1
|
Clostridioides difficile toxin B alone and with pro-inflammatory cytokines induces apoptosis in enteric glial cells by activating three different signalling pathways mediated by caspases, calpains and cathepsin B. Cell Mol Life Sci 2022; 79:442. [PMID: 35864342 PMCID: PMC9304068 DOI: 10.1007/s00018-022-04459-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Clostridioides difficile infection (CDI) causes nosocomial/antibiotic-associated gastrointestinal diseases with dramatically increasing global incidence and mortality rates. The main C. difficile virulence factors, toxins A and B (TcdA/TcdB), cause cytopathic/cytotoxic effects and inflammation. We demonstrated that TcdB induces caspase-dependent, mitochondria-independent enteric glial cell (EGC) apoptosis that is enhanced by the pro-inflammatory cytokines TNF-α and IFN-γ (CKs) by increasing caspase-3/7/9 and PARP activation. Because this cytotoxic synergism is important for CDI pathogenesis, we investigated the apoptotic pathways involved in TcdB- and TcdB + CK-induced apoptosis indepth. EGCs were pre-treated with the inhibitors BAF or Q-VD-OPh (pan-caspase), Z-DEVD-fmk (caspase-3/7), Z-IETD-fmk (caspase-8), PD150606 (calpains), and CA-074Me (cathepsin B) 1 h before TcdB exposure, while CKs were given 1.5 h after TcdB exposure, and assays were performed at 24 h. TcdB and TcdB + CKs induced apoptosis through three signalling pathways activated by calpains, caspases and cathepsins, which all are involved both in induction and execution apoptotic signalling under both conditions but to different degrees in TcdB and TcdB + CKs especially as regards to signal transduction mediated by these proteases towards downstream effects (apoptosis). Calpain activation by Ca2+ influx is the first pro-apoptotic event in TcdB- and TcdB + CK-induced EGC apoptosis and causes caspase-3, caspase-7 and PARP activation. PARP is also directly activated by calpains which are responsible of about 75% of apoptosis in TcdB and 62% in TcdB + CK which is both effector caspase-dependent and -independent. Initiator caspase-8 activation mediated by TcdB contributes to caspase-3/caspase-7 and PARP activation and is responsible of about 28% of apoptosis in both conditions. Caspase-3/caspase-7 activation is weakly responsible of apoptosis, indeed we found that it mediates 27% of apoptosis only in TcdB. Cathepsin B contributes to triggering pro-apoptotic signal and is responsible in both conditions of about 35% of apoptosis by a caspase-independent manner, and seems to regulate the caspase-3 and caspase-7 cleaved fragment levels, highlighting the complex interaction between these cysteine protease families activated during TcdB-induced apoptosis. Further a relevant difference between TcdB- and TcdB + CK-induced apoptosis is that TcdB-induced apoptosis increased slowly reaching at 72 h the value of 18.7%, while TcdB + CK-induced apoptosis increased strongly reaching at 72 h the value of 60.6%. Apoptotic signalling activation by TcdB + CKs is enriched by TNF-α-induced NF-κB signalling, inhibition of JNK activation and activation of AKT. In conclusion, the ability of C. difficile to activate three apoptotic pathways represents an important strategy to overcome resistance against its cytotoxic activity.
Collapse
|
2
|
Perez-Siles G, Ellis M, Ashe A, Grosz B, Vucic S, Kiernan MC, Morris KA, Reddel SW, Kennerson ML. A Compound Heterozygous Mutation in Calpain 1 Identifies a New Genetic Cause for Spinal Muscular Atrophy Type 4 (SMA4). Front Genet 2022; 12:801253. [PMID: 35126465 PMCID: PMC8807693 DOI: 10.3389/fgene.2021.801253] [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: 10/25/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
Spinal Muscular Atrophy (SMA) is a heterogeneous group of neuromuscular diseases characterized by degeneration of anterior horn cells of the spinal cord, leading to muscular atrophy and weakness. Although the major cause of SMA is autosomal recessive exon deletions or loss-of-function mutations of survival motor neuron 1 (SMN1) gene, next generation sequencing technologies are increasing the genetic heterogeneity of SMA. SMA type 4 (SMA4) is an adult onset, less severe form of SMA for which genetic and pathogenic causes remain elusive.Whole exome sequencing in a 30-year-old brother and sister with SMA4 identified a compound heterozygous mutation (p. G492R/p. F610C) in calpain-1 (CAPN1). Mutations in CAPN1 have been previously associated with cerebellar ataxia and hereditary spastic paraplegia. Using skin fibroblasts from a patient bearing the p. G492R/p. F610C mutation, we demonstrate reduced levels of CAPN1 protein and protease activity. Functional characterization of the SMA4 fibroblasts revealed no changes in SMN protein levels and subcellular distribution. Additional cellular pathways associated with SMA remain unaffected in the patient fibroblasts, highlighting the tissue specificity of CAPN1 dysfunction in SMA4 pathophysiology. This study provides genetic and functional evidence of CAPN1 as a novel gene for the SMA4 phenotype and expands the phenotype of CAPN1 mutation disorders.
Collapse
Affiliation(s)
- G. Perez-Siles
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- *Correspondence: G. Perez-Siles , ; M. L. Kennerson,
| | - M. Ellis
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
| | - A. Ashe
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - B. Grosz
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - S. Vucic
- Brain and Nerve Research Center, Concord Clinical School, University of Sydney, Sydney, NSW, Australia
| | - M. C. Kiernan
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - K. A. Morris
- Department of Neurology, Concord Repatriation General Hospital, Sydney, Sydney, NSW, Australia
| | - S. W. Reddel
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - M. L. Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Molecular Medicine Laboratory, Concord Repatriation General Hospital, Sydney, NSW, Australia
- *Correspondence: G. Perez-Siles , ; M. L. Kennerson,
| |
Collapse
|
3
|
Small-molecule suppression of calpastatin degradation reduces neuropathology in models of Huntington's disease. Nat Commun 2021; 12:5305. [PMID: 34489447 PMCID: PMC8421361 DOI: 10.1038/s41467-021-25651-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 08/19/2021] [Indexed: 11/08/2022] Open
Abstract
Mitochondrial dysfunction is a common hallmark of neurological disorders, and reducing mitochondrial damage is considered a promising neuroprotective therapeutic strategy. Here, we used high-throughput small molecule screening to identify CHIR99021 as a potent enhancer of mitochondrial function. CHIR99021 improved mitochondrial phenotypes and enhanced cell viability in several models of Huntington’s disease (HD), a fatal inherited neurodegenerative disorder. Notably, CHIR99201 treatment reduced HD-associated neuropathology and behavioral defects in HD mice and improved mitochondrial function and cell survival in HD patient-derived neurons. Independent of its known inhibitory activity against glycogen synthase kinase 3 (GSK3), CHIR99021 treatment in HD models suppressed the proteasomal degradation of calpastatin (CAST), and subsequently inhibited calpain activation, a well-established effector of neural death, and Drp1, a driver of mitochondrial fragmentation. Our results established CAST-Drp1 as a druggable signaling axis in HD pathogenesis and highlighted CHIR99021 as a mitochondrial function enhancer and a potential lead for developing HD therapies. Mitochondrial dysfunction is a common hallmark of neurological disorders. Here, the authors identify CHIR99021 as a potent enhancer of mitochondrial function, which improved mitochondrial phenotypes in Huntington’s disease models. CHIR99021 was shown to stabilize calpastatin, which suppressed calpain activation and Drp1-induced mitochondrial fragmentation.
Collapse
|
4
|
Meng C, Zeng W, Lv J, Wang Y, Gao M, Chang R, Li Q, Wang X. 1,8-cineole ameliorates ischaemic brain damage via TRPC6/CREB pathways in rats. J Pharm Pharmacol 2021; 73:979-985. [PMID: 33877307 DOI: 10.1093/jpp/rgab035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/12/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVES A previous in vitro study reported that the monoterpene oxide 1,8-cineole (cineole) attenuates neuronal caused by oxygen-glucose deprivation/reoxygenation in culture. However, to date, there is no in vivo evidence showing neuroprotective effects of cineole against stroke. This study aimed to investigate whether cineole attenuates cerebral ischaemic damage in rats. METHODS A rat model of middle cerebral artery occlusion (MCAO) followed by 24 h reperfusion was applied. Male rats were treated with oral cineole (100 mg/kg) for 7 consecutive days, then subjected to MCAO surgery. Infarct volume, neurologic deficits, apoptosis and expression levels of all-spectrin breakdown products of 145 kDa (SBDP145), transient receptor potential canonical (subtype) 6 (TRPC6) and phosphorylated CREB (p-CREB) were measured in ischaemic brain tissues. KEY FINDINGS Cineole treatment significantly reduced infarct volume, neurological dysfunction, neuronal apoptosis, SBDP145 formation and TRPC6 degradation and enhanced p-CREB expression in MCAO rats compared with vehicle treatment. These neuroprotective effects were markedly suppressed by pharmacological inhibition of MEK or CaMKIV signalling. CONCLUSIONS Our study provides in vivo evidence demonstrating that cineole pretreatment attenuates ischaemic stroke-induced brain damage, mainly through blocking calpain-induced TRPC6 degradation and activating CREB via MEK/CREB and CaMKIV/CREB signalling pathways.
Collapse
Affiliation(s)
- Chen Meng
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenjing Zeng
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jing Lv
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Meiling Gao
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ruijie Chang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qing Li
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xianyu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Anesthesiology, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
5
|
Great balls of fire: activation and signalling of inflammatory caspases. Biochem Soc Trans 2021; 49:1311-1324. [PMID: 34060593 PMCID: PMC8286819 DOI: 10.1042/bst20200986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 11/17/2022]
Abstract
Innate immune responses are tightly regulated by various pathways to control infections and maintain homeostasis. One of these pathways, the inflammasome pathway, activates a family of cysteine proteases called inflammatory caspases. They orchestrate an immune response by cleaving specific cellular substrates. Canonical inflammasomes activate caspase-1, whereas non-canonical inflammasomes activate caspase-4 and -5 in humans and caspase-11 in mice. Caspases are highly specific enzymes that select their substrates through diverse mechanisms. During inflammation, caspase activity is responsible for the secretion of inflammatory cytokines and the execution of a form of lytic and inflammatory cell death called pyroptosis. This review aims to bring together our current knowledge of the biochemical processes behind inflammatory caspase activation, substrate specificity, and substrate signalling.
Collapse
|
6
|
Britti E, Delaspre F, Tamarit J, Ros J. Calpain-Inhibitors Protect Frataxin-Deficient Dorsal Root Ganglia Neurons from Loss of Mitochondrial Na +/Ca 2+ Exchanger, NCLX, and Apoptosis. Neurochem Res 2021; 46:108-119. [PMID: 32249386 DOI: 10.1007/s11064-020-03020-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/19/2020] [Accepted: 03/28/2020] [Indexed: 12/16/2022]
Abstract
Calpains are calcium-dependent proteases activated in apoptotic cell death and neurodegeneration. Friedreich Ataxia is a neurodegenerative rare disease caused by frataxin deficiency, a mitochondrial protein. Dorsal root ganglion (DRG) sensory neurons are among the cellular types most affected in this disease. We have previously demonstrated that frataxin-deficient DRGs show calpain activation, alteration in calcium levels and decreased content of the Na+/Ca2+ exchanger (NCLX). This transporter is involved in mitochondrial calcium efflux. In this study, we have performed a time-course analysis of several parameters altered in a frataxin-deficient DRGs. These include decline of NCLX levels, calcium accumulation, mitochondrial depolarization, α-fodrin fragmentation and apoptotic cell death. Furthermore, we have analysed the effect of the calpain inhibitors MDL28170 and Calpeptin on these parameters. We have observed that these inhibitors increase NCLX levels, protect sensory neurons from neurite degeneration and calcium accumulation, and restore mitochondrial membrane potential. In addition, calpain 1 reduction alleviated neurodegeneration in frataxin-deficient DRG neurons. These results strengthen the hypothesis of a central role for calcium homeostasis and calpains in frataxin-deficient dorsal root ganglia neurons.
Collapse
Affiliation(s)
- Elena Britti
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Fabien Delaspre
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain.
| |
Collapse
|
7
|
de la Fuente S, Sansa A, Hidalgo I, Vivancos N, Romero-Guevara R, Garcera A, Soler RM. Calpain system is altered in survival motor neuron-reduced cells from in vitro and in vivo spinal muscular atrophy models. Cell Death Dis 2020; 11:487. [PMID: 32587237 PMCID: PMC7316821 DOI: 10.1038/s41419-020-2688-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 12/31/2022]
Abstract
Spinal muscular atrophy (SMA) is a severe neuromuscular disorder caused by loss of the survival motor neuron 1 (SMN1) gene. SMA is characterized by the degeneration of spinal cord motoneurons (MNs), progressive skeletal muscle atrophy, and weakness. The cellular and molecular mechanisms causing MN loss of function are only partially known. Recent advances in SMA research postulate the role of calpain protease regulating survival motor neuron (SMN) protein and the positive effect on SMA phenotype of treatment with calpain inhibitors. We analyzed the level of calpain pathway members in mice and human cellular SMA models. Results indicate an increase of calpain activity in SMN-reduced MNs. Spinal cord analysis of SMA mice treated with calpeptin, a calpain inhibitor, showed an increase of SMN, calpain, and its endogenous inhibitor calpastatin in MNs. Finally, in vitro calpeptin treatment prevented microtubule-associated protein 1A/1B-light chain 3 (LC3) increase in MNs neurites, indicating that calpain inhibition may reduce autophagosome accumulation in neuron prolongations, but not in soma. Thus, our results show that calpain activity is increased in SMA MNs and its inhibition may have a beneficial effect on SMA phenotype through the increase of SMN in spinal cord MNs.
Collapse
Affiliation(s)
- Sandra de la Fuente
- Neuronal Signaling Unit, Experimental Medicine Department, Universitat de Lleida-IRBLleida, Rovira Roure, 80, Lleida, 25198, Spain
| | - Alba Sansa
- Neuronal Signaling Unit, Experimental Medicine Department, Universitat de Lleida-IRBLleida, Rovira Roure, 80, Lleida, 25198, Spain
| | - Iván Hidalgo
- Neuronal Signaling Unit, Experimental Medicine Department, Universitat de Lleida-IRBLleida, Rovira Roure, 80, Lleida, 25198, Spain
| | - Nuria Vivancos
- Neuronal Signaling Unit, Experimental Medicine Department, Universitat de Lleida-IRBLleida, Rovira Roure, 80, Lleida, 25198, Spain
| | - Ricardo Romero-Guevara
- Metabolic Physiopathology Group, Experimental Medicine Department, Universitat de Lleida-IRBLleida, Lleida, Spain
| | - Ana Garcera
- Neuronal Signaling Unit, Experimental Medicine Department, Universitat de Lleida-IRBLleida, Rovira Roure, 80, Lleida, 25198, Spain
| | - Rosa M Soler
- Neuronal Signaling Unit, Experimental Medicine Department, Universitat de Lleida-IRBLleida, Rovira Roure, 80, Lleida, 25198, Spain.
| |
Collapse
|
8
|
Mourão LC, Cardoso-Oliveira GP, Braga ÉM. Autoantibodies and Malaria: Where We Stand? Insights Into Pathogenesis and Protection. Front Cell Infect Microbiol 2020; 10:262. [PMID: 32596165 PMCID: PMC7300196 DOI: 10.3389/fcimb.2020.00262] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Abstract
Autoantibodies are frequently reported in patients with malaria, but whether they contribute to protection or to pathology is an issue of debate. A large body of evidence indicates that antibodies against host-self components are associated to malaria clinical outcomes such as cerebral malaria, renal dysfunction and anemia. Nonetheless, self-reactive immunoglobulins induced during an infection can also mediate protection. In light of these controversies, we summarize here the latest findings in our understanding of autoimmune responses in malaria, focusing on Plasmodium falciparum and Plasmodium vivax. We review the main targets of self-antibody responses in malaria as well as the current, but still limited, knowledge of their role in disease pathogenesis or protection.
Collapse
Affiliation(s)
| | | | - Érika Martins Braga
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Traumatic brain injury and methamphetamine: A double-hit neurological insult. J Neurol Sci 2020; 411:116711. [DOI: 10.1016/j.jns.2020.116711] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 11/27/2019] [Accepted: 01/29/2020] [Indexed: 11/17/2022]
|
10
|
In vitro cellular uptake and neuroprotective efficacy of poly-arginine-18 (R18) and poly-ornithine-18 (O18) peptides: critical role of arginine guanidinium head groups for neuroprotection. Mol Cell Biochem 2019; 464:27-38. [DOI: 10.1007/s11010-019-03646-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
|
11
|
Li W, Yang F, Gao J, Tang Y, Wang J, Pan Y. Over-Expression of TRPC6 via CRISPR Based Synergistic Activation Mediator in BMSCs Ameliorates Brain Injury in a Rat Model of Cerebral Ischemia/Reperfusion. Neuroscience 2019; 415:147-160. [PMID: 31369718 DOI: 10.1016/j.neuroscience.2019.06.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/23/2022]
Abstract
Stroke is a major life-threatening and disabling disease with a restricted therapeutic approach. Bone marrow stromal cells (BMSCs) possess proliferative ability and a multi-directional differentiation potential, and secrete a range of trophic/growth factors that can protect neurons after cerebral ischemia/reperfusion. Transient receptor potential canonical (TRPC) is a family of non-selective channels permeable to Ca2+, with several functions including neuronal survival. Over-expression of TRPC6, a subtype of the TRPC family, was shown to protect neurons against cerebral ischemia/reperfusion injury. However, it remains unclear whether over-expression of TRPC6 in BMSCs can further reduce brain injury after ischemia/reperfusion. In the present study, we report that over-expression of TRPC6 via a CRISPR-based synergistic activation mediator in BMSCs provided a greater reduction of brain injury in a rat model of ischemia/reperfusion. Further, the improved neurofunctional outcomes were associated with increased TRPC6 and brain derived neurotrophic factor expression levels. Overall, these data suggest that TRPC6 over-expressing BMSCs may be a promising therapeutic agent for ischemic stroke.
Collapse
Affiliation(s)
- Wenbin Li
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Fan Yang
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Jinxing Gao
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Yushi Tang
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Jing Wang
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Yujun Pan
- Department of Neurology, First Clinical College of Harbin Medical University, Room 501, Building 3, 23 Youzheng Street, Harbin, Heilongjiang Province, 150001, People's Republic of China.
| |
Collapse
|
12
|
Jantos K, Kling A, Mack H, Hornberger W, Moeller A, Nimmrich V, Lao Y, Nijsen M. Discovery of ABT-957: 1-Benzyl-5-oxopyrrolidine-2-carboxamides as selective calpain inhibitors with enhanced metabolic stability. Bioorg Med Chem Lett 2019; 29:1968-1973. [DOI: 10.1016/j.bmcl.2019.05.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/13/2023]
|
13
|
Choi JH, Pile-Spellman J. Reperfusion Changes After Stroke and Practical Approaches for Neuroprotection. Neuroimaging Clin N Am 2019; 28:663-682. [PMID: 30322601 DOI: 10.1016/j.nic.2018.06.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Reperfusion is the first line of care in a growing number of eligible acute ischemic stroke patients. Early reperfusion with thrombolytic drugs and endovascular mechanical devices is associated with improved outcome and lower mortality rates compared with natural history. Reperfusion is not without risk, however, and may result in reperfusion injury, which manifests in hemorrhagic transformation, brain edema, infarct progression, and neurologic worsening. In this article, the functional and structural changes and underlying molecular mechanisms of ischemia and reperfusion are reviewed. The pathways that lead to reperfusion injury and novel neuroprotective strategies with endogenous properties are discussed.
Collapse
Affiliation(s)
- Jae H Choi
- Center for Unruptured Brain Aneurysms, Neurological Surgery PC, 1991 Marcus Avenue, Suite 108, Lake Success, NY 11042, USA; Department of Neurology, State University of New York Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; Hybernia Medical LLC, 626 RexCorp Plaza, Uniondale, NY 11556, USA.
| | - John Pile-Spellman
- Center for Unruptured Brain Aneurysms, Neurological Surgery PC, 1991 Marcus Avenue, Suite 108, Lake Success, NY 11042, USA; Hybernia Medical LLC, 626 RexCorp Plaza, Uniondale, NY 11556, USA
| |
Collapse
|
14
|
de la Fuente S, Sansa A, Periyakaruppiah A, Garcera A, Soler RM. Calpain Inhibition Increases SMN Protein in Spinal Cord Motoneurons and Ameliorates the Spinal Muscular Atrophy Phenotype in Mice. Mol Neurobiol 2018; 56:4414-4427. [PMID: 30327977 PMCID: PMC6505520 DOI: 10.1007/s12035-018-1379-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/01/2018] [Indexed: 01/07/2023]
Abstract
Spinal muscular atrophy (SMA), a leading genetic cause of infant death, is caused by the loss of survival motor neuron 1 (SMN1) gene. SMA is characterized by the degeneration and loss of spinal cord motoneurons (MNs), muscular atrophy, and weakness. SMN2 is the centromeric duplication of the SMN gene, whose numbers of copies determine the intracellular levels of SMN protein and define the disease onset and severity. It has been demonstrated that elevating SMN levels can be an important strategy in treating SMA and can be achieved by several mechanisms, including promotion of protein stability. SMN protein is a direct target of the calcium-dependent protease calpain and induces its proteolytic cleavage in muscle cells. In this study, we examined the involvement of calpain in SMN regulation on MNs. In vitro experiments showed that calpain activation induces SMN cleavage in CD1 and SMA mouse spinal cord MNs. Additionally, calpain 1 knockdown or inhibition increased SMN level and prevent neurite degeneration in these cells. We examined the effects of calpain inhibition on the phenotype of two severe SMA mouse models. Treatment with the calpain inhibitor, calpeptin, significantly improved the lifespan and motor function of these mice. Our observations show that calpain regulates SMN level in MNs and calpeptin administration improves SMA phenotype demonstrating the potential utility of calpain inhibitors in SMA therapy.
Collapse
Affiliation(s)
- Sandra de la Fuente
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Alba Sansa
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Ambika Periyakaruppiah
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Ana Garcera
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Rosa M Soler
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain.
| |
Collapse
|
15
|
Tan XL, Sun M, Brady RD, Liu S, Llanos R, Cheung S, Wright DK, Casillas-Espinosa PM, Sashindranath M, O'Brien TJ, McDonald SJ, Turner BJ, Shultz SR. Transactive Response DNA-Binding Protein 43 Abnormalities after Traumatic Brain Injury. J Neurotrauma 2018; 36:87-99. [PMID: 29901412 DOI: 10.1089/neu.2017.5491] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Initial studies have found some evidence for transactive response DNA-binding protein 43 (TDP-43) abnormalities after traumatic brain injury (TBI), and the presence of protein inclusions consisting of TDP-43 are a pathological hallmark of amyotrophic lateral sclerosis (ALS), a condition associated with TBI. However, no study has characterized changes in TDP-43 phosphorylation, mislocalization, and fragmentation (i.e., abnormalities linked to hallmark TDP-43 pathology) after TBI, and how these relate to functional outcomes. Further, how TBI affects an individual with a known predisposition to TDP-43 pathology is unknown. Therefore, this study examined the effects of TBI on TDP-43 post-translational processing, localization, and behavioral outcomes in wild-type (WT) mice and mutant TDP-43A315T mice (i.e., mice predisposed to TDP-43 pathology) at 24 h and 1 week after TBI. Post-mortem brain tissue from human patients with acute TBI was also examined. Western blots found that WT mice given TBI had increased TDP-43 phosphorylation, mislocalization, and fragmentation compared with sham-injured WT mice. The TDP-43A315T mice given a TBI had exacerbated TDP-43 abnormalities, worse cell death, and cognitive deficits compared with all other groups. In the human TBI patients, the only significant finding was increased nuclear accumulation of phosphorylated TDP-43 fragments. The discrepancy between the robust mouse findings and the largely non-significant human findings may be due to factors including heterogeneity in clinical TBI, the small group sizes, and temporal complexities with TDP-43 abnormalities. These findings indicate that TBI can induce a number of TDP-43 abnormalities that may contribute to the neurological consequences of TBI, though further research is still needed.
Collapse
Affiliation(s)
- Xin Lin Tan
- 1 Department of Medicine, The University of Melbourne , Parkville, Victoria, Australia
| | - Mujun Sun
- 1 Department of Medicine, The University of Melbourne , Parkville, Victoria, Australia
| | - Rhys D Brady
- 1 Department of Medicine, The University of Melbourne , Parkville, Victoria, Australia
- 2 Department of Neuroscience, Monash University , Melbourne, Victoria, Australia
| | - Shijie Liu
- 2 Department of Neuroscience, Monash University , Melbourne, Victoria, Australia
| | - Roxana Llanos
- 3 Life and Environmental Sciences, Deakin University , Burwood, Victoria, Australia
| | - Steve Cheung
- 3 Life and Environmental Sciences, Deakin University , Burwood, Victoria, Australia
| | - David K Wright
- 2 Department of Neuroscience, Monash University , Melbourne, Victoria, Australia
| | - Pablo M Casillas-Espinosa
- 1 Department of Medicine, The University of Melbourne , Parkville, Victoria, Australia
- 2 Department of Neuroscience, Monash University , Melbourne, Victoria, Australia
| | - Maithili Sashindranath
- 4 Australian Center for Blood Disease, Monash University , Melbourne, Victoria, Australia
| | - Terence J O'Brien
- 1 Department of Medicine, The University of Melbourne , Parkville, Victoria, Australia
- 2 Department of Neuroscience, Monash University , Melbourne, Victoria, Australia
| | - Stuart J McDonald
- 5 Physiology, Anatomy, and Microbiology, La Trobe University , Bundoora, Victoria, Australia
| | - Bradley J Turner
- 6 The Florey Institute of Neuroscience and Mental Health , Parkville, Victoria, Australia
| | - Sandy R Shultz
- 1 Department of Medicine, The University of Melbourne , Parkville, Victoria, Australia
- 2 Department of Neuroscience, Monash University , Melbourne, Victoria, Australia
| |
Collapse
|
16
|
Liu XF, Zhou DD, Xie T, Hao JL, Malik TH, Lu CB, Qi J, Pant OP, Lu CW. The Nrf2 Signaling in Retinal Ganglion Cells under Oxidative Stress in Ocular Neurodegenerative Diseases. Int J Biol Sci 2018; 14:1090-1098. [PMID: 29989056 PMCID: PMC6036726 DOI: 10.7150/ijbs.25996] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 04/22/2018] [Indexed: 12/28/2022] Open
Abstract
Retinal ganglion cells (RGCs) are one of the important cell types affected in many ocular neurodegenerative diseases. Oxidative stress is considered to be involved in retinal RGCs death in ocular neurodegenerative diseases. More and more attention has been focused on studying the agents that may have neuroprotective effects. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a key nuclear transcription factor for the systemic antioxidant defense system. This review elucidates the underlying mechanism of the Nrf2-mediated neuroprotective effects on RGCs in ocular neurodegenerative diseases, such as diabetic retinopathy and retinal ischemia-reperfusion injury. Several Nrf2 inducers that shield RGCs from oxidative stress-induced neurodegeneration via regulating Nrf2 signaling are discussed.
Collapse
Affiliation(s)
- Xiu-Fen Liu
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Dan-Dan Zhou
- Department of Radiology, The First Hospital of Jilin University, Jilin, China
| | - Tian Xie
- Department of . Neurosurgery, The People's Hospital of Jilin Province, Jilin, China
| | - Ji-Long Hao
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Tayyab Hamid Malik
- Department of Gastroenterology, The First Hospital of Jilin University, Jilin, China
| | - Cheng-Bo Lu
- Department of Cardiology, The First Hospital of Jiamusi University, Heilongjiang, China
| | - Jing Qi
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Om Prakash Pant
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| | - Cheng-Wei Lu
- Department of Ophthalmology, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
17
|
Kling A, Jantos K, Mack H, Hornberger W, Backfisch G, Lao Y, Nijsen M, Rendenbach-Mueller B, Moeller A. Mitigating the Metabolic Liability of Carbonyl Reduction: Novel Calpain Inhibitors with P1' Extension. ACS Med Chem Lett 2018. [PMID: 29541364 DOI: 10.1021/acsmedchemlett.7b00494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Dysregulation of calpains 1 and 2 has been implicated in a variety of pathological disorders including ischemia/reperfusion injuries, kidney diseases, cataract formation, and neurodegenerative diseases such as Alzheimer's disease (AD). 2-(3-Phenyl-1H)-pyrazol-1-yl)nicotinamides represent a series of novel and potent calpain inhibitors with high selectivity and in vivo efficacy. However, carbonyl reduction leading to the formation of the inactive hydroxyamide was identified as major metabolic liability in monkey and human, a pathway not reflected by routine absorption, distribution, metabolism, and excretion (ADME) assays. Using cytosolic clearance as a tailored in vitro ADME assay coupled with in vitro hepatocyte metabolism enabled the identification of analogues with enhanced stability against carbonyl reduction. These efforts led to the identification of P1' modified calpain inhibitors with significantly improved pharmacokinetic profile including P1' N-methoxyamide 23 as potential candidate compound for non-central nervous system indications.
Collapse
Affiliation(s)
- Andreas Kling
- Neuroscience Research, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Katja Jantos
- Neuroscience Research, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Helmut Mack
- Neuroscience Research, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Wilfried Hornberger
- Neuroscience Research, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Gisela Backfisch
- Development Sciences, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Yanbin Lao
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, Illinois 60064-6125, United States
| | - Marjoleen Nijsen
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, Illinois 60064-6125, United States
| | | | - Achim Moeller
- Neuroscience Research, AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| |
Collapse
|
18
|
Britti E, Delaspre F, Feldman A, Osborne M, Greif H, Tamarit J, Ros J. Frataxin-deficient neurons and mice models of Friedreich ataxia are improved by TAT-MTScs-FXN treatment. J Cell Mol Med 2018; 22:834-848. [PMID: 28980774 PMCID: PMC5783845 DOI: 10.1111/jcmm.13365] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/28/2017] [Indexed: 01/02/2023] Open
Abstract
Friedreich ataxia (FA) is a rare disease caused by deficiency of frataxin, a mitochondrial protein. As there is no cure available for this disease, many strategies have been developed to reduce the deleterious effects of such deficiency. One of these approaches is based on delivering frataxin to the tissues by coupling the protein to trans-activator of transcription (TAT) peptides, which enables cell membranes crossing. In this study, we tested the efficiency of TAT-MTScs-FXN fusion protein to decrease neurodegeneration markers on frataxin-depleted neurons obtained from dorsal root ganglia (DRG), one of the most affected tissues. In mice models of the disease, we tested the ability of TAT-MTScs-FXN to penetrate the mitochondria and its effect on lifespan. In DRG neurons, treatment with TAT-MTScs-FXN increased cell survival, decreased neurite degeneration and reduced apoptotic markers, such as α-fodrin cleavage and caspase 9 activation. Also, we show that heat-shock protein 60 (HSP60), a molecular chaperone targeted to mitochondria, suffered an impaired processing in frataxin-deficient neurons that was relieved by TAT-MTScs-FXN addition. In mice models of the disease, administration of TAT-MTScs-FXN was able to reach muscle mitochondria, restore the activity of the succinate dehydrogenase and produce a significant lifespan increase. These results support the use of TAT-MTScs-FXN as a treatment for Friedreich ataxia.
Collapse
Affiliation(s)
- Elena Britti
- Departament de Ciències Mèdiques BàsiquesIRBLleidaUniversitat de LleidaLleidaSpain
| | - Fabien Delaspre
- Departament de Ciències Mèdiques BàsiquesIRBLleidaUniversitat de LleidaLleidaSpain
| | | | | | | | - Jordi Tamarit
- Departament de Ciències Mèdiques BàsiquesIRBLleidaUniversitat de LleidaLleidaSpain
| | - Joaquim Ros
- Departament de Ciències Mèdiques BàsiquesIRBLleidaUniversitat de LleidaLleidaSpain
| |
Collapse
|
19
|
Wang FZ, Dai XL, Liu HY. Molecular mechanisms underlying the α-tomatine-directed apoptosis in human malignant glioblastoma cell lines A172 and U-118 MG. Exp Ther Med 2017; 14:6183-6192. [PMID: 29250143 DOI: 10.3892/etm.2017.5294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 03/06/2017] [Indexed: 12/25/2022] Open
Abstract
In the present study, the molecular mechanisms involved in the α-tomatine-induced apoptosis in human glioblastoma cell lines A172 and U-118 MG were investigated. Wright staining and ApopTag assays were conducted to confirm the apoptosis induced by α-tomatine treatment. Fura-2 assay determined an enhancement in free Ca2+ intracellularly, indicating the occurrence of Ca2+-dependent apoptosis induction. Western blot experiments were also performed to predict the apoptosis by measuring the changes in the Bax:Bcl-2 ratio. Increase of calpain activity triggered caspase-12 expression, which in turn further activated caspase-9. In addition, an increase in the ratio of Bax:Bcl-2 accounted for the mitochondrial release of cytochrome c into the cytosol for caspase-3 and caspase-9 activation. Elevated activity of calpain and caspase-3 yielded spectrin breakdown products with 145 and 120 kDa, respectively. Caspase-3 activation further cleaved the inhibitor of caspase activated DNase, while the apoptosis-inducing factor detected in the cytosol suggested that apoptosis was independent of caspase. The apoptosis induction was further supported by decreased expression levels of nuclear factor-κB and increased expression of the inhibitor of nuclear factor, IκBα. In conclusion, the presented experimental results revealed the stimulation of different molecular mechanisms for α-tomatine-mediated apoptosis in A172 and U-118 MG human glioblastoma cell lines.
Collapse
Affiliation(s)
- Fa-Zhao Wang
- Department of Neurosurgery, The People's Hospital of Zoucheng, Zoucheng, Shandong 273500, P.R. China
| | - Xue-Liang Dai
- Department of Neurosurgery, The People's Hospital of Zoucheng, Zoucheng, Shandong 273500, P.R. China
| | - Hong-Yi Liu
- Department of Neurosurgery, The People's Hospital of Zoucheng, Zoucheng, Shandong 273500, P.R. China
| |
Collapse
|
20
|
Maekawa S, Sato K, Fujita K, Daigaku R, Tawarayama H, Murayama N, Moritoh S, Yabana T, Shiga Y, Omodaka K, Maruyama K, Nishiguchi KM, Nakazawa T. The neuroprotective effect of hesperidin in NMDA-induced retinal injury acts by suppressing oxidative stress and excessive calpain activation. Sci Rep 2017; 7:6885. [PMID: 28761134 PMCID: PMC5537259 DOI: 10.1038/s41598-017-06969-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/22/2017] [Indexed: 12/21/2022] Open
Abstract
We found that hesperidin, a plant-derived bioflavonoid, may be a candidate agent for neuroprotective treatment in the retina, after screening 41 materials for anti-oxidative properties in a primary retinal cell culture under oxidative stress. We found that the intravitreal injection of hesperidin in mice prevented reductions in markers of the retinal ganglion cells (RGCs) and RGC death after N-methyl-D-aspartate (NMDA)-induced excitotoxicity. Hesperidin treatment also reduced calpain activation, reactive oxygen species generation and TNF-α gene expression. Finally, hesperidin treatment improved electrophysiological function, measured with visual evoked potential, and visual function, measured with optomotry. Thus, we found that hesperidin suppressed a number of cytotoxic factors associated with NMDA-induced cell death signaling, such as oxidative stress, over-activation of calpain, and inflammation, thereby protecting the RGCs in mice. Therefore, hesperidin may have potential as a therapeutic supplement for protecting the retina against the damage associated with excitotoxic injury, such as occurs in glaucoma and diabetic retinopathy.
Collapse
Affiliation(s)
- Shigeto Maekawa
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kota Sato
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kosuke Fujita
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Reiko Daigaku
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Hiroshi Tawarayama
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Namie Murayama
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Satoru Moritoh
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Takeshi Yabana
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Yukihiro Shiga
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kazuichi Maruyama
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Toru Nakazawa
- Department of Ophthalmology and Visual Science, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
| |
Collapse
|
21
|
Liu X, Song X, Yuan T, He J, Wang X, Wang Q. Effects of calpain on sevoflurane-induced aged rats hippocampal neuronal apoptosis. Aging Clin Exp Res 2016; 28:633-9. [PMID: 26482746 DOI: 10.1007/s40520-015-0466-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 09/22/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUND Sevoflurane is one of the most commonly used volatile anesthetics and it has been shown to induce widespread apoptotic neurodegeneration in aged rat. Calpain is also activated during apoptosis in several types of cells. We hypothesized that calpain resulted in apoptosis under long time sevoflurane exposure, and it might play a role in the sevoflurane-induced memory impairment in aged rats. METHODS Seventy-two 18-month-old male Sprague-Dawley rats were randomly divided into three groups (n = 24): Control group rats were exposed to simply humid 50 % O2 balanced by N2 for 3 h; While M group rats received calpain inhibitor 10 mg/kg via the tail vein intravenously at 30 min before the animals inhaled 3 % sevoflurane for 3 h, subsequently received MDL 28170 3.33 mg/kg/h for 3 h. Sev group rats were only exposed to 3 % sevoflurane for 3 h without calpain inhibitor. Morris Water Maze was used to test the ability of learning and memory. Cytosolic calcium concentration was measured by using flow cytometry. Annexin-V labeled with a fluorophore or biotin can identify apoptotic cells by binding to PS. The expression of calpain in the hippocampus of rats was tested by Western blots. RESULTS The results showed that the M group had a shorter latency and had a larger number of times crossing over the previous platform site than that of the Sev group. Compared with Sev group, apoptosis rate and 76/80 kDa ratio of μ-calpain were significantly decreased in M group on the 1st day. CONCLUSIONS Sevoflurane might induce apoptosis through increasing [Ca(2+)]c and the activity of μ-calpain, which might be identified at least partially the molecular mechanism by which sevoflurane induces apoptosis.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 ZiQiang Road, Shi Jiazhuang, 050051, China
| | - Xueyin Song
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 ZiQiang Road, Shi Jiazhuang, 050051, China
| | - Tianbao Yuan
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 ZiQiang Road, Shi Jiazhuang, 050051, China
| | - Jihua He
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 ZiQiang Road, Shi Jiazhuang, 050051, China
| | - Xiuli Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 ZiQiang Road, Shi Jiazhuang, 050051, China
| | - Qiujun Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, No. 139 ZiQiang Road, Shi Jiazhuang, 050051, China.
| |
Collapse
|
22
|
Periyakaruppiah A, de la Fuente S, Arumugam S, Bahí N, Garcera A, Soler RM. Autophagy modulators regulate survival motor neuron protein stability in motoneurons. Exp Neurol 2016; 283:287-97. [PMID: 27373203 DOI: 10.1016/j.expneurol.2016.06.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/06/2016] [Accepted: 06/28/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Ambika Periyakaruppiah
- Unitat de Senyalització Neuronal, Dep. Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Rovira Roure 80, 25198 Lleida, Spain
| | - Sandra de la Fuente
- Unitat de Senyalització Neuronal, Dep. Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Rovira Roure 80, 25198 Lleida, Spain
| | - Saravanan Arumugam
- Unitat de Senyalització Neuronal, Dep. Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Rovira Roure 80, 25198 Lleida, Spain
| | - Núria Bahí
- Unitat de Senyalització Neuronal, Dep. Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Rovira Roure 80, 25198 Lleida, Spain
| | - Ana Garcera
- Unitat de Senyalització Neuronal, Dep. Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Rovira Roure 80, 25198 Lleida, Spain
| | - Rosa M Soler
- Unitat de Senyalització Neuronal, Dep. Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Rovira Roure 80, 25198 Lleida, Spain.
| |
Collapse
|
23
|
Serapio-Palacios A, Navarro-Garcia F. EspC, an Autotransporter Protein Secreted by Enteropathogenic Escherichia coli, Causes Apoptosis and Necrosis through Caspase and Calpain Activation, Including Direct Procaspase-3 Cleavage. mBio 2016; 7:e00479-16. [PMID: 27329750 PMCID: PMC4916375 DOI: 10.1128/mbio.00479-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/12/2016] [Indexed: 01/30/2023] Open
Abstract
UNLABELLED Enteropathogenic Escherichia coli (EPEC) has the ability to antagonize host apoptosis during infection through promotion and inhibition of effectors injected by the type III secretion system (T3SS), but the total number of these effectors and the overall functional relationships between these effectors during infection are poorly understood. EspC produced by EPEC cleaves fodrin, paxillin, and focal adhesion kinase (FAK), which are also cleaved by caspases and calpains during apoptosis. Here we show the role of EspC in cell death induced by EPEC. EspC is involved in EPEC-mediated cell death and induces both apoptosis and necrosis in epithelial cells. EspC induces apoptosis through the mitochondrial apoptotic pathway by provoking (i) a decrease in the expression levels of antiapoptotic protein Bcl-2, (ii) translocation of the proapoptotic protein Bax from cytosol to mitochondria, (iii) cytochrome c release from mitochondria to the cytoplasm, (iv) loss of mitochondrial membrane potential, (v) caspase-9 activation, (vi) cleavage of procaspase-3 and (vii) an increase in caspase-3 activity, (viii) PARP proteolysis, and (ix) nuclear fragmentation and an increase in the sub-G1 population. Interestingly, EspC-induced apoptosis was triggered through a dual mechanism involving both independent and dependent functions of its EspC serine protease motif, the direct cleavage of procaspase-3 being dependent on this motif. This is the first report showing a shortcut for induction of apoptosis by the catalytic activity of an EPEC protein. Furthermore, this atypical intrinsic apoptosis appeared to induce necrosis through the activation of calpain and through the increase of intracellular calcium induced by EspC. Our data indicate that EspC plays a relevant role in cell death induced by EPEC. IMPORTANCE EspC, an autotransporter protein with serine protease activity, has cytotoxic effects on epithelial cells during EPEC infection. EspC causes cytotoxicity by cleaving fodrin, a cytoskeletal actin-associated protein, and focal adhesion proteins (i.e., FAK); interestingly, these proteins are also cleaved during apoptosis and necrosis. Here we show that EspC is able to cause cell death, which is characterized by apoptosis: by dissecting the apoptotic pathway and considering that EspC is translocated by an injectisome, we found that EspC induces the mitochondrial apoptotic pathway. Remarkably, EspC activates this pathway by two distinct mechanisms-either by using or not using its serine protease motif. Thus, we show for the first time that this serine protease motif is able to cleave procaspase-3, thereby reaching the terminal stages of caspase cascade activation leading to apoptosis. Furthermore, this overlapped apoptosis appears to potentiate cell death through necrosis, where EspC induces calpain activation and increases intracellular calcium.
Collapse
Affiliation(s)
- Antonio Serapio-Palacios
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México City, Mexico
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México City, Mexico
| |
Collapse
|
24
|
Zamzow DR, Elias V, Acosta VA, Escobedo E, Magnusson KR. Higher levels of phosphorylated Y1472 on GluN2B subunits in the frontal cortex of aged mice are associated with good spatial reference memory, but not cognitive flexibility. AGE (DORDRECHT, NETHERLANDS) 2016; 38:50. [PMID: 27094400 PMCID: PMC5005925 DOI: 10.1007/s11357-016-9913-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 04/06/2016] [Indexed: 06/05/2023]
Abstract
The N-methyl-D-aspartate receptor (NMDAr) is particularly vulnerable to aging. The GluN2B subunit of the NMDAr, compared to other NMDAr subunits, suffers the greatest losses of expression in the aging brain, especially in the frontal cortex. While expression levels of GluN2B mRNA and protein in the aged brain are well documented, there has been little investigation into age-related posttranslational modifications of the subunit. In this study, we explored some of the mechanisms that may promote differences in the NMDAr complex in the frontal cortex of aged animals. Two ages of mice, 3 and 24 months, were behaviorally tested in the Morris water maze. The frontal cortex and hippocampus from each mouse were subjected to differential centrifugation followed by solubilization in Triton X-100. Proteins from Triton-insoluble membranes, Triton-soluble membranes, and intracellular membranes/cytosol were examined by Western blot. Higher levels of GluN2B tyrosine 1472 phosphorylation in frontal cortex synaptic fractions of old mice were associated with better reference learning but poorer cognitive flexibility. Levels of GluN2B phosphotyrosine 1336 remained steady, but there were greater levels of the calpain-induced 115 kDa GluN2B cleavage product on extrasynaptic membranes in these old good learners. There was an age-related increase in calpain activity, but it was not associated with better learning. These data highlight a unique aging change for aged mice with good spatial learning that might be detrimental to cognitive flexibility. This study also suggests that higher levels of truncated GluN2B on extrasynaptic membranes are not deleterious to spatial memory in aged mice.
Collapse
Affiliation(s)
| | - Val Elias
- Oregon State University, Corvallis, OR, USA
| | | | | | | |
Collapse
|
25
|
Kim JH, Kwon SJ, Stankewich MC, Huh GY, Glantz SB, Morrow JS. Reactive protoplasmic and fibrous astrocytes contain high levels of calpain-cleaved alpha 2 spectrin. Exp Mol Pathol 2015; 100:1-7. [PMID: 26551084 DOI: 10.1016/j.yexmp.2015.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/04/2015] [Indexed: 12/13/2022]
Abstract
Calpain, a family of calcium-dependent neutral proteases, plays important roles in neurophysiology and pathology through the proteolytic modification of cytoskeletal proteins, receptors and kinases. Alpha 2 spectrin (αII spectrin) is a major substrate for this protease family, and the presence of the αII spectrin breakdown product (αΙΙ spectrin BDP) in a cell is evidence of calpain activity triggered by enhanced intracytoplasmic Ca(2+) concentrations. Astrocytes, the most dynamic CNS cells, respond to micro-environmental changes or noxious stimuli by elevating intracytoplasmic Ca(2+) concentration to become activated. As one measure of whether calpains are involved with reactive glial transformation, we examined paraffin sections of the human cerebral cortex and white matter by immunohistochemistry with an antibody specific for the calpain-mediated αΙΙ spectrin BDP. We also performed conventional double immunohistochemistry as well as immunofluorescent studies utilizing antibodies against αΙΙ spectrin BDP as well as glial fibrillary acidic protein (GFAP). We found strong immunopositivity in selected protoplasmic and fibrous astrocytes, and in transitional forms that raise the possibility of some of fibrous astrocytes emerging from protoplasmic astrocytes. Immunoreactive astrocytes were numerous in brain sections from cases with severe cardiac and/or respiratory diseases in the current study as opposed to our previous study of cases without significant clinical conditions that failed to reveal such remarkable immunohistochemical alterations. Our study suggests that astrocytes become αΙΙ spectrin BDP immunopositive in various stages of activation, and that spectrin cleavage product persists even in fully reactive astrocytes. Immunohistochemistry for αΙΙ spectrin BDP thus marks reactive astrocytes, and highlights the likelihood that calpains and their proteolytic processing of spectrin participate in the morphologic and physiologic transition from resting protoplasmic astrocytes to reactive fibrous astrocytes.
Collapse
Affiliation(s)
- Jung H Kim
- Department of Pathology, Yale Univ. School of Medicine, 310 Cedar Street, New Haven, CT 06510-8023, USA.
| | - Soojung J Kwon
- Department of Pathology, Yale Univ. School of Medicine, 310 Cedar Street, New Haven, CT 06510-8023, USA
| | - Michael C Stankewich
- Department of Pathology, Yale Univ. School of Medicine, 310 Cedar Street, New Haven, CT 06510-8023, USA
| | - Gi-Yeong Huh
- Department of Forensic Medicine, School of Medicine, Pusan National University, Pusan, Korea
| | - Susan B Glantz
- Department of Pathology, Yale Univ. School of Medicine, 310 Cedar Street, New Haven, CT 06510-8023, USA
| | - Jon S Morrow
- Department of Pathology, Yale Univ. School of Medicine, 310 Cedar Street, New Haven, CT 06510-8023, USA
| |
Collapse
|
26
|
Jantzie LL, Winer JL, Corbett CJ, Robinson S. Erythropoietin Modulates Cerebral and Serum Degradation Products from Excess Calpain Activation following Prenatal Hypoxia-Ischemia. Dev Neurosci 2015; 38:15-26. [PMID: 26551007 DOI: 10.1159/000441024] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/10/2015] [Indexed: 01/20/2023] Open
Abstract
Preterm infants suffer central nervous system (CNS) injury from hypoxia-ischemia and inflammation - termed encephalopathy of prematurity. Mature CNS injury activates caspase and calpain proteases. Erythropoietin (EPO) limits apoptosis mediated by activated caspases, but its role in modulating calpain activation has not yet been investigated extensively following injury to the developing CNS. We hypothesized that excess calpain activation degrades developmentally regulated molecules essential for CNS circuit formation, myelination and axon integrity, including neuronal potassium-chloride co-transporter (KCC2), myelin basic protein (MBP) and phosphorylated neurofilament (pNF), respectively. Further, we predicted that post-injury EPO treatment could mitigate CNS calpain-mediated degradation. Using prenatal transient systemic hypoxia-ischemia (TSHI) in rats to mimic CNS injury from extreme preterm birth, and postnatal EPO treatment with a clinically relevant dosing regimen, we found sustained postnatal excess cortical calpain activation following prenatal TSHI, as shown by the cleavage of alpha II-spectrin (αII-spectrin) into 145-kDa αII-spectrin degradation products (αII-SDPs) and p35 into p25. Postnatal expression of the endogenous calpain inhibitor calpastatin was also reduced following prenatal TSHI. Calpain substrate expression following TSHI, including cortical KCC2, MBP and NF, was modulated by postnatal EPO treatment. Calpain activation was reflected in serum levels of αII-SDPs and KCC2 fragments, and notably, EPO treatment also modulated KCC2 fragment levels. Together, these data indicate that excess calpain activity contributes to the pathogenesis of encephalopathy of prematurity. Serum biomarkers of calpain activation may detect ongoing cerebral injury and responsiveness to EPO or similar neuroprotective strategies.
Collapse
Affiliation(s)
- Lauren L Jantzie
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, Mass., USA
| | | | | | | |
Collapse
|
27
|
Inhibition of interleukin-6 abolishes the promoting effects of pair housing on post-stroke neurogenesis. Neuroscience 2015; 307:160-70. [DOI: 10.1016/j.neuroscience.2015.08.055] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/25/2015] [Accepted: 08/22/2015] [Indexed: 11/18/2022]
|
28
|
Kobeissy FH, Liu MC, Yang Z, Zhang Z, Zheng W, Glushakova O, Mondello S, Anagli J, Hayes RL, Wang KK. Degradation of βII-Spectrin Protein by Calpain-2 and Caspase-3 Under Neurotoxic and Traumatic Brain Injury Conditions. Mol Neurobiol 2015; 52:696-709. [PMID: 25270371 PMCID: PMC4383741 DOI: 10.1007/s12035-014-8898-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/10/2014] [Indexed: 12/22/2022]
Abstract
A major consequence of traumatic brain injury (TBI) is the rapid proteolytic degradation of structural cytoskeletal proteins. This process is largely reflected by the interruption of axonal transport as a result of extensive axonal injury leading to neuronal cell injury. Previous work from our group has described the extensive degradation of the axonally enriched cytoskeletal αII-spectrin protein which results in molecular signature breakdown products (BDPs) indicative of injury mechanisms and to specific protease activation both in vitro and in vivo. In the current study, we investigated the integrity of βII-spectrin protein and its proteolytic profile both in primary rat cerebrocortical cell culture under apoptotic, necrotic, and excitotoxic challenge and extended to in vivo rat model of experimental TBI (controlled cortical impact model). Interestingly, our results revealed that the intact 260-kDa βII-spectrin is degraded into major fragments (βII-spectrin breakdown products (βsBDPs)) of 110, 108, 85, and 80 kDa in rat brain (hippocampus and cortex) 48 h post-injury. These βsBDP profiles were further characterized and compared to an in vitro βII-spectrin fragmentation pattern of naive rat cortex lysate digested by calpain-2 and caspase-3. Results revealed that βII-spectrin was degraded into major fragments of 110/85 kDa by calpain-2 activation and 108/80 kDa by caspase-3 activation. These data strongly support the hypothesis that in vivo activation of multiple protease system induces structural protein proteolysis involving βII-spectrin proteolysis via a specific calpain and/or caspase-mediated pathway resulting in a signature, protease-specific βsBDPs that are dependent upon the type of neural injury mechanism. This work extends on previous published work that discusses the interplay spectrin family (αII-spectrin and βII-spectrin) and their susceptibility to protease proteolysis and their implication to neuronal cell death mechanisms.
Collapse
Affiliation(s)
- Firas H Kobeissy
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Ming Cheng Liu
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Zhihui Yang
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Zhiqun Zhang
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Wenrong Zheng
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Olena Glushakova
- Banyan Laboratory, Banyan Biomarkers, Inc., Alachua, FL 32615,
USA
| | - Stefania Mondello
- Department of Neurosciences, University of Messina, 98125
Messina, Italy
| | - John Anagli
- Banyan Laboratory, Banyan Biomarkers, Inc., Alachua, FL 32615,
USA
| | - Ronald L. Hayes
- Banyan Laboratory, Banyan Biomarkers, Inc., Alachua, FL 32615,
USA
| | - Kevin K.W. Wang
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
29
|
Messaoud NB, Yue J, Valent D, Katzarova I, López JM. Osmostress-induced apoptosis in Xenopus oocytes: role of stress protein kinases, calpains and Smac/DIABLO. PLoS One 2015; 10:e0124482. [PMID: 25866890 PMCID: PMC4395108 DOI: 10.1371/journal.pone.0124482] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Abstract
Hyperosmotic shock induces cytochrome c release and caspase-3 activation in Xenopus oocytes, but the regulators and signaling pathways involved are not well characterized. Here we show that hyperosmotic shock induces rapid calpain activation and high levels of Smac/DIABLO release from the mitochondria before significant amounts of cytochrome c are released to promote caspase-3 activation. Calpain inhibitors or EGTA microinjection delays osmostress-induced apoptosis, and blockage of Smac/DIABLO with antibodies markedly reduces cytochrome c release and caspase-3 activation. Hyperosmotic shock also activates the p38 and JNK signaling pathways very quickly. Simultaneous inhibition of both p38 and JNK pathways reduces osmostress-induced apoptosis, while sustained activation of these kinases accelerates the release of cytochrome c and caspase-3 activation. Therefore, at least four different pathways early induced by osmostress converge on the mitochondria to trigger apoptosis. Deciphering the mechanisms of hyperosmotic shock-induced apoptosis gives insight for potential treatments of human diseases that are caused by perturbations in fluid osmolarity.
Collapse
Affiliation(s)
- Nabil Ben Messaoud
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica, Facultad de Medicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Jicheng Yue
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica, Facultad de Medicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Daniel Valent
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica, Facultad de Medicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Ilina Katzarova
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica, Facultad de Medicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - José M. López
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica, Facultad de Medicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
- * E-mail:
| |
Collapse
|
30
|
Park YH, Mueller BH, McGrady NR, Ma HY, Yorio T. AMPA receptor desensitization is the determinant of AMPA receptor mediated excitotoxicity in purified retinal ganglion cells. Exp Eye Res 2015; 132:136-50. [PMID: 25643624 DOI: 10.1016/j.exer.2015.01.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/08/2014] [Accepted: 01/29/2015] [Indexed: 01/15/2023]
Abstract
The ionotropic glutamate receptors (iGLuR) have been hypothesized to play a role in neuronal pathogenesis by mediating excitotoxic death. Previous studies on iGluR in the retina have focused on two broad classes of receptors: NMDA and non-NMDA receptors including the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptor (AMPAR) and kainate receptor. In this study, we examined the role of receptor desensitization on the specific excitotoxic effects of AMPAR activation on primary retinal ganglion cells (RGCs). Purified rat RGCs were isolated from postnatal day 4-7 Sprague-Dawley rats. Calcium imaging was used to identify the functionality of the AMPARs and selectivity of the s-AMPA agonist. Phosphorylated CREB and ERK1/2 expression were performed following s-AMPA treatment. s-AMPA excitotoxicity was determined by JC-1 mitochondrial membrane depolarization assay, caspase 3/7 luciferase activity assay, immunoblot analysis for α-fodrin, and Live (calcein AM)/Dead (ethidium homodimer-1) assay. RGC cultures of 98% purity, lacking Iba1 and GFAP expression were used for the present studies. Isolated prenatal RGCs expressed calcium permeable AMPAR and s-AMPA (100 μM) treatment of cultured RGCs significantly increased phosphorylation of CREB but not that of ERK1/2. A prolonged (6 h) AMPAR activation in purified RGCs using s-AMPA (100 μM) did not depolarize the RGC mitochondrial membrane potential. In addition, treatment of cultured RGCs with s-AMPA, both in the presence and absence of trophic factors (BDNF and CNTF), did not increase caspase 3/7 activities or the cleavage of α-fodrin (neuronal apoptosis marker), as compared to untreated controls. Lastly, a significant increase in cell survival of RGCs was observed after s-AMPA treatment as compared to control untreated RGCs. However, preventing the desensitization of AMPAR with the treatment with either kainic acid (100 μM) or the combination of s-AMPA and cyclothiazide (50 μM) significantly reduced cell survivability. Activation of the AMPAR in RGCs does not appear to activate a signaling cascade to apoptosis, suggesting that RGCs in vitro are not susceptible to AMPA excitotoxicity as previously hypothesized. Conversely, preventing AMPAR desensitization through differential agonist activation caused AMPAR mediated excitotoxicity. Activation of the AMPAR in increasing CREB phosphorylation was dependent on the presence of calcium, which may help explain this action in increasing RGC survival.
Collapse
Affiliation(s)
- Yong H Park
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA.
| | - Brett H Mueller
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Nolan R McGrady
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Hai-Ying Ma
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Thomas Yorio
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
31
|
Marković V, Debeljak N, Stanojković T, Kolundžija B, Sladić D, Vujčić M, Janović B, Tanić N, Perović M, Tešić V, Antić J, Joksović MD. Anthraquinone–chalcone hybrids: Synthesis, preliminary antiproliferative evaluation and DNA-interaction studies. Eur J Med Chem 2015; 89:401-10. [DOI: 10.1016/j.ejmech.2014.10.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/15/2014] [Accepted: 10/18/2014] [Indexed: 12/01/2022]
|
32
|
Müller GJ, Hasseldam H, Rasmussen RS, Johansen FF. Dexamethasone enhances necrosis-like neuronal death in ischemic rat hippocampus involving μ-calpain activation. Exp Neurol 2014; 261:711-9. [PMID: 25135859 DOI: 10.1016/j.expneurol.2014.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/07/2014] [Accepted: 08/12/2014] [Indexed: 11/29/2022]
Abstract
Transient forebrain ischemia (TFI) leads to hippocampal CA1 pyramidal cell death which is aggravated by glucocorticoids (GC). It is unknown how GC affect apoptosis and necrosis in cerebral ischemia. We therefore investigated the co-localization of activated caspase-3 (casp-3) with apoptosis- and necrosis-like cell death morphologies in CA1 of rats treated with dexamethasone prior to TFI (DPTI). In addition, apoptosis- (casp-9, casp-3, casp-3-cleaved PARP and cleaved α-spectrin 145/150 and 120kDa) and necrosis-related (calpain-specific casp-9 cleavage, μ-calpain upregulation and cleaved α-spectrin 145/150kDa) cell death mechanisms were investigated by Western blot analysis. DPTI expedited CA1 neuronal death from day 4 to day 1 and increased the magnitude of CA1 neuronal death from 66.2% to 91.3% at day 7. Furthermore, DPTI decreased the overall (days 1-7) percentage of dying neurons displaying apoptosis-like morphology from 4.7% to 0.3% and, conversely, increased the percentage of neurons with necrosis-like morphology from 95.3% to 99.7%. In animals subjected to TFI without dexamethasone (ischemia-only), 7.4% of all dying CA1 neurons were casp-3-immunoreactive (IR), of which 3.1% co-localized with apoptosis-like and 4.3% with necrosis-like changes. By contrast, DPTI decreased the percentage of dying neurons with casp-3 IR to 1.4%, of which 0.3% co-localized with apoptosis-like changes and 1.1% with necrosis-like changes. Western blot analysis from DPTI animals showed a significant elevation of μ-calpain, a calpain-produced necrosis-related casp-9 fragment (25kDa) and cleavage of α-spectrin into 145/150kDa fragments at day 4, whereas in ischemia-only animals a significant increase of casp-3-cleaved PARP, cleavage of α-spectrin into 145/150 and 120kDa fragments was detected at day 7. We conclude that DPTI, in addition to augmenting and expediting CA1 neuronal death, causes a shift from apoptosis-like cell death to necrosis involving μ-calpain activation.
Collapse
Affiliation(s)
- Georg Johannes Müller
- Department of Biomedical Sciences, University of Copenhagen, Ole Maaloesvej 5, Copenhagen 2200, Denmark; Department of Neurology, Donauspital, Langobardenstrasse 122, A-1220 Vienna, Austria.
| | - Henrik Hasseldam
- Department of Biomedical Sciences, University of Copenhagen, Ole Maaloesvej 5, Copenhagen 2200, Denmark
| | - Rune Skovgaard Rasmussen
- Department of Biomedical Sciences, University of Copenhagen, Ole Maaloesvej 5, Copenhagen 2200, Denmark
| | - Flemming Fryd Johansen
- Department of Biomedical Sciences, University of Copenhagen, Ole Maaloesvej 5, Copenhagen 2200, Denmark
| |
Collapse
|
33
|
Zhang J, Mao X, Zhou T, Cheng X, Lin Y. IL-17A contributes to brain ischemia reperfusion injury through calpain-TRPC6 pathway in mice. Neuroscience 2014; 274:419-28. [DOI: 10.1016/j.neuroscience.2014.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/31/2014] [Accepted: 06/02/2014] [Indexed: 10/25/2022]
|
34
|
Gou-Fabregas M, Ramírez-Núñez O, Cacabelos D, Bahi N, Portero M, Garcera A, Soler RM. Calpain activation and CaMKIV reduction in spinal cords from hSOD1G93A mouse model. Mol Cell Neurosci 2014; 61:219-25. [PMID: 25063475 DOI: 10.1016/j.mcn.2014.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 07/17/2014] [Accepted: 07/21/2014] [Indexed: 01/21/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS), a severe neurodegenerative disease, affects the upper and lower motor neurons in the brain and spinal cord. In some studies, ALS disease progression has been associated with an increase in calcium-dependent degeneration processes. Motoneurons are specifically vulnerable to sustained membrane depolarization and excessive elevation of intracellular calcium concentration. The present study analyzed intracellular events in embryonic motoneurons and adult spinal cords of the hSOD1G93A ALS mouse model. We observed activation of calpain, a calcium-dependent cysteine protease that degrades a variety of substrates, and a reduction in calcium-calmodulin dependent protein kinase type IV (CaMKIV) levels in protein extracts from spinal cords obtained at several time-points of hSOD1G93A mice disease progression. However, in cultured embryonic motoneurons these differences between controls and hSOD1G93A mutants are not evident. Our results support the hypothesis that age-dependent changes in calcium homeostasis and resulting events, e.g., calpain activation and CaMKIV processing, are involved in ALS pathogenesis.
Collapse
Affiliation(s)
- Myriam Gou-Fabregas
- Unitat de Senyalització Neuronal, Dept Ciencies Mediques Basiques, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Rovira Roure, 80, 25198 Lleida, Spain
| | - Omar Ramírez-Núñez
- Dept Medicina Experimental, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Rovira Roure, 80, 25198 Lleida, Spain
| | - Daniel Cacabelos
- Dept Medicina Experimental, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Rovira Roure, 80, 25198 Lleida, Spain
| | - Nuria Bahi
- Unitat de Senyalització Neuronal, Dept Ciencies Mediques Basiques, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Rovira Roure, 80, 25198 Lleida, Spain
| | - Manuel Portero
- Dept Medicina Experimental, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Rovira Roure, 80, 25198 Lleida, Spain
| | - Ana Garcera
- Unitat de Senyalització Neuronal, Dept Ciencies Mediques Basiques, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Rovira Roure, 80, 25198 Lleida, Spain
| | - Rosa M Soler
- Unitat de Senyalització Neuronal, Dept Ciencies Mediques Basiques, Facultat de Medicina, Universitat de Lleida, IRBLLEIDA, Rovira Roure, 80, 25198 Lleida, Spain.
| |
Collapse
|
35
|
Recent updates on drug abuse analyzed by neuroproteomics studies: Cocaine, Methamphetamine and MDMA. TRANSLATIONAL PROTEOMICS 2014. [DOI: 10.1016/j.trprot.2014.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
36
|
Limoniastrum guyonianum aqueous gall extract induces apoptosis in colorectal cancer cells by inhibiting calpain activity. Tumour Biol 2014; 35:7877-85. [PMID: 24828012 DOI: 10.1007/s13277-014-1993-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/17/2014] [Indexed: 01/09/2023] Open
Abstract
Several studies have reported that plant-derived natural products have cancer chemopreventive and chemotherapeutic properties. The aim of the present study was to determine the antiproliferative and pro-apoptotic potential of Limoniastrum guyonianum aqueous gall extract (G extract) on human colorectal cancer BE cell line and, if so, to characterize the mechanism involved. The G extract-induced growth inhibitory effect was associated with an arrest of cell cycle progression in G2/M phase as shown by the cell phase distribution. In addition, G extract promoted in a concentration-dependent manner these cells towards apoptosis as indicated by the presence of cleaved poly(ADP-ribose) polymerase (PARP). In order to characterize the mechanism involved in the antiproliferative and pro-apoptotic signaling pathway activated by G extract, calpain activity and the expression of the cell cycle inhibitor p16(INK4A) were determined. The present findings indicated that G extract exhibited significant inhibitory activity against calpain and caused a marked and concentration-dependent upregulation of p16(INK4A). These effects could be ascribed to the presence of condensed tannins and polyphenols such as epicatechin and epigallocatechin gallate in G extract.
Collapse
|
37
|
Suzuki R, Oka T, Tamada Y, Shearer TR, Azuma M. Degeneration and dysfunction of retinal neurons in acute ocular hypertensive rats: involvement of calpains. J Ocul Pharmacol Ther 2014; 30:419-28. [PMID: 24660785 DOI: 10.1089/jop.2013.0100] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Retinal ischemic diseases primarily lead to damage of the inner retinal neurons. Electrophysiological studies also suggest impairment of the inner retinal neurons. Our recent studies with acute ocular hypertensive rats confirmed damage predominantly in the inner retinal layer along with the ganglion cell layer, changes that are ameliorated by the calpain inhibitor SNJ-1945. However, we do not know which specific neuronal cells in the inner retinal layer are damaged by calpains. Thus, the purpose of the present study was to identify specific calpain-damaged neuronal cells in the inner retina from acute ocular hypertensive rats. METHODS Intraocular pressure was elevated to 110 mm Hg for 40 min. One hour after ocular hypertension (OH), SNJ-1945 was administrated as a single oral dose of 50 mg/kg. Retinal function was assessed by scotopic electroretinography (ERG). Histological degeneration was evaluated by hematoxylin and eosin, terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end-labeling (TUNEL), and immunostaining in thin sections and flat mounts of the retina. Calpain activation was determined by proteolysis of the calpain substrate α-spectrin. RESULTS OH caused calpain activation, increased TUNEL-positive staining, decreased thickness of the inner nuclear layer (INL), and decreased amplitudes of the ERG a- and b-waves and oscillatory potentials (OPs). SNJ-1945 significantly inhibited calpain activation and the decrease in ERG values. Interestingly, the changes in the b-wave and OPs amplitudes were significantly correlated to changes in the thickness of the INL. In the inner retinal layer, the numbers of rod bipolar, cone-ON bipolar, and amacrine cells were decreased after OH. SNJ-1945 suppressed the loss of cone-ON bipolar and amacrine cells, but did not inhibit the loss of rod bipolar cells. We also observed increased glial fibrillary acid protein-positive staining in the Müller cells after OH and the treatment with SNJ-1945. CONCLUSIONS Calpains may contribute to ischemic retinal dysfunction by causing the loss of cone-ON bipolar and amacrine cells and causing the activation of Müller cells. Calpain inhibitor SNJ-1945 may be a candidate compound for treatment of retinal ischemic disease.
Collapse
Affiliation(s)
- Rie Suzuki
- 1 Senju Laboratory of Ocular Sciences, Senju Pharmaceutical Co., Ltd. , Kobe, Japan
| | | | | | | | | |
Collapse
|
38
|
Konze SA, van Diepen L, Schröder A, Olmer R, Möller H, Pich A, Weißmann R, Kuss AW, Zweigerdt R, Buettner FFR. Cleavage of E-cadherin and β-catenin by calpain affects Wnt signaling and spheroid formation in suspension cultures of human pluripotent stem cells. Mol Cell Proteomics 2014; 13:990-1007. [PMID: 24482122 DOI: 10.1074/mcp.m113.033423] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The envisioned clinical and industrial use of human pluripotent stem cells and their derivatives has given major momentum to the establishment of suspension culture protocols that enable the mass production of cells. Understanding molecular changes accompanying the transfer from adherent to suspension culture is of utmost importance because this information can have a direct effect on the development of optimized culture conditions. In this study we assessed the gene expression of human embryonic stem cells and induced pluripotent stem cells grown in surface-adherent culture (two-dimensional) versus free-floating suspension culture spheroids (three-dimensional). We combined a quantitative proteomic approach based on stable isotope labeling by amino acids in cell culture with deep-sequencing-based transcriptomics. Cells in three-dimensional culture showed reduced expression of proteins forming structural components of cell-cell and cell-extracellular matrix junctions. However, fully unexpected, we found up-regulation of secreted inhibitors of the canonical Wnt signaling pathway and, concomitantly, a reduction in the level of active β-catenin and in the expression of Wnt target genes. In Western blot analyses the cysteine protease calpain was shown to cleave E-cadherin and β-catenin under three-dimensional culture conditions. Our data allowed the development of a model in which calpain cleavage of E-cadherin induces the disintegration of focal cell contacts and generates a 100-kDa E-cadherin fragment required for the formation of three-dimensional cell-cell contacts in spheroids. The parallel release of β-catenin and its potential activation by calpain cleavage are counterbalanced by the overexpression of soluble Wnt pathway inhibitors. According to this model, calpain has a key function in the interplay between E-cadherin and β-catenin-mediated intercellular adhesion and the canonical Wnt signaling pathway. Supporting this model, we show that pharmacological modulation of calpain activity prevents spheroid formation and causes disassembly of preexisting spheroids into single cells, thereby providing novel strategies for improving suspension culture conditions for human pluripotent stem cells in the future.
Collapse
Affiliation(s)
- Sarah A Konze
- Institute for Cellular Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
YAO CHENGYE, ZHANG JIANCHENG, LIU GONGPING, CHEN FANG, LIN YUN. Neuroprotection by (−)-epigallocatechin-3-gallate in a rat model of stroke is mediated through inhibition of endoplasmic reticulum stress. Mol Med Rep 2013; 9:69-76. [DOI: 10.3892/mmr.2013.1778] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 10/25/2013] [Indexed: 11/06/2022] Open
|
40
|
Protective effects of resveratrol in experimental retinal detachment. PLoS One 2013; 8:e75735. [PMID: 24040416 PMCID: PMC3770540 DOI: 10.1371/journal.pone.0075735] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 08/21/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Oxidative stress is one of the major factors that trigger photoreceptor apoptosis. To investigate whether resveratrol, a potent antioxidant and small molecule activator of the FoxO pathway, would be neuroprotective against photoreceptor cell death in a rodent model of retinal detachment. METHODS Retinal detachment was created in adult Brown Norway rats by subretinal injection of sodium hyaluronate. The animals were treated daily with vehicle or resveratrol (20 mg/kg) intraperitoneal injection. Photoreceptor death was assessed by counting the number of apoptotic cells with TdT-dUTP terminal nick-end labeling (TUNEL) and measurement of the outer nuclear layer (ONL) thickness 3 days after RD. Changes in expression of FoxO1a, FoxO3a, and FoxO4 were analyzed by western blot. The activity of caspase 3, caspase 8, caspase 9, spectrin and their cleavage forms were studied. RESULTS Three days after retinal detachment, caspase 3, caspase 8 and caspase 9 were significantly activated in the detached retina. Spectrin cleavage products at 120 and 145 kDa were also detected. Both caspase and calpain activation are involved in apoptotic photoreceptor cell death in detached retinas. Treatment with resveratrol increases FoxO1a, FoxO3a, and FoxO4 protein expression in detached retinas only. Resveratrol treatment decreases activation of intrinsic and extrinsic caspase apoptotic pathways triggered by RD. The number of TUNEL-positive cells decreases from 1301±51 cells/mm(2) in control groups to 430±35 cells/mm(2) in treatment groups (p<0.05). Resveratrol treatment also demonstrates 59% less ONL thickness loss compared to controls. CONCLUSIONS Resveratrol treatment up-regulates the FoxO family and blocks Caspase3, 8, and 9 activation. Resveratrol has the potential to be used as a novel therapeutic agent for preventing vision loss in diseases characterized by photoreceptor detachment.
Collapse
|
41
|
Manfredi LH, Zanon NM, Garófalo MA, Navegantes LCC, Kettelhut IC. Effect of short-term cold exposure on skeletal muscle protein breakdown in rats. J Appl Physiol (1985) 2013; 115:1496-505. [PMID: 23908317 DOI: 10.1152/japplphysiol.00474.2013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Although it is well established that carbohydrate and lipid metabolism are profoundly altered by cold stress, the effects of short-term cold exposure on protein metabolism in skeletal muscle are still poorly understood. Because cold acclimation requires that an organism adjust its metabolic flux, and muscle amino acids may be an important energy source for heat production, we hypothesize that muscle proteolysis is increased and protein synthesis is decreased under such a stress condition. Herein, cold exposure for 24 h decreased rates of protein synthesis and increased overall proteolysis in both soleus and extensor digitorum longus (EDL) muscles, but it did not affect muscle weight. An increase in proteolysis was accompanied by hyperactivity of the ubiquitin-proteasome system (UPS) in both soleus and EDL, and Ca(2+)-dependent proteolysis in EDL. Furthermore, muscles of rats exposed to cold showed increased mRNA and protein levels of atrogin-1 and muscle RING finger enzyme-1 (MuRF1). Additionally, cold stress reduced phosphorylation of Akt and Forkhead box class O1 (FoxO1), a well-known effect that increases FoxO translocation to the nucleus and leads to activation of proteolysis. Plasma insulin levels were lower, whereas catecholamines, corticosterone, and thyroid hormones were higher in cold-exposed rats compared with control rats. The present data provide the first direct evidence that short-term cold exposure for 24 h decreases rates of protein synthesis and increases the UPS and Ca(2+)-dependent proteolytic processes, and increases expression of atrogin-1 and MuRF1 in skeletal muscles of young rats. The activation of atrophy induced by acute cold stress seems to be mediated at least in part through the inactivation of Akt/FoxO signaling and activation of AMP-activated protein kinase.
Collapse
Affiliation(s)
- L H Manfredi
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
42
|
Ayuso MI, Martínez-Alonso E, Cid C, Alonso de Leciñana M, Alcázar A. The translational repressor eIF4E-binding protein 2 (4E-BP2) correlates with selective delayed neuronal death after ischemia. J Cereb Blood Flow Metab 2013; 33:1173-81. [PMID: 23591646 PMCID: PMC3734765 DOI: 10.1038/jcbfm.2013.60] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 02/20/2013] [Accepted: 03/18/2013] [Indexed: 01/08/2023]
Abstract
Transient brain ischemia induces an inhibition of translational rates and causes delayed neuronal death in selective regions and cognitive deficits, whereas these effects do not occur in resistant areas. The translational repressor eukaryotic initiation factor (eIF) 4E-binding protein-2 (4E-BP2) specifically binds to eIF4E and is critical in the control of protein synthesis. To link neuronal death to translation inhibition, we study the eIF4E association with 4E-BP2 under ischemia reperfusion in a rat model of transient forebrain ischemia. Upon reperfusion, a selective neuronal apoptosis in the hippocampal cornu ammonis 1 (CA1) region was induced, while it did not occur in the cerebral cortex. Confocal microscopy analysis showed a decrease in 4E-BP2/eIF4E colocalization in resistant cortical neurons after reperfusion. In contrast, in vulnerable CA1 neurons, 4E-BP2 remains associated to eIF4E with a higher degree of 4E-BP2/eIF4E colocalization and translation inhibition. Furthermore, the binding of a 4E-BP2 peptide to eIF4E induced neuronal apoptosis in the CA1 region. Finally, pharmacological-induced protection of CA1 neurons inhibited neuronal apoptosis, decreased 4E-BP2/eIF4E association, and recovered translation. These findings documented specific changes in 4E-BP2/eIF4E association during ischemic reperfusion, linking the translation inhibition to selective neuronal death, and identifying 4E-BP2 as a novel target for protection of vulnerable neurons in ischemic injury.
Collapse
Affiliation(s)
- María Irene Ayuso
- Department of Investigation, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| | | | | | | | | |
Collapse
|
43
|
Han C, Zhao R, Kroger J, Qu M, Wani AA, Wang QE. Caspase-2 short isoform interacts with membrane-associated cytoskeleton proteins to inhibit apoptosis. PLoS One 2013; 8:e67033. [PMID: 23840868 PMCID: PMC3698186 DOI: 10.1371/journal.pone.0067033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/13/2013] [Indexed: 12/21/2022] Open
Abstract
Caspase-2 (casp-2) is the most conserved caspase across species, and is one of the initiator caspases activated by various stimuli. The casp-2 gene produces several alternative splicing isoforms. It is believed that the long isoform, casp-2L, promotes apoptosis, whereas the short isoform, casp-2S, inhibits apoptosis. The actual effect of casp-2S on apoptosis is still controversial, however, and the underlying mechanism for casp-2S-mediated apoptosis inhibition is unclear. Here, we analyzed the effects of casp-2S on DNA damage induced apoptosis through "gain-of-function" and "loss-of-function" strategies in ovarian cancer cell lines. We clearly demonstrated that the over-expression of casp-2S inhibited, and the knockdown of casp-2S promoted, the cisplatin-induced apoptosis of ovarian cancer cells. To explore the mechanism by which casp-2S mediates apoptosis inhibition, we analyzed the proteins which interact with casp-2S in cells by using immunoprecipitation (IP) and mass spectrometry. We have identified two cytoskeleton proteins, Fodrin and α-Actinin 4, which interact with FLAG-tagged casp-2S in HeLa cells and confirmed this interaction through reciprocal IP. We further demonstrated that casp-2S (i) is responsible for inhibiting DNA damage-induced cytoplasmic Fodrin cleavage independent of cellular p53 status, and (ii) prevents cisplatin-induced membrane blebbing. Taken together, our data suggests that casp-2S affects cellular apoptosis through its interaction with membrane-associated cytoskeletal Fodrin protein.
Collapse
Affiliation(s)
- Chunhua Han
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Ran Zhao
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - John Kroger
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Meihua Qu
- Department of Pharmacology, Weifang Medical University, Weifang, China
| | - Altaf A. Wani
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Qi-En Wang
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
44
|
Shear DA, Tortella FC. A military-centered approach to neuroprotection for traumatic brain injury. Front Neurol 2013; 4:73. [PMID: 23781213 PMCID: PMC3679469 DOI: 10.3389/fneur.2013.00073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/31/2013] [Indexed: 12/14/2022] Open
Abstract
Studies in animals show that many compounds and therapeutics have the potential to greatly reduce the morbidity and post-injury clinical sequela for soldiers experiencing TBI. However, to date there are no FDA approved drugs for the treatment of TBI. In fact, expert opinion suggests that combination therapies will be necessary to treat any stage of TBI recovery. Our approach to this research effort is to conduct comprehensive pre-clinical neuroprotection studies in military-relevant animal models of TBI using the most promising neuroprotective agents. In addition, emerging efforts incorporating novel treatment strategies such as stem cell based therapies and alternative therapeutic approaches will be discussed. The development of a non-surgical, non-invasive brain injury therapeutic clearly addresses a major, unresolved medical problem for the Combat Casualty Care Research Program. Since drug discovery is too expensive to be pursued by DOD in the TBI arena, this effort capitalizes on partnerships with the Private Sector (Pharmaceutical Companies) and academic collaborations (Operation Brain Trauma Therapy Consortium) to study therapies already under advanced development. Candidate therapies selected for research include drugs that are aimed at reducing the acute and delayed effects of the traumatic incident, stem cell therapies aimed at brain repair, and selective brain cooling to stabilize cerebral metabolism. Each of these efforts can also focus on combination therapies targeting multiple mechanisms of neuronal injury.
Collapse
Affiliation(s)
- Deborah A. Shear
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Frank C. Tortella
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
45
|
Cao M, Prima V, Nelson D, Svetlov S. Composite fatty acid ether amides suppress growth of liver cancer cells in vitro and in an in vivo allograft mouse model. Cell Oncol (Dordr) 2013; 36:247-57. [PMID: 23619943 DOI: 10.1007/s13402-013-0132-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The heterogeneity of liver cancer, in particular hepatocellular carcinoma (HCC), portrays the requirement of multiple targets for both its treatment and prevention. Multifaceted agents, minimally or non-toxic for normal hepatocytes, are required to address the molecular diversity of HCC, including the resistance of putative liver cancer stem cells to chemotherapy. METHODS We designed and synthesized two fatty acid ethers of isopropylamino propanol, C16:0-AIP-1 and C18:1-AIP-2 (jointly named AIPs), and evaluated their anti-proliferative effects on the human HCC cell line Huh7 and the murine hepatoma cell line BNL 1MEA.7R.1, both in vitro and in an in vivo allograft mouse model. RESULTS We found that AIP-1 and AIP-2 inhibited proliferation and caused cell death in both Huh7 and BNL 1MEA.7R.1 cells. Importantly, AIP-1 and AIP-2 were found to block the activation of putative liver cancer stem cells as manifested by suppression of clonal 'carcinosphere' development in growth factor-free and anchorage-free medium. The AIPs exhibited a relatively low toxicity against normal human or rat hepatocytes in primary cultures. In addition, we found that the AIPs utilized multifaceted pathways that mediate both autophagy and apoptosis in HCC, including the inhibition of AKTs and CAMK-1. In immune-competent mice, the AIPs significantly reduced BNL 1MEA.7R.1 cell-driven tumor allograft development, with a higher efficiency than sorafenib. A combination of AIP-1 + AIP-2 was most effective in reducing the tumor allograft incidence. CONCLUSIONS AIPs represent a novel class of simple fatty acid derivatives that are effective against liver tumors via diverse pathways. They show a low toxicity towards normal hepatocytes. The addition of AIPs may represent a new avenue towards the management of chronic liver injury and, ultimately, the prevention and treatment of HCC.
Collapse
Affiliation(s)
- Mengde Cao
- Banyan Laboratories, Inc., Alachua, FL, 32615, USA.
| | | | | | | |
Collapse
|
46
|
Knox R, Zhao C, Miguel-Perez D, Wang S, Yuan J, Ferriero D, Jiang X. Enhanced NMDA receptor tyrosine phosphorylation and increased brain injury following neonatal hypoxia-ischemia in mice with neuronal Fyn overexpression. Neurobiol Dis 2013; 51:113-9. [PMID: 23127881 PMCID: PMC3595007 DOI: 10.1016/j.nbd.2012.10.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 09/12/2012] [Accepted: 10/26/2012] [Indexed: 10/27/2022] Open
Abstract
The Src family kinases (SFKs) Src and Fyn are implicated in hypoxic-ischemic (HI) injury in the developing brain. However, it is unclear how these particular SFKs contribute to brain injury. Using neuron-specific Fyn overexpressing (OE) mice, we investigated the role of neuronal Fyn in neonatal brain HI. Wild type (WT) and Fyn OE mice were subjected to HI using the Vannucci model at postnatal day 7. Brains were scored five days later for evaluation of damage using cresyl violet and iron staining. Western blotting with postsynaptic density (PSD)-associated synaptic membrane proteins and co-immunoprecipitation with cortical lysates were performed at various time points after HI to determine NMDA receptor tyrosine phosphorylation and Fyn kinase activity. Fyn OE mice had significantly higher mortality and brain injury compared to their WT littermates. Neuronal Fyn overexpression led to sustained NR2A and NR2B tyrosine phosphorylation and enhanced NR2B phosphorylation at tyrosine (Y) 1472 and Y1252 in synaptic membranes. These early changes correlated with higher calpain activity 24h after HI in Fyn OE mice relative to WT animals. Our findings suggest a role for Fyn kinase in neuronal death after neonatal HI, possibly via up-regulation of NMDA receptor tyrosine phosphorylation.
Collapse
Affiliation(s)
- Renatta Knox
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, 94158, USA
- Medical Scientist Training Program, University of California, San Francisco, CA, 94158, USA
| | - Chong Zhao
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Dario Miguel-Perez
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Steven Wang
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Jinwei Yuan
- Icon Clinical Research, Redwood City, CA 94065, USA
| | - Donna Ferriero
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA, 94158, USA
| | - Xiangning Jiang
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
47
|
Induction of apoptosis by low-molecular-weight fucoidan through calcium- and caspase-dependent mitochondrial pathways in MDA-MB-231 breast cancer cells. Biosci Biotechnol Biochem 2013; 77:235-42. [PMID: 23391903 DOI: 10.1271/bbb.120631] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fucoidan, a fucose-rich polysaccharide extracted from brown seaweed, has antitumor, anticoagulant, antiviral, anti-inflammatory, and antibacterial activities. Several studies have shown that a fucoidan treatment of cancer cells induced cytotoxicity and apoptosis and inhibited angiogenesis and invasion. We investigated in the present study the effect of low-molecular-weight fucoidan (LMWF) on apoptosis in estrogen receptor-negative MDA-MB-231 human breast cancer cells. The LMWF treatment of MDA-MB-231 cells was associated with the activation of caspases and mitochondrial dysfunction, including dissipation of the mitochondrial membrane potential (ΔΨm), alteration of Ca(2+) homeostasis, cytochrome c release, and decreased expression of antiapoptotic Bcl-2 family proteins. Understanding the molecular events that mediated LMWF-induced MDA-MB-231 cell death will contribute to a more rational approach to cancer chemotherapy.
Collapse
|
48
|
Hyperforin attenuates brain damage induced by transient middle cerebral artery occlusion (MCAO) in rats via inhibition of TRPC6 channels degradation. J Cereb Blood Flow Metab 2013; 33:253-62. [PMID: 23149561 PMCID: PMC3564196 DOI: 10.1038/jcbfm.2012.164] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hyperforin, a lipophilic constituent of medicinal herb St John's wort, has been identified as the main active ingredient of St John's wort extract for antidepressant action by experimental and clinical studies. Hyperforin is currently known to activate transient receptor potential canonical (subtype) 6 (TRPC6) channel, increase the phosphorylated CREB (p-CREB), and has N-methyl-D-aspartate receptor-antagonistic effect that convert potential neuroprotective effects in vitro. However, the protective effects of hyperforin on ischemic stroke in vivo remain controversial and its neuroprotective mechanisms are still unclear. This study was designed to examine the effects of intracerebroventricular (i.c.v.) injection of hyperforin on transient focal cerebral ischemia in rats. Hyperforin, when applied immediately after middle cerebral artery occlusion (MCAO) onset, significantly reduced infarct volumes and apoptotic cells, and also increased neurologic scores at 24 hours after reperfusion accompanied by elevated TRPC6 and p-CREB activity and decreased SBDP145 activity. When MEK or CaMKIV activity was specifically inhibited, the neuroprotective effect of hyperforin was attenuated, and we observed a correlated decrease in CREB activity. In conclusion, our results clearly showed that i.c.v. injection of hyperforin immediately after MCAO onset blocked calpain-mediated TRPC6 channels degradation, and then to stimulate the Ras/MEK/ERK and CaMKIV pathways that converge on CREB activation, contributed to neuroprotection.
Collapse
|
49
|
Actin cytoskeleton manipulation by effector proteins secreted by diarrheagenic Escherichia coli pathotypes. BIOMED RESEARCH INTERNATIONAL 2012; 2013:374395. [PMID: 23509714 PMCID: PMC3591105 DOI: 10.1155/2013/374395] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/22/2012] [Indexed: 11/18/2022]
Abstract
The actin cytoskeleton is a dynamic structure necessary for cell and tissue organization, including the maintenance of epithelial barriers. Disruption of the epithelial barrier coincides with alterations of the actin cytoskeleton in several disease states. These disruptions primarily affect the paracellular space, which is normally regulated by tight junctions. Thereby, the actin cytoskeleton is a common and recurring target of bacterial virulence factors. In order to manipulate the actin cytoskeleton, bacteria secrete and inject toxins and effectors to hijack the host cell machinery, which interferes with host-cell pathways and with a number of actin binding proteins. An interesting model to study actin manipulation by bacterial effectors is Escherichia coli since due to its genome plasticity it has acquired diverse genetic mobile elements, which allow having different E. coli varieties in one bacterial species. These E. coli pathotypes, including intracellular and extracellular bacteria, interact with epithelial cells, and their interactions depend on a specific combination of virulence factors. In this paper we focus on E. coli effectors that mimic host cell proteins to manipulate the actin cytoskeleton. The study of bacterial effector-cytoskeleton interaction will contribute not only to the comprehension of the molecular causes of infectious diseases but also to increase our knowledge of cell biology.
Collapse
|
50
|
Duregotti E, Tedesco E, Montecucco C, Rigoni M. Calpains participate in nerve terminal degeneration induced by spider and snake presynaptic neurotoxins. Toxicon 2012; 64:20-8. [PMID: 23266309 DOI: 10.1016/j.toxicon.2012.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 11/13/2012] [Accepted: 12/13/2012] [Indexed: 11/19/2022]
Abstract
α-latrotoxin and snake presynaptic phospholipases A2 neurotoxins target the presynaptic membrane of axon terminals of the neuromuscular junction causing paralysis. These neurotoxins display different biochemical activities, but similarly alter the presynaptic membrane permeability causing Ca(2+) overload within the nerve terminals, which in turn induces nerve degeneration. Using different methods, here we show that the calcium-activated proteases calpains are involved in the cytoskeletal rearrangements that we have previously documented in neurons exposed to α-latrotoxin or to snake presynaptic phospholipases A2 neurotoxins. These results indicate that calpains, activated by the massive calcium influx from the extracellular medium, target fundamental components of neuronal cytoskeleton such as spectrin and neurofilaments, whose cleavage is functional to the ensuing nerve terminal fragmentation.
Collapse
Affiliation(s)
- Elisa Duregotti
- Department of Biomedical Sciences, CNR Institute of Neuroscience, University of Padova, Italy
| | | | | | | |
Collapse
|