing active caspases (e.g., apoptotic cells). Caspase inhibitors, which contain a moiety that covalently attaches to the active site of the caspase, can be used in these assays. These assays will ascertain whether caspases are involved in a specific process (e.g., whether caspases are activated after an apoptotic stimulus) and are particularly informative if a purified caspase is used. However, the substrates and inhibitors are not specific for a particular caspase in an environment containing multiple caspases. So, if cytosolic or apoptotic cell extracts are used in these assays, additional experiments must be performed to identify exactly which caspases are involved.
Collapse
Affiliation(s)
- Gavin P McStay
- Department of Life Sciences, New York Institute of Technology, Old Westbury, New York 11568
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| |
Collapse
|
47
|
|
48
|
McCord MC, Aizenman E. The role of intracellular zinc release in aging, oxidative stress, and Alzheimer's disease. Front Aging Neurosci 2014; 6:77. [PMID: 24860495 PMCID: PMC4028997 DOI: 10.3389/fnagi.2014.00077] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/02/2014] [Indexed: 01/26/2023] Open
Abstract
Brain aging is marked by structural, chemical, and genetic changes leading to cognitive decline and impaired neural functioning. Further, aging itself is also a risk factor for a number of neurodegenerative disorders, most notably Alzheimer’s disease (AD). Many of the pathological changes associated with aging and aging-related disorders have been attributed in part to increased and unregulated production of reactive oxygen species (ROS) in the brain. ROS are produced as a physiological byproduct of various cellular processes, and are normally detoxified by enzymes and antioxidants to help maintain neuronal homeostasis. However, cellular injury can cause excessive ROS production, triggering a state of oxidative stress that can lead to neuronal cell death. ROS and intracellular zinc are intimately related, as ROS production can lead to oxidation of proteins that normally bind the metal, thereby causing the liberation of zinc in cytoplasmic compartments. Similarly, not only can zinc impair mitochondrial function, leading to excess ROS production, but it can also activate a variety of extra-mitochondrial ROS-generating signaling cascades. As such, numerous accounts of oxidative neuronal injury by ROS-producing sources appear to also require zinc. We suggest that zinc deregulation is a common, perhaps ubiquitous component of injurious oxidative processes in neurons. This review summarizes current findings on zinc dyshomeostasis-driven signaling cascades in oxidative stress and age-related neurodegeneration, with a focus on AD, in order to highlight the critical role of the intracellular liberation of the metal during oxidative neuronal injury.
Collapse
Affiliation(s)
- Meghan C McCord
- Department of Neurobiology, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| |
Collapse
|
49
|
Moschou PN, Roubelakis-Angelakis KA. Polyamines and programmed cell death. JOURNAL OF EXPERIMENTAL BOTANY 2014; 65:1285-96. [PMID: 24218329 DOI: 10.1093/jxb/ert373] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Polyamines (PAs) have been considered as important molecules for survival. However, evidence reinforces that PAs are also implicated, directly or indirectly, in pathways regulating programmed cell death (PCD). Direct correlation of PAs with cell death refers to their association with particular biological processes, and their physical contact with molecules or structures involved in cell death. Indirectly, PAs regulate PCD through their metabolic derivatives, such as catabolic and interconversion products. Cytotoxic products of PA metabolism are involved in PCD cascades, whereas it remains largely elusive how PAs directly control pathways leading to PCD. In this review, we present and compare advances in PA-dependent PCD in animals and plants.
Collapse
Affiliation(s)
- Panagiotis N Moschou
- Department of Plant Biology and Forest Genetics, Uppsala BioCenter, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, 75007 Uppsala, Sweden
| | | |
Collapse
|
50
|
Bortner CD, Cidlowski JA. Ion channels and apoptosis in cancer. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130104. [PMID: 24493752 DOI: 10.1098/rstb.2013.0104] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Humans maintain a constant cell number throughout their lifespan. This equilibrium of cell number is accomplished when cell proliferation and cell death are kept balanced, achieving a steady-state cell number. Abnormalities in cell growth or cell death can lead to an overabundance of cells known as neoplasm or tumours. While the perception of cancer is often that of an uncontrollable rate of cell growth or increased proliferation, a decrease in cell death can also lead to tumour formation. Most cells when detached from their normal tissue die. However, cancer cells evade cell death, tipping the balance to an overabundance of cell number. Therefore, overcoming this resistance to cell death is a decisive factor in the treatment of cancer. Ion channels play a critical role in cancer in regards to cell proliferation, malignant angiogenesis, migration and metastasis. Additionally, ion channels are also known to be critical components of apoptosis. In this review, we discuss the modes of cell death focusing on the ability of cancer cells to evade apoptosis. Specifically, we focus on the role ion channels play in controlling and regulating life/death decisions and how they can be used to overcome resistance to apoptosis in the treatment of cancer.
Collapse
Affiliation(s)
- Carl D Bortner
- The Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Department of Health and Human Services, National Institutes of Health, , Research Triangle Park, NC 27709, USA
| | | |
Collapse
|