1
|
Chukai Y, Furukawa N, Kosegawa O, Bai L, Sugano E, Fukuda T, Tomita H, Ozaki T. Mitochondrial calpain-1 truncates ATP synthase beta subunit. Biochem Biophys Res Commun 2025; 765:151829. [PMID: 40262470 DOI: 10.1016/j.bbrc.2025.151829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
Calpains cleave proteins in a calcium concentration-dependent manner, modulating their intracellular functions. Calpain-1, a member of the calpain family, is localized in the cytosol and mitochondria. Mitochondrial calpain-1 induces mitochondrial dysfunction and apoptosis by cleaving its substrate. Thus, identifying the substrate of calpain-1 is essential to understand its function. However, little is known about the substrates of mitochondrial calpain-1. To address this issue, we screened mitochondrial proteins using bioinformatics approaches and two-dimensional gel electrophoresis. We identified ATP5B as a potential substrate of mitochondrial calpain-1. Calpeptin, a pan-calpain inhibitor, and Tat-μCL, a mitochondrial calpain-1 specific inhibitor, prevented the truncation of ATP5B during in vitro Ca2+ incubation. Using recombinant human calpain-1 and ATP5B proteins, we demonstrated that calpain-1 directly cleaved ATP5B, generating a fragment of ATP5B. Based on the predicted cleavage sites in ATP5B, this cleavage may disrupt its interaction with ATP5A1, leading to mitochondrial dysfunction in ATP production. This study identified ATP5B as a novel substrate of mitochondrial calpain-1. The results provide new insights into mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yusaku Chukai
- Laboratory of Cell Biochemistry, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan; Laboratory of Cell Biochemistry, Department of Biological Science, Graduate School of Science and Engineering, Iwate University, Morioka, Iwate, Japan
| | - Nanami Furukawa
- Laboratory of Cell Biochemistry, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan; Laboratory of Cell Biochemistry, Department of Biological Science, Graduate School of Science and Engineering, Iwate University, Morioka, Iwate, Japan
| | - On Kosegawa
- Laboratory of Cell Biochemistry, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Lanlan Bai
- Laboratory of Cell Engineering and Molecular Genetics, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Eriko Sugano
- Laboratory of Visual Neuroscience, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Tomokazu Fukuda
- Laboratory of Cell Engineering and Molecular Genetics, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Hiroshi Tomita
- Laboratory of Visual Neuroscience, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Taku Ozaki
- Laboratory of Cell Biochemistry, Department of Life Sciences, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan; Laboratory of Cell Biochemistry, Department of Biological Science, Graduate School of Science and Engineering, Iwate University, Morioka, Iwate, Japan.
| |
Collapse
|
2
|
Sahagun DA, Lopuszynski JB, Feldman KS, Pogodzinski N, Zahid M. Toxicity Studies of Cardiac-Targeting Peptide Reveal a Robust Safety Profile. Pharmaceutics 2024; 16:73. [PMID: 38258084 PMCID: PMC10818749 DOI: 10.3390/pharmaceutics16010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
Targeted delivery of therapeutics specifically to cardiomyocytes would open up new frontiers for common conditions like heart failure. Our prior work using a phage display methodology identified a 12-amino-acid-long peptide that selectively targets cardiomyocytes after an intravenous injection in as little as 5 min and was hence termed a cardiac-targeting peptide (CTP: APHLSSQYSRT). CTP has been used to deliver imaging agents, small drug molecules, photosensitizing nanoparticles, exosomes, and even miRNA to cardiomyocytes. As a natural extension to the development of CTP as a clinically viable cardiac vector, we now present toxicity studies performed with the peptide. In vitro viability studies were performed in a human left ventricular myocyte cell line with 10 µM of Cyanine-5.5-labeled CTP (CTP-Cy5.5). In vitro ion channel profiles were completed for CTP followed by extensive studies in stably transfected cell lines for several GPCR-coupled receptors. Positive data for GPCR-coupled receptors were interrogated further with RT-qPCRs performed on mouse heart tissue. In vivo studies consisted of pre- and post-blood pressure monitoring acutely after a single CTP (10 mg/Kg) injection. Further in vivo toxicity studies consisted of injecting CTP (150 µg/Kg) in 60, 6-week-old, wild-type CD1, male/female mice (1:1), with cohorts of mice euthanized on days 0, 1, 2, 7, and 14 with inhalational CO2, followed by blood collection via cardiac puncture, complete blood count analysis, metabolic profiling, and finally, liver, renal, and thyroid studies. Lastly, mouse cardiac MRI was performed immediately before and after CTP (150 µg/Kg) injection to assess changes in cardiac size or function. Human left ventricular cardiomyocytes showed no decrease in viability after a 30 min incubation with CTP-Cy5.5. No significant activation or inhibition of any of seventy-eight protein channels was observed other than OPRM1 and COX2 at the highest tested concentration, neither of which were expressed in mouse heart tissue as assessed using RT-qPCR. CTP (10 mg/Kg) injections led to no change in blood pressure. Blood counts and chemistries showed no evidence of significant hematological, hepatic, or renal toxicities. Lastly, there was no difference in cardiac function, size, or mass acutely in response to CTP injections. Our studies with CTP showed no activation or inhibition of GPCR-associated receptors in vitro. We found no signals indicative of toxicity in vivo. Most importantly, cardiac functions remained unchanged acutely in response to CTP uptake. Further studies using good laboratory practices are needed with prolonged, chronic administration of CTP conjugated to a specific cargo of choice before human studies can be contemplated.
Collapse
Affiliation(s)
- Daniella A. Sahagun
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (D.A.S.); (J.B.L.)
| | - Jack B. Lopuszynski
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (D.A.S.); (J.B.L.)
| | - Kyle S. Feldman
- Clinical Virology Laboratory, Yale New Haven Hospital, New Haven, CT 06511, USA;
| | - Nicholas Pogodzinski
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Maliha Zahid
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (D.A.S.); (J.B.L.)
| |
Collapse
|
3
|
Ito G, Tatara Y, Itoh K, Yamada M, Yamashita T, Sakamoto K, Nozaki T, Ishida K, Wake Y, Kaneko T, Fukuda T, Sugano E, Tomita H, Ozaki T. Novel dicarbonyl metabolic pathway via mitochondrial ES1 possessing glyoxalase III activity. BBA ADVANCES 2023; 3:100092. [PMID: 37250100 PMCID: PMC10209487 DOI: 10.1016/j.bbadva.2023.100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023] Open
Abstract
Glycation, caused by reactive dicarbonyls, plays a role in various diseases by forming advanced glycation end products. In live cells, reactive dicarbonyls such as glyoxal (GO) and methylglyoxal (MGO) are produced during cell metabolism, and these should be removed consistently. However, the dicarbonyl metabolic system in the mitochondria remains unclear. It has been speculated that the mammalian mitochondrial protein ES1 is a homolog of bacterial elbB possessing glyoxalase III (GLO3) activity. Therefore, in this study, to investigate ES1 functions and GLO3 activity, we generated ES1-knockout (KO) mice and recombinant mouse ES1 protein and investigated the biochemical and histological analyses. In the mitochondrial fraction obtained from ES1-KO mouse brains, the GO metabolism and cytochrome c oxidase activity were significantly lower than those in the mitochondrial fraction obtained from wildtype (WT) mouse brains. However, the morphological features of the mitochondria did not change noticeably in the ES1-KO mouse brains compared with those in the WT mouse brains. The mitochondrial proteome analysis showed that the MGO degradation III pathway and oxidative phosphorylation-related proteins were increased. These should be the response to the reduced GO metabolism caused by ES1 deletion to compensate for the dicarbonyl metabolism and damaged cytochrome c oxidase by elevated GO. Recombinant mouse ES1 protein exhibited catalytic activity of converting GO to glycolic acid. These results indicate that ES1 possesses GLO3 activity and modulates the metabolism of GO in the mitochondria. To our knowledge, this is the first study to show a novel metabolic pathway for reactive dicarbonyls in mitochondria.
Collapse
Affiliation(s)
- Ginga Ito
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Yota Tatara
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifuchou, Hirosaki, Aomori 036-8562, Japan
| | - Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifuchou, Hirosaki, Aomori 036-8562, Japan
| | - Miwa Yamada
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Kimitoshi Sakamoto
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Aomori 036-8561, Japan
| | - Takayuki Nozaki
- Technical Support Center for Life Science Research, Iwate Medical University, 1-1-1 Idaidori, Yahaba, Iwate 028-3694, Japan
| | - Kinji Ishida
- Technical Support Center for Life Science Research, Iwate Medical University, 1-1-1 Idaidori, Yahaba, Iwate 028-3694, Japan
| | - Yui Wake
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Takehito Kaneko
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Tomokazu Fukuda
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Eriko Sugano
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Hiroshi Tomita
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Taku Ozaki
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| |
Collapse
|
4
|
Nhàn NTT, Maidana DE, Yamada KH. Ocular Delivery of Therapeutic Agents by Cell-Penetrating Peptides. Cells 2023; 12:1071. [PMID: 37048144 PMCID: PMC10093283 DOI: 10.3390/cells12071071] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Cell-penetrating peptides (CPPs) are short peptides with the ability to translocate through the cell membrane to facilitate their cellular uptake. CPPs can be used as drug-delivery systems for molecules that are difficult to uptake. Ocular drug delivery is challenging due to the structural and physiological complexity of the eye. CPPs may be tailored to overcome this challenge, facilitating cellular uptake and delivery to the targeted area. Retinal diseases occur at the posterior pole of the eye; thus, intravitreal injections are needed to deliver drugs at an effective concentration in situ. However, frequent injections have risks of causing vision-threatening complications. Recent investigations have focused on developing long-acting drugs and drug delivery systems to reduce the frequency of injections. In fact, conjugation with CPP could deliver FDA-approved drugs to the back of the eye, as seen by topical application in animal models. This review summarizes recent advances in CPPs, protein/peptide-based drugs for eye diseases, and the use of CPPs for drug delivery based on systematic searches in PubMed and clinical trials. We highlight targeted therapies and explore the potential of CPPs and peptide-based drugs for eye diseases.
Collapse
Affiliation(s)
- Nguyễn Thị Thanh Nhàn
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Daniel E. Maidana
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Kaori H. Yamada
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|
5
|
Calpains as mechanistic drivers and therapeutic targets for ocular disease. Trends Mol Med 2022; 28:644-661. [PMID: 35641420 PMCID: PMC9345745 DOI: 10.1016/j.molmed.2022.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022]
Abstract
Ophthalmic neurodegenerative diseases encompass a wide array of molecular pathologies unified by calpain dysregulation. Calpains are calcium-dependent proteases that perpetuate cellular death and inflammation when hyperactivated. Calpain inhibition trials in other organs have faced pharmacological challenges, but the eye offers many advantages for the development and testing of targeted molecular therapeutics, including small molecules, peptides, engineered proteins, drug implants, and gene-based therapies. This review highlights structural mechanisms underlying calpain activation, distinct cellular expression patterns, and in vivo models that link calpain hyperactivity to human retinal and developmental disease. Optimizing therapeutic approaches for calpain-mediated eye diseases can help accelerate clinically feasible strategies for treating calpain dysregulation in other diseased tissues.
Collapse
|
6
|
Aluja D, Delgado-Tomás S, Ruiz-Meana M, Barrabés JA, Inserte J. Calpains as Potential Therapeutic Targets for Myocardial Hypertrophy. Int J Mol Sci 2022; 23:ijms23084103. [PMID: 35456920 PMCID: PMC9032729 DOI: 10.3390/ijms23084103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/26/2022] [Accepted: 04/06/2022] [Indexed: 11/25/2022] Open
Abstract
Despite advances in its treatment, heart failure remains a major cause of morbidity and mortality, evidencing an urgent need for novel mechanism-based targets and strategies. Myocardial hypertrophy, caused by a wide variety of chronic stress stimuli, represents an independent risk factor for the development of heart failure, and its prevention constitutes a clinical objective. Recent studies performed in preclinical animal models support the contribution of the Ca2+-dependent cysteine proteases calpains in regulating the hypertrophic process and highlight the feasibility of their long-term inhibition as a pharmacological strategy. In this review, we discuss the existing evidence implicating calpains in the development of cardiac hypertrophy, as well as the latest advances in unraveling the underlying mechanisms. Finally, we provide an updated overview of calpain inhibitors that have been explored in preclinical models of cardiac hypertrophy and the progress made in developing new compounds that may serve for testing the efficacy of calpain inhibition in the treatment of pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- David Aluja
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
| | - Sara Delgado-Tomás
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José A. Barrabés
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-934894038
| |
Collapse
|
7
|
Oikawa T, Fukuda T, Yamashita T, Tomita H, Ozaki T. Lentiviral expression of calpain-1 C2-like domain peptide prevents glutamate-induced cell death in mouse hippocampal neuronal HT22 cells. In Vitro Cell Dev Biol Anim 2022; 58:289-294. [PMID: 35469046 DOI: 10.1007/s11626-022-00683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/30/2022] [Indexed: 11/05/2022]
Abstract
Glutamate neurotoxicity is involved in neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. Excess glutamate causes caspase-independent programmed cell death via oxidative stress and calcium influx. Our previous study showed that calpain-1 localizes to both the cytoplasm and mitochondria, where apoptosis-inducing factor (AIF) is cleaved by calpain-1 and translocates to the nucleus to induce DNA fragmentation. The autoinhibitory region of calpain-1 conjugated with the cell-penetrating peptide HIV1-Tat (namely Tat-μCL) specifically prevents the activity of mitochondrial calpain-1 and attenuates neuronal cell death in animal models of retinitis pigmentosa, as well as glutamate-induced cell death in mouse hippocampal HT22 cells. In the present study, we constructed a lentiviral vector expressing the Tat-μCL peptide and evaluated its protective effect against glutamate-induced cell death in HT22 cells. Lentiviral transduction with Tat-μCL significantly suppressed glutamate-induced nuclear translocation of AIF and DNA fragmentation. The findings of the present study suggest that the stable expression of Tat-μCL may be a potential gene therapy modality for neurodegenerative diseases.
Collapse
Affiliation(s)
- Takenori Oikawa
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 020-8550, Japan
| | - Tomokazu Fukuda
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 020-8550, Japan
| | - Hiroshi Tomita
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Taku Ozaki
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan.
| |
Collapse
|
8
|
Calpain-1 C2L domain peptide protects mouse hippocampus-derived neuronal HT22 cells against glutamate-induced oxytosis. Biochem Biophys Rep 2021; 27:101101. [PMID: 34430716 PMCID: PMC8374356 DOI: 10.1016/j.bbrep.2021.101101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 01/13/2023] Open
Abstract
Calpains are Ca2+-dependent cysteine proteases; their aberrant activation is associated with several neurodegenerative diseases. The μ-calpain catalytic subunit, calpain-1, is located in the cytoplasm as well as in the mitochondria. Mitochondrial calpain-1 cleaves apoptosis-inducing factor (AIF), leading to apoptotic cell death. We have previously reported that short peptides of calpain-1 C2-like domain conjugated with cell penetrating peptide HIV-Tat (Tat-μCL) selectively inhibit mitochondrial calpain-1 and effectively prevent neurodegenerative diseases of the eye. In this study, we determined whether mitochondrial calpain-1 mediates oxytosis (oxidative glutamate toxicity) in hippocampal HT22 cells using Tat-μCL and newly generated polyhistidine-conjugated μCL peptide and compared their efficacies in preventing oxytosis. TUNEL assay and single strand DNA staining revealed that both μCL peptides inhibited glutamate-induced oxytosis. Additionally, both the peptides suppressed the mitochondrial AIF translocation into the nucleus. All polyhistidine-μCL peptides (containing 4–16 histidine residues) showed higher cell permeability than Tat-μCL. Notably, tetrahistidine (H4)-μCL exerted the highest cytoprotective activity. Thus, H4-μCL may be a potential peptide drug for calpain-1-mediated neurodegenerative diseases such as Alzheimer's disease. Mitochondrial calpain-1 mediates glutamate-induced oxytosis in HT22 cells. CPP-μCL inhibits AIF translocation and DNA fragmentation in oxytosis. Polyhistidine conjugation enhances intracellular μCL peptide uptake efficiency.
Collapse
Key Words
- AIF, apoptosis-inducing factor
- BBB, blood–brain barrier
- CPP, cell penetrating peptide
- Cell penetrating peptide
- DAPI, 4′,6-diamidino-2-phenylindole
- DMEM, Dulbecco's Modified Eagle Medium
- Hippocampal HT22 cells
- Hn-μCL, polyhistidine-conjugated μCL
- MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium
- Mitochondrial calpain-1
- Neurodegeneration
- Oxytosis
- PBS, phosphate-buffered saline
- ssDNA, single stranded DNA
Collapse
|
9
|
Tang PH, Kinnick TR, Folk JC, Mahajan M, Bassuk AG, Tsang SH, Mahajan VB. PROGRESSION OF SCOTOPIC SINGLE-FLASH ELECTRORETINOGRAPHY IN THE STAGES OF CAPN5 VITREORETINOPATHY. Retin Cases Brief Rep 2021; 15:473-478. [PMID: 30300311 PMCID: PMC6453748 DOI: 10.1097/icb.0000000000000828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To characterize the changes found in the electroretinography (ERG) recordings of patients with autosomal dominant neovascular inflammatory vitreoretinopathy and correlate with clinical stages of the disease. METHODS Retrospective chart review. Bright- and dim-flash full-field scotopic, photopic, and 30-Hz flicker ERGs were obtained according to international standards. The scotopic ERGs were further processed to analyze the oscillatory potential. The patient described in the case report underwent full ERG testing; five patients composed the archival case series data and included scotopic ERG recordings. RESULTS Stage I autosomal dominant neovascular inflammatory vitreoretinopathy is characterized by a decrease in the b-wave amplitude on scotopic flash ERG and the disappearance of late OPs; however, the a-wave amplitude is normal. In Stage II, attenuation of early OPs and the c-wave are observed in scotopic ERG recordings, but both a- and b-wave amplitudes are unchanged. For patients in Stage III, there is a continued decline of both a- and b-wave amplitudes in scotopic ERG recordings. There was a loss of recordable scotopic ERG response in patients with Stage IV disease. CONCLUSION Electroretinography may be valuable in determining optimal timing for therapeutic intervention and response before loss of recordable retinal function in CAPN5 vitreoretinopathy.
Collapse
Affiliation(s)
- Peter H. Tang
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA
| | | | - James C. Folk
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA
| | - MaryAnn Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA
| | | | - Stephen H. Tsang
- College of Physicians & Surgeons, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Vinit B. Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA
- Palo Alto Veterans Administration, Palo Alto, CA
| |
Collapse
|
10
|
Wert KJ, Koch SF, Velez G, Hsu CW, Mahajan M, Bassuk AG, Tsang SH, Mahajan VB. CAPN5 genetic inactivation phenotype supports therapeutic inhibition trials. Hum Mutat 2019; 40:2377-2392. [PMID: 31403230 DOI: 10.1002/humu.23894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/20/2019] [Accepted: 08/09/2019] [Indexed: 01/08/2023]
Abstract
Small molecule pharmacological inhibition of dominant human genetic disease is a feasible treatment that does not rely on the development of individual, patient-specific gene therapy vectors. However, the consequences of protein inhibition as a clinical therapeutic are not well-studied. In advance of human therapeutic trials for CAPN5 vitreoretinopathy, genetic inactivation can be used to infer the effect of protein inhibition in vivo. We created a photoreceptor-specific knockout (KO) mouse for Capn5 and compared the retinal phenotype to both wild-type and an existing Capn5 KO mouse model. In humans, CAPN5 loss-of-function (LOF) gene variants were ascertained in large exome databases from 60,706 unrelated subjects without severe disease phenotypes. Ocular examination of the retina of Capn5 KO mice by histology and electroretinography showed no significant abnormalities. In humans, there were 22 LOF CAPN5 variants located throughout the gene and in all major protein domains. Structural modeling of coding variants showed these LOF variants were nearby known disease-causing variants within the proteolytic core and in regions of high homology between human CAPN5 and 150 homologs, yet the LOF of CAPN5 was tolerated as opposed to gain-of-function disease-causing variants. These results indicate that localized inhibition of CAPN5 is a viable strategy for hyperactivating disease alleles.
Collapse
Affiliation(s)
- Katherine J Wert
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California
| | - Susanne F Koch
- Department of Physiological Genomics, Biomedical Center, Ludwig Maximillian University, Munich, Germany
| | - Gabriel Velez
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California.,Department of Ophthalmology, Medical Scientist Training Program, University of Iowa, Iowa City, Iowa
| | - Chun-Wei Hsu
- Department of Ophthalmology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital, New York, New York.,Departments of Ophthalmology, Pathology, and Cell Biology, Jonas Children's Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia Stem Cell Initiative (CSCI), Columbia University, New York, New York
| | - MaryAnn Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California
| | | | - Stephen H Tsang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital, New York, New York.,Departments of Ophthalmology, Pathology, and Cell Biology, Jonas Children's Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia Stem Cell Initiative (CSCI), Columbia University, New York, New York
| | - Vinit B Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California.,Department of Ophthalmology, Veterans Affairs, Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
11
|
Iwamoto T, Ishiyama E, Ishida K, Yamashita T, Tomita H, Ozaki T. Presence of calpain-5 in mitochondria. Biochem Biophys Res Commun 2018; 504:454-459. [DOI: 10.1016/j.bbrc.2018.08.144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/24/2018] [Indexed: 01/10/2023]
|
12
|
Randazzo NM, Shanks ME, Clouston P, MacLaren RE. Two Novel CAPN5 Variants Associated with Mild and Severe Autosomal Dominant Neovascular Inflammatory Vitreoretinopathy Phenotypes. Ocul Immunol Inflamm 2017; 27:693-698. [PMID: 29040051 DOI: 10.1080/09273948.2017.1370651] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Purpose: We report two new CAPN5 mutations associated with a phenotype of Autosomal Dominant Neovascular Inflammatory Vitreoretinopathy. Methods: We performed next generation sequencing in two patients with ADNIV phenotype; the variants identified were explored further. Results: Patient 1 was heterozygous for CAPN5 c.799G>A, p.(Gly267Ser). Patient 2 was heterozygous for CAPN5 c.1126G>A, p.(Gly376Ser). Both amino acids are highly conserved across species. Patient 1 had a severe phenotype and his mutation lies within the protein's catalytic domain. Patient 2 had a mild phenotype and her mutation is the first ADNIV-causing mutation to be described in the regulatory domain of Calpain-5. Conclusions: Our findings potentially add two new ADNIV-causing CAPN5 mutations to the three previously described. We recommend CAPN5 genetic testing in all patients with a possible ADNIV phenotype, to develop our understanding of Calpain-5; a protein which could potentially provide therapeutically accessible targets for the treatment of many leading causes of blindness.
Collapse
Affiliation(s)
- Nadia M Randazzo
- a NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust , Oxford , United Kingdom.,b Oxford Eye Hospital, University of Oxford NHS Trust, John Radcliffe Hospital , Oxford , United Kingdom
| | - Morag E Shanks
- c Oxford Medical Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust Churchill Hospital , Oxford , United Kingdom
| | - Penny Clouston
- c Oxford Medical Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust Churchill Hospital , Oxford , United Kingdom
| | - Robert E MacLaren
- a NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust , Oxford , United Kingdom.,b Oxford Eye Hospital, University of Oxford NHS Trust, John Radcliffe Hospital , Oxford , United Kingdom
| |
Collapse
|
13
|
Nikaido Y, Furukawa T, Shimoyama S, Yamada J, Migita K, Koga K, Kushikata T, Hirota K, Kanematsu T, Hirata M, Ueno S. Propofol Anesthesia Is Reduced in Phospholipase C-Related Inactive Protein Type-1 Knockout Mice. J Pharmacol Exp Ther 2017; 361:367-374. [PMID: 28404686 DOI: 10.1124/jpet.116.239145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/04/2017] [Indexed: 11/22/2022] Open
Abstract
The GABA type A receptor (GABAA-R) is a major target of intravenous anesthetics. Phospholipase C-related inactive protein type-1 (PRIP-1) is important in GABAA-R phosphorylation and membrane trafficking. In this study, we investigated the role of PRIP-1 in general anesthetic action. The anesthetic effects of propofol, etomidate, and pentobarbital were evaluated in wild-type and PRIP-1 knockout (PRIP-1 KO) mice by measuring the latency and duration of loss of righting reflex (LORR) and loss of tail-pinch withdrawal response (LTWR). The effect of pretreatment with okadaic acid (OA), a protein phosphatase 1/2A inhibitor, on propofol- and etomidate-induced LORR was also examined. PRIP-1 deficiency provided the reduction of LORR and LTWR induced by propofol but not by etomidate or pentobarbital, indicating that PRIP-1 could determine the potency of the anesthetic action of propofol. Pretreatment with OA recovered the anesthetic potency induced by propofol in PRIP-1 KO mice. OA injection enhanced phosphorylation of cortical the GABAA-R β3 subunit in PRIP-1 KO mice. These results suggest that PRIP-1-mediated GABAA-R β3 subunit phosphorylation might be involved in the general anesthetic action induced by propofol but not by etomidate or pentobarbital.
Collapse
Affiliation(s)
- Yoshikazu Nikaido
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Tomonori Furukawa
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Shuji Shimoyama
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Junko Yamada
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Keisuke Migita
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Kohei Koga
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Tetsuya Kushikata
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Kazuyoshi Hirota
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Takashi Kanematsu
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Masato Hirata
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Shinya Ueno
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| |
Collapse
|
14
|
Ono Y, Saido TC, Sorimachi H. Calpain research for drug discovery: challenges and potential. Nat Rev Drug Discov 2016; 15:854-876. [PMID: 27833121 DOI: 10.1038/nrd.2016.212] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Calpains are a family of proteases that were scientifically recognized earlier than proteasomes and caspases, but remain enigmatic. However, they are known to participate in a multitude of physiological and pathological processes, performing 'limited proteolysis' whereby they do not destroy but rather modulate the functions of their substrates. Calpains are therefore referred to as 'modulator proteases'. Multidisciplinary research on calpains has begun to elucidate their involvement in pathophysiological mechanisms. Therapeutic strategies targeting malfunctions of calpains have been developed, driven primarily by improvements in the specificity and bioavailability of calpain inhibitors. Here, we review the calpain superfamily and calpain-related disorders, and discuss emerging calpain-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yasuko Ono
- Calpain Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science (IGAKUKEN), 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hiroyuki Sorimachi
- Calpain Project, Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science (IGAKUKEN), 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
15
|
Tabata K, Sugano E, Murakami F, Yamashita T, Ozaki T, Tomita H. Improved transduction efficiencies of adeno-associated virus vectors by synthetic cell-permeable peptides. Biochem Biophys Res Commun 2016; 478:1732-8. [PMID: 27614311 DOI: 10.1016/j.bbrc.2016.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/02/2016] [Indexed: 12/01/2022]
Abstract
Various serotypes of adeno-associated virus (AAV) vectors have been used for gene therapy and as research tools. Among these serotypes, the AAV type 2 vector has been used successfully in human gene therapies. However, the transduction efficiency of AAV2 depends on the cell type, and this poses a problem in the efficacy of gene therapy. To improve the transduction efficiency of AAV2, we designed a small peptide consisting of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor peptide and the HIV-Tat sequence Tat-Y1068. Pre- or co-treatment of CYNOM-K1 cells from cynomolgus monkey embryo skin with Tat-Y1068 increased the transduction efficiencies in a dose-dependent manner and caused p38 phosphorylation. The transduction efficiency of AAV2 into the rat fibroblast cell line RAT-1 highly expressing EGFR was less than the transduction efficiency of AAV2 into CYNOM-K1 cells. Tat-Y1068 increased the transduction efficiency in RAT-1 cells in the same manner as in CYNOM-K1 cells. In conclusion, cell-permeable peptides possessing the EGFR tyrosine kinase inhibitor function might serve as a useful ingredient of AAV2 vector solution for increasing the transduction efficiency of gene therapies.
Collapse
Affiliation(s)
- Kitako Tabata
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan.
| | - Eriko Sugano
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan; Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan.
| | - Fumika Murakami
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan.
| | - Tetsuro Yamashita
- Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan; Department of Biological Chemistry, Iwate University Faculty of Agriculture, Morioka, Japan.
| | - Taku Ozaki
- Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan.
| | - Hiroshi Tomita
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan; Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan; Clinical Research, Innovation and Education Center, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai, Miyagi 980-8574, Japan.
| |
Collapse
|
16
|
Ozaki T, Yamashita T, Tomita H, Sugano E, Ishiguro SI. The protection of rat retinal ganglion cells from ischemia/reperfusion injury by the inhibitory peptide of mitochondrial μ-calpain. Biochem Biophys Res Commun 2016; 478:1700-5. [PMID: 27596965 DOI: 10.1016/j.bbrc.2016.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
Abstract
Intracellular Ca(2+)-dependent cysteine proteases such as calpains have been suggested as critical factors in retinal ganglion cell (RGC) death. However, it is unknown whether mitochondrial calpains are involved in RGC death. The purpose of the present study was to determine whether the inhibition of mitochondrial μ-calpain activity protects against RGC death during ischemia/reperfusion (I/R) injury. This study used a well-established rat model of experimental acute glaucoma involving I/R injury. A specific peptide inhibitor of mitochondrial μ-calpain, Tat-μCL, was topically applied to rats via eye drops three times a day for 5 days after I/R. RGC death was determined by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. The truncation of apoptosis-inducing factor (AIF) was determined by western blot analyses. Retinal morphology was determined after staining with hematoxyline and eosin. In addition, the number of Fluoro Gold-labeled RGCs in flat-mounted retinas was used to determine the percentage of surviving RGCs after I/R injury. After 1 day of I/R, RGC death was observed in the ganglion cell layer. Treatment with Tat-μCL eye drops significantly prevented the death of RGCs and the truncation of AIF. After 5 days of I/R, RGC death decreased by approximately 40%. However, Tat-μCL significantly inhibited the decrease in the retinal sections and flat-mounted retinas. The results suggested that mitochondrial μ-calpain is associated with RGC death during I/R injury via truncation of AIF. In addition, the inhibition of mitochondrial μ-calpain activity by Tat-μCL had a neuroprotective effect against I/R-induced RGC death.
Collapse
Affiliation(s)
- Taku Ozaki
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, Morioka, Japan; Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan.
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Hiroshi Tomita
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, Morioka, Japan
| | - Eriko Sugano
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, Morioka, Japan
| | - Sei-Ichi Ishiguro
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan; Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
17
|
Shinde V, Kotla P, Strang C, Gorbatyuk M. Unfolded protein response-induced dysregulation of calcium homeostasis promotes retinal degeneration in rat models of autosomal dominant retinitis pigmentosa. Cell Death Dis 2016; 7:e2085. [PMID: 26844699 PMCID: PMC4670931 DOI: 10.1038/cddis.2015.325] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/15/2015] [Accepted: 09/30/2015] [Indexed: 12/12/2022]
Abstract
The molecular mechanism of autosomal dominant retinitis pigmentosa (ADRP) in rats is closely associated with a persistently activated unfolded protein response (UPR). If unchecked, the UPR might trigger apoptosis, leading to photoreceptor death. One of the UPR-activated cellular signaling culminating in apoptotic photoreceptor cell death is linked to an increase in intracellular Ca(2+). Therefore, we validated whether ADRP retinas experience a cytosolic Ca(2+) overload, and whether sustained UPR in the wild-type retina could promote retinal degeneration through Ca(2+)-mediated calpain activation. We performed an ex vivo experiment to measure intracellular Ca(2+) in ADRP retinas as well as to detect the expression levels of proteins that act as Ca(2+) sensors. In separate experiments with the subretinal injection of tunicamycin (UPR inducer) and a mixture of calcium ionophore (A231278) and thapsigargin (SERCA2b inhibitor) we assessed the consequences of a sustained UPR activation and increased intracellular Ca(2+) in the wild-type retina, respectively, by performing scotopic ERG, histological, and western blot analyses. Results of the study revealed that induced UPR in the retina activates calpain-mediated signaling, and increased intracellular Ca(2+) is capable of promoting retinal degeneration. A significant decline in ERG amplitudes at 6 weeks post treatment was associated with photoreceptor cell loss that occurred through calpain-activated CDK5-pJNK-Csp3/7 pathway. Similar calpain activation was found in ADRP rat retinas. A twofold increase in intracellular Ca(2+) and up- and downregulations of ER membrane-associated Ca(2+)-regulated IP3R channels and SERCA2b transporters were detected. Therefore, sustained UPR activation in the ADRP rat retinas could promote retinal degeneration through increased intracellular Ca(2+) and calpain-mediated apoptosis.
Collapse
Affiliation(s)
- V Shinde
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - P Kotla
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C Strang
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M Gorbatyuk
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
18
|
Furukawa A, Koriyama Y. A role of Heat Shock Protein 70 in Photoreceptor Cell Death: Potential as a Novel Therapeutic Target in Retinal Degeneration. CNS Neurosci Ther 2015; 22:7-14. [PMID: 26507240 DOI: 10.1111/cns.12471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 01/17/2023] Open
Abstract
Retinal degenerative diseases (RDs) such as retinitis pigmentosa (RP) are a genetically heterogeneous group of disorders characterized by night blindness and peripheral vision loss, which caused by the dysfunction and death of photoreceptor cells. Although many causative gene mutations have been reported, the final common end stage is photoreceptor cell death. Unfortunately, no effective treatments or therapeutic agents have been discovered. Heat shock protein 70 (HSP70) is highly conserved and has antiapoptotic activities. A few reports have shown that HSP70 plays a role in RDs. Thus, we focused on the role of HSP70 in photoreceptor cell death. Using the N-methyl-N-nitrosourea (MNU)-induced photoreceptor cell death model in mice, we could examine two stages of the novel cell death mechanism; the early stage, including HSP70 cleavage through protein carbonylation by production of reactive oxygen species, lipid peroxidation and Ca(2+) influx/calpain activation, and the late stage of cathepsin and/or caspase activation. The upregulation of intact HSP70 expression by its inducer is likely to protect photoreceptor cells. In this review, we focus on the role of HSP70 and the novel cell death signaling process in RDs. We also describe candidate therapeutic agents for RDs.
Collapse
Affiliation(s)
- Ayako Furukawa
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| | - Yoshiki Koriyama
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| |
Collapse
|
19
|
Fernández-Sánchez L, Lax P, Noailles A, Angulo A, Maneu V, Cuenca N. Natural Compounds from Saffron and Bear Bile Prevent Vision Loss and Retinal Degeneration. Molecules 2015; 20:13875-93. [PMID: 26263962 PMCID: PMC6332441 DOI: 10.3390/molecules200813875] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/23/2015] [Accepted: 07/28/2015] [Indexed: 12/13/2022] Open
Abstract
All retinal disorders, regardless of their aetiology, involve the activation of oxidative stress and apoptosis pathways. The administration of neuroprotective factors is crucial in all phases of the pathology, even when vision has been completely lost. The retina is one of the most susceptible tissues to reactive oxygen species damage. On the other hand, proper development and functioning of the retina requires a precise balance between the processes of proliferation, differentiation and programmed cell death. The life-or-death decision seems to be the result of a complex balance between pro- and anti-apoptotic signals. It has been recently shown the efficacy of natural products to slow retinal degenerative process through different pathways. In this review, we assess the neuroprotective effect of two compounds used in the ancient pharmacopoeia. On one hand, it has been demonstrated that administration of the saffron constituent safranal to P23H rats, an animal model of retinitis pigmentosa, preserves photoreceptor morphology and number, the capillary network and the visual response. On the other hand, it has been shown that systemic administration of tauroursodeoxycholic acid (TUDCA), the major component of bear bile, to P23H rats preserves cone and rod structure and function, together with their contact with postsynaptic neurons. The neuroprotective effects of safranal and TUDCA make these compounds potentially useful for therapeutic applications in retinal degenerative diseases.
Collapse
Affiliation(s)
- Laura Fernández-Sánchez
- Departament of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain.
| | - Pedro Lax
- Departament of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain.
| | - Agustina Noailles
- Departament of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain.
| | - Antonia Angulo
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain.
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain.
| | - Nicolás Cuenca
- Departament of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain.
| |
Collapse
|
20
|
Ozaki T, Nakazawa M, Yamashita T, Ishiguro SI. Delivery of Topically Applied Calpain Inhibitory Peptide to the Posterior Segment of the Rat Eye. PLoS One 2015; 10:e0130986. [PMID: 26107400 PMCID: PMC4479448 DOI: 10.1371/journal.pone.0130986] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/27/2015] [Indexed: 11/19/2022] Open
Abstract
We developed an inhibitory peptide that specifically acts against mitochondrial μ-calpain (Tat-μCL, 23 amino acid, 2857.37 Da) and protects photoreceptors in retinal dystrophic rats. In the present study, we topically administered Tat-μCL to the eyes of Sprague-Dawley rats for 7 days to determine both the delivery route of the peptide to the posterior segment of the eye and the kinetics after topical application in adult rats. Distribution of the peptide was determined by immunohistochemical analysis, and enzyme-linked immune-absorbent assay was used to quantify the accumulation in the retina. Peptides were prominently detected in both the anterior and posterior segments of the eye at 1 h after the final eye drop application. Immunohistochemically positive reactions were observed in the retina, optic nerve, choroid, sclera and the retrobulbar tissues, even in the posterior portion of the eye. Immunoactivities gradually diminished at 3 and 6 h after the final eye drop. Quantitative estimations of the amount of peptide in the retina were 15.3, 5.8 and 1.0 pg/μg protein at 1, 3 and 6 h after the final instillation, respectively. Current results suggest that while the topically applied Tat-μCL peptide reaches the posterior segment of the retina and the optic nerve, the sufficient concentration (> IC50) is maintained for at least 6 h in the rat retina. Our findings suggest that delivery of topically applied peptide to the posterior segment and optic nerve occurs through the conjunctiva, periocular connective tissue, sclera and optic nerve sheath.
Collapse
Affiliation(s)
- Taku Ozaki
- Department of Ophthalmology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Mitsuru Nakazawa
- Department of Ophthalmology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
- * E-mail:
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Iwate University Faculty of Agriculture, Morioka, Japan
| | - Sei-ichi Ishiguro
- Department of Biochemistry and Molecular Biology, Hirosaki University Faculty of Agriculture and Life Science, Hirosaki, Japan
| |
Collapse
|
21
|
Wert KJ, Bassuk AG, Wu WH, Gakhar L, Coglan D, Mahajan M, Wu S, Yang J, Lin CS, Tsang SH, Mahajan VB. CAPN5 mutation in hereditary uveitis: the R243L mutation increases calpain catalytic activity and triggers intraocular inflammation in a mouse model. Hum Mol Genet 2015; 24:4584-98. [PMID: 25994508 DOI: 10.1093/hmg/ddv189] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/18/2015] [Indexed: 12/21/2022] Open
Abstract
A single amino acid mutation near the active site of the CAPN5 protease was linked to the inherited blinding disorder, autosomal dominant neovascular inflammatory vitreoretinopathy (ADNIV, OMIM #193235). In homology modeling with other calpains, this R243L CAPN5 mutation was situated in a mobile loop that gates substrate access to the calcium-regulated active site. In in vitro activity assays, the mutation increased calpain protease activity and made it far more active at low concentrations of calcium. To test whether the disease allele could yield an animal model of ADNIV, we created transgenic mice expressing human (h) CAPN5(R243L) only in the retina. The resulting hCAPN5(R243L) transgenic mice developed a phenotype consistent with human uveitis and ADNIV, at the clinical, histological and molecular levels. The fundus of hCAPN5(R243L) mice showed enhanced autofluorescence (AF) and pigment changes indicative of reactive retinal pigment epithelial cells and photoreceptor degeneration. Electroretinography showed mutant mouse eyes had a selective loss of the b-wave indicating an inner-retina signaling defect. Histological analysis of mutant mouse eyes showed protein extravasation from dilated vessels into the anterior chamber and vitreous, vitreous inflammation, vitreous and retinal fibrosis and retinal degeneration. Analysis of gene expression changes in the hCAPN5(R243L) mouse retina showed upregulation of several markers, including members of the Toll-like receptor pathway, chemokines and cytokines, indicative of both an innate and adaptive immune response. Since many forms of uveitis share phenotypic characteristics of ADNIV, this mouse offers a model with therapeutic testing utility for ADNIV and uveitis patients.
Collapse
Affiliation(s)
- Katherine J Wert
- Barbara and Donald Jonas Laboratory of Stem Cells and Regenerative Medicine and Bernard and Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, Institute of Human Nutrition, College of Physicians and Surgeons
| | | | - Wen-Hsuan Wu
- Barbara and Donald Jonas Laboratory of Stem Cells and Regenerative Medicine and Bernard and Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute
| | - Lokesh Gakhar
- Department of Biochemistry, Protein Crystallography Facility
| | - Diana Coglan
- Omics Laboratory and Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - MaryAnn Mahajan
- Omics Laboratory and Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Shu Wu
- Department of Pediatrics and Neurology
| | - Jing Yang
- Protein Crystallography Facility, Omics Laboratory and
| | | | - Stephen H Tsang
- Barbara and Donald Jonas Laboratory of Stem Cells and Regenerative Medicine and Bernard and Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, Institute of Human Nutrition, College of Physicians and Surgeons, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA,
| | - Vinit B Mahajan
- Omics Laboratory and Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
22
|
Abstract
INTRODUCTION Calpain is a family of cysteine proteases found in eukaryotes and a few bacteria. There is considerable interest in the search for calpain inhibitors because the enzyme has been implicated in several diseases including ocular disorders, neurodegenerative disorders, metabolic disorders and cancer. AREAS COVERED An overview of calpain inhibitors disclosed between 2012 and 2014 is presented. Among these are epoxysuccinates, dipeptide imaging agents, macrocyclic inhibitors, α-helical peptidomimetic inhibitors, carboxamides, 5-azolones and α-mercaptoacrylates. Additionally, preclinical studies of calpain inhibitors in pathologies such blood disorders, ocular disorders, neurological disorders and muscle disorders are discussed. EXPERT OPINION Major advances made in calpain inhibitor research between 2012 and 2014 include: i) the discovery of cytosolic-stable carboxamide calpain inhibitors; ii) synthesis of epoxysuccinates with excellent bioavailability; iii) disclosure of the X-ray crystal structures of novel α-mercaptoacrylates bound to the pentaEF hand region from human calpain; and iv) disclosure of calpain inhibitors as anti-sickling agents. Several calpain inhibitors were reported but limited effort was directed towards the discovery of calpain isoform selective agents, which continues to dampen the therapeutic potential of calpain inhibitors.
Collapse
Affiliation(s)
- Isaac O Donkor
- The University of Tennessee Health Science Center , Memphis, TN , USA
| |
Collapse
|
23
|
Cuenca N, Fernández-Sánchez L, Campello L, Maneu V, De la Villa P, Lax P, Pinilla I. Cellular responses following retinal injuries and therapeutic approaches for neurodegenerative diseases. Prog Retin Eye Res 2014; 43:17-75. [PMID: 25038518 DOI: 10.1016/j.preteyeres.2014.07.001] [Citation(s) in RCA: 313] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/03/2014] [Accepted: 07/07/2014] [Indexed: 01/17/2023]
Abstract
Retinal neurodegenerative diseases like age-related macular degeneration, glaucoma, diabetic retinopathy and retinitis pigmentosa each have a different etiology and pathogenesis. However, at the cellular and molecular level, the response to retinal injury is similar in all of them, and results in morphological and functional impairment of retinal cells. This retinal degeneration may be triggered by gene defects, increased intraocular pressure, high levels of blood glucose, other types of stress or aging, but they all frequently induce a set of cell signals that lead to well-established and similar morphological and functional changes, including controlled cell death and retinal remodeling. Interestingly, an inflammatory response, oxidative stress and activation of apoptotic pathways are common features in all these diseases. Furthermore, it is important to note the relevant role of glial cells, including astrocytes, Müller cells and microglia, because their response to injury is decisive for maintaining the health of the retina or its degeneration. Several therapeutic approaches have been developed to preserve retinal function or restore eyesight in pathological conditions. In this context, neuroprotective compounds, gene therapy, cell transplantation or artificial devices should be applied at the appropriate stage of retinal degeneration to obtain successful results. This review provides an overview of the common and distinctive features of retinal neurodegenerative diseases, including the molecular, anatomical and functional changes caused by the cellular response to damage, in order to establish appropriate treatments for these pathologies.
Collapse
Affiliation(s)
- Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain; Multidisciplinary Institute for Environmental Studies "Ramon Margalef", University of Alicante, Alicante, Spain.
| | - Laura Fernández-Sánchez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Pedro De la Villa
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Isabel Pinilla
- Department of Ophthalmology, Lozano Blesa University Hospital, Aragon Institute of Health Sciences, Zaragoza, Spain
| |
Collapse
|
24
|
Ozaki T, Nakazawa M, Kudo T, Hirano S, Suzuki K, Ishiguro SI. Protection of Cone Photoreceptor M-Opsin Degradation with 9-Cis-β-Carotene-Rich AlgaDunaliella bardawilinRpe65−/−Mouse Retinal Explant Culture. Curr Eye Res 2014; 39:1221-31. [DOI: 10.3109/02713683.2014.907430] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
25
|
Wert KJ, Skeie JM, Bassuk AG, Olivier AK, Tsang SH, Mahajan VB. Functional validation of a human CAPN5 exome variant by lentiviral transduction into mouse retina. Hum Mol Genet 2013; 23:2665-77. [PMID: 24381307 DOI: 10.1093/hmg/ddt661] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Exome sequencing indicated that the gene encoding the calpain-5 protease, CAPN5, is the likely cause of retinal degeneration and autoimmune uveitis in human patients with autosomal dominant neovascular inflammatory vitreoretinopathy (ADNIV, OMIM #193235). To explore the mechanism of ADNIV, a human CAPN5 disease allele was expressed in mouse retinas with a lentiviral vector created to express either the wild-type human (h) CAPN5 or the ADNIV mutant hCAPN5-R243L allele under a rhodopsin promoter with tandem green fluorescent protein (GFP) expression. Vectors were injected into the subretinal space of perinatal mice. Mouse phenotypes were analyzed using electroretinography, histology and inflammatory gene expression profiling. Mouse calpain-5 showed high homology to its human ortholog with >98% sequence identity that includes the ADNIV mutant residue. Calpain-5 protein was expressed in the inner and outer segments of the photoreceptors and in the outer plexiform layer. Expression of the hCAPN5-R243L allele caused loss of the electroretinogram b-wave, photoreceptor degeneration and induction of immune cell infiltration and inflammatory genes in the retina, recapitulating major features of the ADNIV phenotype. Intraocular neovascularization and fibrosis were not observed during the study period. Our study shows that expression of the hCAPN5-R243L disease allele elicits an ADNIV-like disease in mice. It further suggests that ADNIV is due to CAPN5 gain-of-function rather than haploinsufficiency, and retinal expression may be sufficient to generate an autoimmune response. Genetic models of ADNIV in the mouse can be used to explore protease mechanisms in retinal degeneration and inflammation as well as preclinical therapeutic testing.
Collapse
Affiliation(s)
- Katherine J Wert
- Bernard and Shirlee Brown Glaucoma Laboratory, Department of Pathology and Cell Biology, College of Physicians and Surgeons
| | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Murakami Y, Notomi S, Hisatomi T, Nakazawa T, Ishibashi T, Miller JW, Vavvas DG. Photoreceptor cell death and rescue in retinal detachment and degenerations. Prog Retin Eye Res 2013; 37:114-40. [PMID: 23994436 DOI: 10.1016/j.preteyeres.2013.08.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/08/2013] [Accepted: 08/10/2013] [Indexed: 02/08/2023]
Abstract
Photoreceptor cell death is the ultimate cause of vision loss in various retinal disorders, including retinal detachment (RD). Photoreceptor cell death has been thought to occur mainly through apoptosis, which is the most characterized form of programmed cell death. The caspase family of cysteine proteases plays a central role for inducing apoptosis, and in experimental models of RD, dying photoreceptor cells exhibit caspase activation; however, there is a paradox that caspase inhibition alone does not provide a sufficient protection against photoreceptor cell loss, suggesting that other mechanisms of cell death are involved. Recent accumulating evidence demonstrates that non-apoptotic forms of cell death, such as autophagy and necrosis, are also regulated by specific molecular machinery, such as those mediated by autophagy-related proteins and receptor-interacting protein kinases, respectively. Here we summarize the current knowledge of cell death signaling and its roles in photoreceptor cell death after RD and other retinal degenerative diseases. A body of studies indicate that not only apoptotic but also autophagic and necrotic signaling are involved in photoreceptor cell death, and that combined targeting of these pathways may be an effective neuroprotective strategy for retinal diseases associated with photoreceptor cell loss.
Collapse
Affiliation(s)
- Yusuke Murakami
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Ozaki T, Nakazawa M, Yamashita T, Tomita H, Ebina Y, Ishiguro SI. Decrease of ATP by mitochondrial m-calpain inhibitory peptide in the rat retinas. Cell Struct Funct 2013; 38:207-23. [PMID: 23965546 DOI: 10.1247/csf.13008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Activations of mitochondrial calpains cause apoptosis-inducing factor-dependent apoptosis of retinal photoreceptor cells in the Royal College of Surgeons (RCS) rat, an animal model of retinitis pigmentosa. In the present study, we attempted to develop specific inhibitors of mitochondrial calpains that would prevent the retinal degeneration. We examined the inhibitory potency of 20-mer peptides of the m-calpain for mitochondrial calpains activity, determined the inhibitory regions, and conjugated the cell-penetrating peptides (CPP). The cytotoxicity and delivery of the peptide was evaluated using mouse photoreceptor-derived 661W cells. After intravitreal injection of the peptide in RCS rats, we examined the peptide delivery to the retina, photoreceptor cell death numbers, responses of the electroretinogram (ERG), concentrations of intracellular ATP, and changes of retinal morphology. Results showed that one of the peptides inhibited the activity of the mitochondrial m-calpain. The HIV-1 tat-conjugated m-calpain peptide, HIV-Nm, could preserve the inhibitory potency of the mitochondrial m-calpain, and penetrate into the 661W cells. While intravitreal injection of HIV-Nm made it possible to deliver to the retina, it did not prevent photoreceptor cell death. Furthermore, it caused the ERG attenuation and the decrease in the intracellular ATP only a day after the injection. Although HIV-Nm did not cause histological change of the retina after 1 or 2 days of the administration, the morphological abnormality of the retina was observed after 3-14 days. Our results demonstrated that HIV-Nm failed to prevent the photoreceptor cell death, but rather caused the attenuation of ERG response and the decrease of ATP.
Collapse
Affiliation(s)
- Taku Ozaki
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University
| | | | | | | | | | | |
Collapse
|
29
|
Ozaki T, Ishiguro SI, Hirano S, Baba A, Yamashita T, Tomita H, Nakazawa M. Inhibitory peptide of mitochondrial μ-calpain protects against photoreceptor degeneration in rhodopsin transgenic S334ter and P23H rats. PLoS One 2013; 8:e71650. [PMID: 23951212 PMCID: PMC3739725 DOI: 10.1371/journal.pone.0071650] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/01/2013] [Indexed: 11/23/2022] Open
Abstract
Mitochondrial μ-calpain and apoptosis-inducing factor (AIF)-dependent photoreceptor cell death has been seen in several rat and mouse models of retinitis pigmentosa (RP). Previously, we demonstrated that the specific peptide inhibitor of mitochondrial μ-calpain, Tat-µCL, protected against retinal degeneration following intravitreal injection or topical eye-drop application in Mertk gene-mutated Royal College of Surgeons rats, one of the animal models of RP. Because of the high rate of rhodopsin mutations in RP patients, the present study was performed to confirm the protective effects of Tat-µCL against retinal degeneration in rhodopsin transgenic S334ter and P23H rats. We examined the effects of intravitreal injection or topical application of the peptide on retinal degeneration in S334ter and P23H rats by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay, electroretinogram (ERG), immunohistochemistry for AIF, and histological staining. In S334ter rats, we found that intravitreal injection or topical application of the peptide prevented photoreceptor cell death from postnatal (PN) 15 to 18 days, the time of early-stage retinal degeneration. Topical application of the peptide also delayed attenuation of ERG responses from PN 28 to 56 days. In P23H rats, topical application of the peptide protected against photoreceptor cell death and nuclear translocation of AIF on PN 30, 40, and 50 days, as the primary stages of degeneration. We observed that topical application of the peptide inhibited the thinning of the outer nuclear layer and delayed ERG attenuations from PN 30 to 90 days. Our results demonstrate that the mitochondrial μ-calpain and AIF pathway is involved in early-stage retinal degeneration in rhodopsin transgenic S334ter and P23H rats, and inhibition of this pathway shows curative potential for rhodopsin mutation-caused RP.
Collapse
Affiliation(s)
- Taku Ozaki
- Department of Ophthalmology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
- Department of Biochemistry and Molecular Biology, Hirosaki University Faculty of Agriculture and Life Science, Hirosaki, Japan
| | - Sei-ichi Ishiguro
- Department of Biochemistry and Molecular Biology, Hirosaki University Faculty of Agriculture and Life Science, Hirosaki, Japan
| | - Satoshi Hirano
- Department of Biochemistry and Molecular Biology, Hirosaki University Faculty of Agriculture and Life Science, Hirosaki, Japan
| | - Ayaka Baba
- Department of Biochemistry and Molecular Biology, Hirosaki University Faculty of Agriculture and Life Science, Hirosaki, Japan
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Iwate University Faculty of Agriculture, Morioka, Japan
| | - Hiroshi Tomita
- Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Japan
| | - Mitsuru Nakazawa
- Department of Ophthalmology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
30
|
Cisplatin binding and inactivation of mitochondrial glutamate oxaloacetate transaminase in cisplatin-induced rat nephrotoxicity. Biosci Biotechnol Biochem 2013; 77:1645-9. [PMID: 23924727 DOI: 10.1271/bbb.130172] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cisplatin is a widely used chemotherapeutic agent, but its use is limited by nephrotoxicity associated with mitochondrial dysfunction. Because its mechanisms are poorly understood, we aimed to identify the mitochondrial proteins targeted by cisplatin. We isolated renal mitochondrial proteins from Sprague-Dawley (SD) rats and performed cisplatin-affinity column chromatography. The proteins eluted were detected on SDS-PAGE and subjected to in-gel tryptic digestion and LC-MS/MS analysis. We identified glutamate oxaloacetate transaminase (GOT) and mitochondrial malate dehydrogenase (MDH). Next, we administered cisplatin intraperitoneally to SD rats to induce nephrotoxicity and assayed the activities of the enzymes. The results indicated that cisplatin caused a severe decrease in mitochondrial GOT activity on day 1 after cisplatin administration. Three d later, we also identified a decrease in mitochondrial MDH activity. Our results indicate that cisplatin binds to mitochondrial GOT and inhibits its activity, causing mitochondrial dysfunction and subsequent nephrotoxicity.
Collapse
|