1
|
Policastro PF, Schneider CA, Winkler CW, Leung JM, Peterson KE. Retinoic acid-induced differentiation and oxidative stress inhibitors increase resistance of human neuroblastoma cells to La Crosse virus-induced cell death. J Virol 2024:e0030024. [PMID: 39382324 DOI: 10.1128/jvi.00300-24] [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: 06/24/2024] [Accepted: 08/13/2024] [Indexed: 10/10/2024] Open
Abstract
La Crosse Virus (LACV) encephalitis patients are at risk for long-term deficits in cognitive function due to neuronal apoptosis following virus infection. However, the specific etiology underlying neuronal damage remains elusive. In this study, we examined how differentiation and mitotic inhibition of neuroblastoma cells influence their susceptibility to LACV infection and cell death. Treatment of SH-SY5Y cells with retinoic acid induced a neuronal cell phenotype which was similarly susceptible to LACV infection as untreated cells but had significantly delayed virus-induced cell death. Protein and RNA transcript analysis showed that retinoic acid-treated cells had decreased oxidative stress responses to LACV infection compared to untreated cells. Modulation of oxidative stress in untreated cells with specific compounds also delayed cell death, without substantially impacting virus production. Thus, the oxidative stress response of neurons to virus infection may be a key component of neuronal susceptibility to virus-induced cell death. IMPORTANCE Encephalitic viruses like La Crosse Virus (LACV) infect and kill neurons. Disease onset and progression is rapid meaning the time frame to treat patients before significant and long-lasting damage occurs is limited. Examining how neurons, the primary cells infected by LACV in the brain, resist virus-induced cell death can provide avenues for determining which pathways to target for effective treatments. In the current study, we studied how changing neuroblastoma growth and metabolism with retinoic acid treatment impacted their susceptibility to LACV-induced cell death. We utilized this information to test compounds for preventing death in these cells.
Collapse
Affiliation(s)
- Paul F Policastro
- Neuroimmunology Section, Laboratory of Neurological Infections and Immunity, Hamilton, Montana, USA
| | - Christine A Schneider
- Neuroimmunology Section, Laboratory of Neurological Infections and Immunity, Hamilton, Montana, USA
- Electron Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Hamilton, Montana, USA
| | - Clayton W Winkler
- Neuroimmunology Section, Laboratory of Neurological Infections and Immunity, Hamilton, Montana, USA
| | - Jacqueline M Leung
- Electron Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Hamilton, Montana, USA
| | - Karin E Peterson
- Neuroimmunology Section, Laboratory of Neurological Infections and Immunity, Hamilton, Montana, USA
| |
Collapse
|
2
|
Skobeleva K, Wang G, Kaznacheyeva E. STIM Proteins: The Gas and Brake of Calcium Entry in Neurons. Neurosci Bull 2024:10.1007/s12264-024-01272-5. [PMID: 39266936 DOI: 10.1007/s12264-024-01272-5] [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: 01/22/2024] [Accepted: 04/22/2024] [Indexed: 09/14/2024] Open
Abstract
Stromal interaction molecules (STIM)s are Ca2+ sensors in internal Ca2+ stores of the endoplasmic reticulum. They activate the store-operated Ca2+ channels, which are the main source of Ca2+ entry in non-excitable cells. Moreover, STIM proteins interact with other Ca2+ channel subunits and active transporters, making STIMs an important intermediate molecule in orchestrating a wide variety of Ca2+ influxes into excitable cells. Nevertheless, little is known about the role of STIM proteins in brain functioning. Being involved in many signaling pathways, STIMs replenish internal Ca2+ stores in neurons and mediate synaptic transmission and neuronal excitability. Ca2+ dyshomeostasis is a signature of many pathological conditions of the brain, including neurodegenerative diseases, injuries, stroke, and epilepsy. STIMs play a role in these disturbances not only by supporting abnormal store-operated Ca2+ entry but also by regulating Ca2+ influx through other channels. Here, we review the present knowledge of STIMs in neurons and their involvement in brain pathology.
Collapse
Affiliation(s)
- Ksenia Skobeleva
- Laboratory of Ion Channels of Cell Membranes, Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia, 194064
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Elena Kaznacheyeva
- Laboratory of Ion Channels of Cell Membranes, Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia, 194064.
| |
Collapse
|
3
|
Qiao S, Jia Y, Xie L, Jing W, Xia Y, Song Y, Zhang J, Cao T, Song H, Meng L, Shi L, Zhang X. KLF7 promotes neuroblastoma differentiation through the GTPase signaling pathway by upregulating neuroblast differentiation-associated protein AHNAKs and glycerophosphodiesterase GDPD5. FEBS J 2024; 291:3870-3888. [PMID: 38924469 DOI: 10.1111/febs.17208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
The arrest of neural crest-derived sympathoadrenal neuroblast differentiation contributes to neuroblastoma formation, and overriding this blocked differentiation is a clear strategy for treating high-risk neuroblastoma. A better understanding of neuroblast or neuroblastoma differentiation is essential for developing new therapeutic approaches. It has been proposed that Krueppel-like factor 7 (KLF7) is a neuroblastoma super-enhancer-associated transcription factor gene. Moreover, KLF7 was found to be intensely active in postmitotic neuroblasts of the developing nervous system during embryogenesis. However, the role of KLF7 in the differentiation of neuroblast or neuroblastoma is unknown. Here, we find a strong association between high KLF7 expression and favorable clinical outcomes in neuroblastoma. KLF7 induces differentiation of neuroblastoma cells independently of the retinoic acid (RA) pathway and acts cooperatively with RA to induce neuroblastoma differentiation. KLF7 alters the GTPase activity and multiple differentiation-related genes by binding directly to the promoters of neuroblast differentiation-associated protein (AHNAK and AHNAK2) and glycerophosphodiester phosphodiesterase domain-containing protein 5 (GDPD5) and regulating their expression. Furthermore, we also observe that silencing KLF7 in neuroblastoma cells promotes the adrenergic-to-mesenchymal transition accompanied by changes in enhancer-mediated gene expression. Our results reveal that KLF7 is an inducer of neuroblast or neuroblastoma differentiation with prognostic significance and potential therapeutic value.
Collapse
Affiliation(s)
- Shupei Qiao
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Ying Jia
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Li Xie
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Wenwen Jing
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Yang Xia
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Yue Song
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Jiahui Zhang
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Tianhua Cao
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Huilin Song
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Lingdi Meng
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, China
| | - Lei Shi
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, China
| | - Xue Zhang
- Heilongjiang Province Key Laboratory of Child Development and Genetic Research, Harbin Medical University, China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, China
| |
Collapse
|
4
|
Mignen O, Vannier JP, Schneider P, Renaudineau Y, Abdoul-Azize S. Orai1 Ca 2+ channel modulators as therapeutic tools for treating cancer: Emerging evidence! Biochem Pharmacol 2024; 219:115955. [PMID: 38040093 DOI: 10.1016/j.bcp.2023.115955] [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: 10/25/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
In non-excitable cells, Orai proteins represent the main channel for Store-Operated Calcium Entry (SOCE), and also mediate various store-independent Calcium Entry (SICE) pathways. Deregulation of these pathways contribute to increased tumor cell proliferation, migration, metastasis, and angiogenesis. Among Orais, Orai1 is an attractive therapeutic target explaining the development of specific modulators. Therapeutic trials using Orai1 channel inhibitors have been evaluated for treating diverse diseases such as psoriasis and acute pancreatitis, and emerging data suggest that Orai1 channel modulators may be beneficial for cancer treatment. This review discusses herein the importance of Orai1 channel modulators as potential therapeutic tools and the added value of these modulators for treating cancer.
Collapse
Affiliation(s)
| | | | | | - Yves Renaudineau
- Laboratory of Immunology, CHU Purpan Toulouse, INSERM U1291, CNRS U5051, University Toulouse III, 31062 Toulouse, France
| | - Souleymane Abdoul-Azize
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France; Normandie Univ., UNIROUEN, INSERM, U1234, Rouen 76000, France.
| |
Collapse
|
5
|
Alhasan MA, Tomokiyo A, Hamano S, Sugii H, Ono T, Ipposhi K, Yamashita K, Mardini B, Minowa F, Maeda H. Hyaluronic Acid Induction Promotes the Differentiation of Human Neural Crest-like Cells into Periodontal Ligament Stem-like Cells. Cells 2023; 12:2743. [PMID: 38067170 PMCID: PMC10705959 DOI: 10.3390/cells12232743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Periodontal ligament (PDL) stem-like cells (PDLSCs) are promising for regeneration of the periodontium because they demonstrate multipotency, high proliferative capacity, and the potential to regenerate bone, cementum, and PDL tissue. However, the transplantation of autologous PDLSCs is restricted by limited availability. Since PDLSCs are derived from neural crest cells (NCs) and NCs persist in adult PDL tissue, we devised to promote the regeneration of the periodontium by activating NCs to differentiate into PDLSCs. SK-N-SH cells, a neuroblastoma cell line that reportedly has NC-like features, seeded on the extracellular matrix of PDL cells for 2 weeks, resulted in the significant upregulation of PDL marker expression. SK-N-SH cell-derived PDLSCs (SK-PDLSCs) presented phenotypic characteristics comparable to induced pluripotent stem cell (iPSC)-derived PDLSCs (iPDLSCs). The expression levels of various hyaluronic acid (HA)-related genes were upregulated in iPDLSCs and SK-PDLSCs compared with iPSC-derived NCs and SK-N-SH cells, respectively. The knockdown of CD44 in SK-N-SH cells significantly inhibited their ability to differentiate into SK-PDLSCs, while low-molecular HA (LMWHA) induction enhanced SK-PDLSC differentiation. Our findings suggest that SK-N-SH cells could be applied as a new model to induce the differentiation of NCs into PDLSCs and that the LMWHA-CD44 relationship is important for the differentiation of NCs into PDLSCs.
Collapse
Affiliation(s)
- M. Anas Alhasan
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
| | - Atsushi Tomokiyo
- Department of Restorative Dentistry, Faculty of Dental Medicine, Hokkaido University, Kita13 Nishi7, Kita-ku, Sapporo 060-8586, Japan
| | - Sayuri Hamano
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Hideki Sugii
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
| | - Taiga Ono
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
| | - Keita Ipposhi
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
| | - Kozue Yamashita
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
| | - Bara Mardini
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
| | - Fumiko Minowa
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
| | - Hidefumi Maeda
- Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.A.A.); (S.H.); (H.S.); (T.O.); (K.I.); (K.Y.); (B.M.); (F.M.); (H.M.)
- Department of Endodontology, Kyushu University Hospital, Fukuoka 812-8582, Japan
| |
Collapse
|
6
|
Reichlmeir M, Canet-Pons J, Koepf G, Nurieva W, Duecker RP, Doering C, Abell K, Key J, Stokes MP, Zielen S, Schubert R, Ivics Z, Auburger G. In Cerebellar Atrophy of 12-Month-Old ATM-Null Mice, Transcriptome Upregulations Concern Most Neurotransmission and Neuropeptide Pathways, While Downregulations Affect Prominently Itpr1, Usp2 and Non-Coding RNA. Cells 2023; 12:2399. [PMID: 37830614 PMCID: PMC10572167 DOI: 10.3390/cells12192399] [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: 09/14/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2023] Open
Abstract
The autosomal recessive disorder Ataxia-Telangiectasia is caused by a dysfunction of the stress response protein, ATM. In the nucleus of proliferating cells, ATM senses DNA double-strand breaks and coordinates their repair. This role explains T-cell dysfunction and tumour risk. However, it remains unclear whether this function is relevant for postmitotic neurons and underlies cerebellar atrophy, since ATM is cytoplasmic in postmitotic neurons. Here, we used ATM-null mice that survived early immune deficits via bone-marrow transplantation, and that reached initial neurodegeneration stages at 12 months of age. Global cerebellar transcriptomics demonstrated that ATM depletion triggered upregulations in most neurotransmission and neuropeptide systems. Downregulated transcripts were found for the ATM interactome component Usp2, many non-coding RNAs, ataxia genes Itpr1, Grid2, immediate early genes and immunity factors. Allelic splice changes affected prominently the neuropeptide machinery, e.g., Oprm1. Validation experiments with stressors were performed in human neuroblastoma cells, where ATM was localised only to cytoplasm, similar to the brain. Effect confirmation in SH-SY5Y cells occurred after ATM depletion and osmotic stress better than nutrient/oxidative stress, but not after ATM kinase inhibition or DNA stressor bleomycin. Overall, we provide pioneer observations from a faithful A-T mouse model, which suggest general changes in synaptic and dense-core vesicle stress adaptation.
Collapse
Affiliation(s)
- Marina Reichlmeir
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Júlia Canet-Pons
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Gabriele Koepf
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Wasifa Nurieva
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Ruth Pia Duecker
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Claudia Doering
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany;
| | - Kathryn Abell
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Matthew P. Stokes
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Stefan Zielen
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
- Respiratory Research Institute, Medaimun GmbH, 60596 Frankfurt am Main, Germany
| | - Ralf Schubert
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Zoltán Ivics
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| |
Collapse
|
7
|
Bouron A. Neuronal Store-Operated Calcium Channels. Mol Neurobiol 2023:10.1007/s12035-023-03352-5. [PMID: 37118324 DOI: 10.1007/s12035-023-03352-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/13/2023] [Indexed: 04/30/2023]
Abstract
The endoplasmic reticulum (ER) is the major intracellular calcium (Ca2+) storage compartment in eukaryotic cells. In most instances, the mobilization of Ca2+ from this store is followed by a delayed and sustained uptake of Ca2+ through Ca2+-permeable channels of the cell surface named store-operated Ca2+ channels (SOCCs). This gives rise to a store-operated Ca2+ entry (SOCE) that has been thoroughly investigated in electrically non-excitable cells where it is the principal regulated Ca2+ entry pathway. The existence of this Ca2+ route in neurons has long been a matter of debate. However, a growing body of experimental evidence indicates that the recruitment of Ca2+ from neuronal ER Ca2+ stores generates a SOCE. The present review summarizes the main studies supporting the presence of a depletion-dependent Ca2+ entry in neurons. It also addresses the question of the molecular composition of neuronal SOCCs, their expression, pharmacological properties, as well as their physiological relevance.
Collapse
Affiliation(s)
- Alexandre Bouron
- Université Grenoble Alpes, CNRS, CEA, Inserm UA13 BGE, 38000, Grenoble, France.
| |
Collapse
|
8
|
Yoo JY, Lee YJ, Kim YJ, Baik TK, Lee JH, Lee MJ, Woo RS. Multiple low-dose radiation-induced neuronal cysteine transporter expression and oxidative stress are rescued by N-acetylcysteine in neuronal SH-SY5Y cells. Neurotoxicology 2023; 95:205-217. [PMID: 36796651 DOI: 10.1016/j.neuro.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/13/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Recently, several studies have demonstrated that low-dose radiation (LDR) therapy has positively impacts on the treatment of Alzheimer's disease (AD). LDR suppresses the production of pro-neuroinflammation molecules and improves cognitive function in AD. However, it is unclear whether direct exposure to LDR causes beneficial effects and what mechanism is involved in neuronal cells. In this study, we first determined the effect of high-dose radiation (HDR) alone on C6 cells and SH-SY5Y cells. We found that SH-SY5Y cells were more vulnerable than C6 cells to HDR. Moreover, in neuronal SH-SY5Y cells exposed to single or multiple LDR, N-type cells showed decreased cell viability with increasing radiation exposure time and frequency, but S-type cells were unaffected. Multiple LDR increased proapoptotic molecules such as p53, Bax and cleaved caspase-3, and decreased anti-apoptotic molecule (Bcl2). Multiple LDR also generated free radicals in neuronal SH-SY5Y cells. We detected a change in the expression of the neuronal cysteine transporter EAAC1. Pretreatment with N-acetylcysteine (NAC) rescued the increased in EAAC1 expression and the generation of ROS in neuronal SH-SY5Y cells after multiple LDR. Furthermore, we verified whether the increased in EAAC1 expression induces cell defense or cell death promotion signaling. We showed that transient overexpression of EAAC1 reduced the multiple LDR-induced p53 overexpression in neuronal SH-SY5Y cells. Our results indicate that neuronal cells can be injured by increased production of ROS not only by HDR but also by multiple LDR, which suggests that combination treatment with anti-free radical agents such as NAC may be useful in multiple LDR therapy.
Collapse
Affiliation(s)
- Ji-Young Yoo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Ye-Ji Lee
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Yu-Jin Kim
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Tai-Kyoung Baik
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon 34520, Republic of Korea
| | - Mi-Jo Lee
- Department of Radiation Oncology, Eulji University Hospital, Daejeon 35233, Republic of Korea.
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea.
| |
Collapse
|
9
|
Wang YY, Wang WC, Su CW, Hsu CW, Yuan SS, Chen YK. Overexpression of transient receptor potential melastatin 6 during human oral squamous cell carcinogenesis. J Dent Sci 2023; 18:382-391. [PMID: 36643266 PMCID: PMC9831831 DOI: 10.1016/j.jds.2022.11.002] [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] [Received: 10/30/2022] [Revised: 10/30/2022] [Indexed: 11/16/2022] Open
Abstract
Background/purpose Transient receptor potential melastatin (TRPM) channel is involved in cell proliferation and cell survival. Eight members (TRPM1-8) are within the TRPM subfamily. The current study is aimed to investigate TRPM6 expression in human oral carcinogenesis. Materials and methods Sixty-six oral squamous cell carcinomas (OSCCs), 47 oral potentially malignant disorders (OPMD) with moderate-severe epithelial dysplasia (ED), 28 OPMD with mild ED, and 33 normal oral mucosa (NOM) samples were subjected to immunohistochemical staining. Two human oral cancer cell lines (OCCLs), an oral premalignant cell line (DOK), and a normal oral keratinocyte culture (HOK) were used for Western blot analysis. OCCLs were evaluated for proliferation, migration, invasion assays, and intracellular calcium concentration. Results TRPM6 protein expression in OSCC was significantly increased as compared with normal samples. Protein expression of TRPM6 in OCCLs was significantly higher as compared with HOK. Significant decreases in degrees of proliferation, migration, invasion, and intracellular calcium concentration were noted in OCCLs with TRPM6 siRNA transfection as compared with those without transfection. Significantly increased TRPM6 protein level was noted in OPMD with moderate-severe ED as compared with those with mild ED. Conclusion Our results implicate that TRPM6 overexpression is potentially related to human oral carcinogenesis.
Collapse
Affiliation(s)
- Yen-Yun Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Chen Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Division of Oral Pathology & Maxillofacial Radiology, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan,Oral & Maxillofacial Imaging Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chiang-Wei Su
- Division of Oral & Maxillofacial Surgery, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ching-Wei Hsu
- Division of Oral & Maxillofacial Surgery, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shyng-Shiou Yuan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Corresponding author. Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan.
| | - Yuk-Kwan Chen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Division of Oral Pathology & Maxillofacial Radiology, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan,Oral & Maxillofacial Imaging Center, Kaohsiung Medical University, Kaohsiung, Taiwan,Corresponding author. School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan.
| |
Collapse
|
10
|
Khazeem MM, Casement JW, Schlossmacher G, Kenneth NS, Sumbung NK, Chan JYT, McGow JF, Cowell IG, Austin CA. TOP2B Is Required to Maintain the Adrenergic Neural Phenotype and for ATRA-Induced Differentiation of SH-SY5Y Neuroblastoma Cells. Mol Neurobiol 2022; 59:5987-6008. [PMID: 35831557 PMCID: PMC9463316 DOI: 10.1007/s12035-022-02949-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 06/23/2022] [Indexed: 12/13/2022]
Abstract
The neuroblastoma cell line SH-SY5Y is widely used to study retinoic acid (RA)-induced gene expression and differentiation and as a tool to study neurodegenerative disorders. SH-SY5Y cells predominantly exhibit adrenergic neuronal properties, but they can also exist in an epigenetically interconvertible alternative state with more mesenchymal characteristics; as a result, these cells can be used to study gene regulation circuitry controlling neuroblastoma phenotype. Using a combination of pharmacological inhibition and targeted gene inactivation, we have probed the requirement for DNA topoisomerase IIB (TOP2B) in RA-induced gene expression and differentiation and in the balance between adrenergic neuronal versus mesenchymal transcription programmes. We found that expression of many, but not all genes that are rapidly induced by ATRA in SH-SY5Y cells was significantly reduced in the TOP2B null cells; these genes include BCL2, CYP26A1, CRABP2, and NTRK2. Comparing gene expression profiles in wild-type versus TOP2B null cells, we found that long genes and genes expressed at a high level in WT SH-SY5Y cells were disproportionately dependent on TOP2B. Notably, TOP2B null SH-SY5Y cells upregulated mesenchymal markers vimentin (VIM) and fibronectin (FN1) and components of the NOTCH signalling pathway. Enrichment analysis and comparison with the transcription profiles of other neuroblastoma-derived cell lines supported the conclusion that TOP2B is required to fully maintain the adrenergic neural-like transcriptional signature of SH-SY5Y cells and to suppress the alternative mesenchymal epithelial-like epigenetic state.
Collapse
Affiliation(s)
- Mushtaq M Khazeem
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,National Center of Hematology, Mustansiriyah University, Baghdad, Iraq
| | - John W Casement
- Bioinformatics Support Unit, The Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - George Schlossmacher
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Niall S Kenneth
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Nielda K Sumbung
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Janice Yuen Tung Chan
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Jade F McGow
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Ian G Cowell
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - Caroline A Austin
- The Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
11
|
Streamlining Culture Conditions for the Neuroblastoma Cell Line SH-SY5Y: A Prerequisite for Functional Studies. Methods Protoc 2022; 5:mps5040058. [PMID: 35893584 PMCID: PMC9326679 DOI: 10.3390/mps5040058] [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] [Received: 05/11/2022] [Revised: 06/30/2022] [Accepted: 07/08/2022] [Indexed: 11/23/2022] Open
Abstract
The neuroblastoma cell line SH-SY5Y has been a well-established and very popular in vitro model in neuroscience for decades, especially focusing on neurodevelopmental disorders, such as Parkinson’s disease. The ability of this cell type to differentiate compared with other models in neurobiology makes it one of the few suitable models without having to rely on a primary culture of neuronal cells. Over the years, various, partly contradictory, methods of cultivation have been reported. This study is intended to provide a comprehensive guide to the in vitro cultivation of undifferentiated SH-SY5Y cells. For this purpose, the morphology of the cell line and the differentiation of the individual subtypes are described, and instructions for cell culture practice and long-term cryoconservation are provided. We describe the key growth characteristics of this cell line, including proliferation and confluency data, optimal initial seeding cell numbers, and a comparison of different culture media and cell viability during cultivation. Furthermore, applying an optimized protocol in a long-term cultivation over 60 days, we show that cumulative population doubling (CPD) is constant over time and does not decrease with incremental passage, enabling stable cultivation, for example, for recurrent differentiation to achieve the highest possible reproducibility in subsequent analyses. Therefore, we provide a solid guidance for future research that employs the neuroblastoma cell line SH-SY5Y.
Collapse
|
12
|
Wang YY, Wang WC, Su CW, Hsu CW, Yuan SS, Chen YK. Expression of Orai1 and STIM1 in human oral squamous cell carcinogenesis. J Dent Sci 2022; 17:78-88. [PMID: 35028023 PMCID: PMC8739746 DOI: 10.1016/j.jds.2021.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/07/2021] [Indexed: 12/21/2022] Open
Abstract
Background/purpose Return of Ca2+ to endoplasmic reticulum is mediated by Orai/STIM-mediated store-operated Ca2+ entry (SOCE) channel. We aimed to investigate Orai1 and STIM1 expressions in human oral carcinogenesis. Materials and methods Sixty-six oral squamous cell carcinomas (OSCCs), 14 oral potentially malignant disorders (OPMD) with moderate-severe oral epithelial dysplasia (OED), 19 OPMD with mild OED, and 14 normal oral mucosa (NOM) samples were subjected to immunohistochemical staining. Two human oral cancer cell lines (OCCLs), an oral premalignant cell line (DOK), and a normal oral keratinocyte culture (HOK) were used for Western blot and real-time quantitative reverse transcription-polymerase chain reaction. OCCLs were evaluated for proliferation, migration, and invasion assays. Results Orai1 and STIM1 protein and mRNA expressions in OSCC were significantly enhanced as compared with normal samples. Protein expressions of Orai1 and STIM1 in OCCLs were significantly enhanced as compared with HOK. Significant decreases in degrees of proliferation, migration and invasion were noted in OCCLs with Orai1 and STIM1 siRNA transfection as compared with those without transfection. Significantly increased Orai1 and STIM1 protein levels were noted in OPMD with moderate-severe OED as compared with those with mild OED. Conclusion Our results indicate that Orai1 and STIM1 overexpression is associated with human oral carcinogenesis.
Collapse
Affiliation(s)
- Yen-Yun Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Chen Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Oral Pathology & Maxillofacial Radiology, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Oral & Maxillofacial Imaging Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chiang-Wei Su
- Division of Oral & Maxillofacial Surgery, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ching-Wei Hsu
- Division of Oral & Maxillofacial Surgery, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shyng-Shiou Yuan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Obstetrics and Gynecology and Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuk-Kwan Chen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Oral Pathology & Maxillofacial Radiology, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Oral & Maxillofacial Imaging Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
13
|
Downing K, Prisby R, Varanasi V, Zhou J, Pan Z, Brotto M. Old and new biomarkers for volumetric muscle loss. Curr Opin Pharmacol 2021; 59:61-69. [PMID: 34146835 DOI: 10.1016/j.coph.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022]
Abstract
Volumetric muscle loss (VML) impacts skeletal muscles and causes damage to associated tissues such as blood vessels and other structural tissues. Despite progress in the VML field, current preclinical approaches are often ineffective at restoring muscle volume. Additional research is paramount to develop strategies that improve muscle mass and function, while restoring supporting tissues. We highlight mechanisms that govern normal muscle function that are also key players for VML, including intracellular calcium signaling/homeostasis, mitochondria signaling (calcium, reactiove oxidative species (ROS)/oxidative stress), and angiogenesis. We propose an integration of these processes within the context of emerging biomaterials that provide structural support for muscle regeneration. We posit that new biomarkers (i.e. myokines and lipid signaling mediators) may serve as sentinels of early muscle injury and regeneration. We conclude that as new ideas, approaches, and models come together, new treatments will emerge to allow the full rebuilding of skeletal muscles and functional recovery of skeletal muscles after VML.
Collapse
Affiliation(s)
- Kerrie Downing
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Rhonda Prisby
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Venu Varanasi
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Jingsong Zhou
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA
| | - Zui Pan
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA.
| | - Marco Brotto
- Bone-Muscle Collaborative Sciences, College of Nursing & Health Innovation, The University of Texas at Arlington, Arlington, TX 76010, USA.
| |
Collapse
|
14
|
Capone R, Tiwari A, Hadziselimovic A, Peskova Y, Hutchison JM, Sanders CR, Kenworthy AK. The C99 domain of the amyloid precursor protein resides in the disordered membrane phase. J Biol Chem 2021; 296:100652. [PMID: 33839158 PMCID: PMC8113881 DOI: 10.1016/j.jbc.2021.100652] [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: 11/27/2020] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Processing of the amyloid precursor protein (APP) via the amyloidogenic pathway is associated with the etiology of Alzheimer's disease. The cleavage of APP by β-secretase to generate the transmembrane 99-residue C-terminal fragment (C99) and subsequent processing of C99 by γ-secretase to yield amyloid-β (Aβ) peptides are essential steps in this pathway. Biochemical evidence suggests that amyloidogenic processing of C99 occurs in cholesterol- and sphingolipid-enriched liquid-ordered phase membrane rafts. However, direct evidence that C99 preferentially associates with these rafts has remained elusive. Here, we tested this by quantifying the affinity of C99-GFP for raft domains in cell-derived giant plasma membrane vesicles (GPMVs). We found that C99 was essentially excluded from ordered domains in vesicles from HeLa cells, undifferentiated SH-SY5Y cells, or SH-SY5Y-derived neurons; instead, ∼90% of C99 partitioned into disordered domains. The strong association of C99 with disordered domains occurred independently of its cholesterol-binding activity or homodimerization, or of the presence of the familial Alzheimer disease Arctic mutation (APP E693G). Finally, through biochemical studies we confirmed previous results, which showed that C99 is processed in the plasma membrane by α-secretase, in addition to the well-known γ-secretase. These findings suggest that C99 itself lacks an intrinsic affinity for raft domains, implying that either i) amyloidogenic processing of the protein occurs in disordered regions of the membrane, ii) processing involves a marginal subpopulation of C99 found in rafts, or iii) as-yet-unidentified protein-protein interactions with C99 in living cells drive this protein into membrane rafts to promote its cleavage therein.
Collapse
Affiliation(s)
- Ricardo Capone
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ajit Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Yelena Peskova
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
| | - James M Hutchison
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Charles R Sanders
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anne K Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
15
|
Campos Cogo S, Gradowski Farias da Costa do Nascimento T, de Almeida Brehm Pinhatti F, de França Junior N, Santos Rodrigues B, Regina Cavalli L, Elifio-Esposito S. An overview of neuroblastoma cell lineage phenotypes and in vitro models. Exp Biol Med (Maywood) 2020; 245:1637-1647. [PMID: 32787463 PMCID: PMC7802384 DOI: 10.1177/1535370220949237] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
This review was conducted to present the main neuroblastoma (NB) clinical characteristics and the most common genetic alterations present in these pediatric tumors, highlighting their impact in tumor cell aggressiveness behavior, including metastatic development and treatment resistance, and patients' prognosis. The distinct three NB cell lineage phenotypes, S-type, N-type, and I-type, which are characterized by unique cell surface markers and gene expression patterns, are also reviewed. Finally, an overview of the most used NB cell lines currently available for in vitro studies and their unique cellular and molecular characteristics, which should be taken into account for the selection of the most appropriate model for NB pre-clinical studies, is presented. These valuable models can be complemented by the generation of NB reprogrammed tumor cells or organoids, derived directly from patients' tumor specimens, in the direction toward personalized medicine.
Collapse
Affiliation(s)
- Sheron Campos Cogo
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | | | | | - Nilton de França Junior
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | - Bruna Santos Rodrigues
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | - Luciane Regina Cavalli
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, Brazil
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Selene Elifio-Esposito
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| |
Collapse
|
16
|
Consales C, Butera A, Merla C, Pasquali E, Lopresto V, Pinto R, Pierdomenico M, Mancuso M, Marino C, Benassi B. Exposure of the SH-SY5Y Human Neuroblastoma Cells to 50-Hz Magnetic Field: Comparison Between Two-Dimensional (2D) and Three-Dimensional (3D) In Vitro Cultures. Mol Neurobiol 2020; 58:1634-1649. [PMID: 33230715 PMCID: PMC7932966 DOI: 10.1007/s12035-020-02192-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
We here characterize the response to the extremely low-frequency (ELF) magnetic field (MF, 50 Hz, 1 mT) of SH-SY5Y human neuroblastoma cells, cultured in a three-dimensional (3D) Alvetex® scaffold compared to conventional two-dimensional (2D) monolayers. We proved that the growing phenotype of proliferating SH-SY5Y cells is not affected by the culturing conditions, as morphology, cell cycle distribution, proliferation/differentiation gene expression of 3D-cultures overlap what reported in 2D plates. In response to 72-h exposure to 50-Hz MF, we demonstrated that no proliferation change and apoptosis activation occur in both 2D and 3D cultures. Consistently, no modulation of Ki67, MYCN, CCDN1, and Nestin, of invasiveness and neo-angiogenesis-controlling genes (HIF-1α, VEGF, and PDGF) and of microRNA epigenetic signature (miR-21-5p, miR-222-3p and miR-133b) is driven by ELF exposure. Conversely, intracellular glutathione content and SOD1 expression are exclusively impaired in 3D-culture cells in response to the MF, whereas no change of such redox modulators is observed in SH-SY5Y cells if grown on 2D monolayers. Moreover, ELF-MF synergizes with the differentiating agents to stimulate neuroblastoma differentiation into a dopaminergic (DA) phenotype in the 3D-scaffold culture only, as growth arrest and induction of p21, TH, DAT, and GAP43 are reported in ELF-exposed SH-SY5Y cells exclusively if grown on 3D scaffolds. As overall, our findings prove that 3D culture is a more reliable experimental model for studying SH-SY5Y response to ELF-MF if compared to 2D conventional monolayer, and put the bases for promoting 3D systems in future studies addressing the interaction between electromagnetic fields and biological systems.
Collapse
Affiliation(s)
- Claudia Consales
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Alessio Butera
- Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Caterina Merla
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Emanuela Pasquali
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Vanni Lopresto
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Rosanna Pinto
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Maria Pierdomenico
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Carmela Marino
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
17
|
Moreira NCDS, Lima JEBDF, Chierrito TPC, Carvalho I, Sakamoto-Hojo ET. Novel Hybrid Acetylcholinesterase Inhibitors Induce Differentiation and Neuritogenesis in Neuronal Cells in vitro Through Activation of the AKT Pathway. J Alzheimers Dis 2020; 78:353-370. [PMID: 32986667 DOI: 10.3233/jad-200425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a progressive loss of episodic memory associated with amyloid-β peptide aggregation and the abnormal phosphorylation of the tau protein, leading to the loss of cholinergic function. Acetylcholinesterase (AChE) inhibitors are the main class of drugs used in AD therapy. OBJECTIVE The aim of the current study was to evaluate the potential of two tacrine-donepezil hybrid molecules (TA8Amino and TAHB3), which are AChE inhibitors, to induce neurodifferentiation and neuritogenesis in SH-SY5Y cells. METHODS The experiments were carried out to characterize neurodifferentiation, cellular changes related to responses to oxidative stress and pathways of cell survival in response to drug treatments. RESULTS The results indicated that the compounds did not present cytotoxic effects in SH-SY5Y or HepG2 cells. TA8Amino and TAHB3 induced neurodifferentiation and neuritogenesis in SH-SY5Y cells. These cells showed increased levels of intracellular and mitochondrial reactive oxygen species; the induction of oxidative stress was also demonstrated by an increase in SOD1 expression in TA8Amino and TAHB3-treated cells. Cells treated with the compounds showed an increase in PTEN(Ser380/Thr382/383) and AKT(Ser473) expression, suggesting the involvement of the AKT pathway. CONCLUSION Our results demonstrated that TA8Amino and TAHB3 present advantages as potential drugs for AD therapy and that they are capable of inducing neurodifferentiation and neuritogenesis.
Collapse
Affiliation(s)
| | | | | | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
18
|
Kronenberg NM, Tilston-Lunel A, Thompson FE, Chen D, Yu W, Dholakia K, Gather MC, Gunn-Moore FJ. Willin/FRMD6 Influences Mechanical Phenotype and Neuronal Differentiation in Mammalian Cells by Regulating ERK1/2 Activity. Front Cell Neurosci 2020; 14:552213. [PMID: 33088261 PMCID: PMC7498650 DOI: 10.3389/fncel.2020.552213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022] Open
Abstract
Willin/FRMD6 is part of a family of proteins with a 4.1 ezrin-radixin-moesin (FERM) domain. It has been identified as an upstream activator of the Hippo pathway and, when aberrant in its expression, is associated with human diseases and disorders. Even though Willin/FRMD6 was originally discovered in the rat sciatic nerve, most studies have focused on its functional roles in cells outside of the nervous system, where Willin/FRMD6 is involved in the formation of apical junctional cell-cell complexes and in regulating cell migration. Here, we investigate the biochemical and biophysical role of Willin/FRMD6 in neuronal cells, employing the commonly used SH-SY5Y neuronal model cell system and combining biochemical measurements with Elastic Resonator Interference Stress Micropscopy (ERISM). We present the first direct evidence that Willin/FRMD6 expression influences both the cell mechanical phenotype and neuronal differentiation. By investigating cells with increased and decreased Willin/FRMD6 expression levels, we show that Willin/FRMD6 not only affects proliferation and migration capacity of cells but also leads to changes in cell morphology and an enhanced formation of neurite-like membrane extensions. These changes were accompanied by alterations of biophysical parameters such as cell force, the organization of actin stress fibers and the formation of focal adhesions. At the biochemical level, changes in Willin/FRMD6 expression inversely affected the activity of the extracellular signal-regulated kinases (ERK) pathway and downstream transcriptional factor NeuroD1, which seems to prime SH-SY5Y cells for retinoic acid (RA)-induced neuronal differentiation.
Collapse
Affiliation(s)
- Nils M Kronenberg
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom.,Centre for Nanobiophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Andrew Tilston-Lunel
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom.,Department of Biochemistry, School of Medicine, Boston University, Boston, MA, United States
| | - Frances E Thompson
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom
| | - Doris Chen
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom
| | - Wanjia Yu
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom
| | - Kishan Dholakia
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom.,Department of Physics, College of Science, Yonsei University, Seoul, South Korea
| | - Malte C Gather
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom.,Centre for Nanobiophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Frank J Gunn-Moore
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
19
|
Yang CL, Serra-Roma A, Gualandi M, Bodmer N, Niggli F, Schulte JH, Bode PK, Shakhova O. Lineage-restricted sympathoadrenal progenitors confer neuroblastoma origin and its tumorigenicity. Oncotarget 2020; 11:2357-2371. [PMID: 32595833 PMCID: PMC7299536 DOI: 10.18632/oncotarget.27636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/20/2020] [Indexed: 01/18/2023] Open
Abstract
Neuroblastoma (NB) is the most common cancer in infants and it accounts for six percent of all pediatric malignancies. There are several hypotheses proposed on the origins of NB. While there is little genetic evidence to support this, the prevailing model is that NB originates from neural crest stem cells (NCSCs). Utilizing in vivo mouse models, we demonstrate that targeting MYCN oncogene to NCSCs causes perinatal lethality. During sympathoadrenal (SA) lineage development, SOX transcriptional factors drive the transition from NCSCs to lineage-specific progenitors, characterized by the sequential activation of Sox9/Sox10/Sox4/Sox11 genes. We find the NCSCs factor SOX10 is not expressed in neuroblasts, but rather restricted to the Schwannian stroma and is associated with a good prognosis. On the other hand, SOX9 expression in NB cells was associated with several key biological processes including migration, invasion and differentiation. Moreover, manipulating SOX9 gene predominantly affects lineage-restricted SA progenitors. Our findings highlight a unique molecular SOX signature associated with NB that is highly reminiscent of SA progenitor transcriptional program during embryonic development, providing novel insights into NB pathobiology. In summary, we provide multiple lines of evidence suggesting that multipotent NCSCs do not contribute to NB initiation and maintenance.
Collapse
Affiliation(s)
- Chia-Lung Yang
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - André Serra-Roma
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Marco Gualandi
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Nicole Bodmer
- Department of Oncology, Children Hospital of Zürich, Zürich, Switzerland
| | - Felix Niggli
- Department of Oncology, Children Hospital of Zürich, Zürich, Switzerland
| | | | - Peter Karl Bode
- Department of Surgical Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Olga Shakhova
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
20
|
Calcium signalling in mammalian cell lines expressing wild type and mutant human α1-Antitrypsin. Sci Rep 2019; 9:17293. [PMID: 31754242 PMCID: PMC6872872 DOI: 10.1038/s41598-019-53535-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/28/2019] [Indexed: 11/08/2022] Open
Abstract
A possible role for calcium signalling in the autosomal dominant form of dementia, familial encephalopathy with neuroserpin inclusion bodies (FENIB), has been proposed, which may point towards a mechanism by which cells could sense and respond to the accumulation of mutant serpin polymers in the endoplasmic reticulum (ER). We therefore explored possible defects in Ca2+-signalling, which may contribute to the pathology associated with another serpinopathy, α1-antitrypsin (AAT) deficiency. Using CHO K1 cell lines stably expressing a wild type human AAT (MAAT) and a disease-causing polymer-forming variant (ZAAT) and the truncated variant (NHK AAT), we measured basal intracellular free Ca2+, its responses to thapsigargin (TG), an ER Ca2+-ATPase blocker, and store-operated Ca2+-entry (SOCE). Our fura2 based Ca2+ measurements detected no differences between these 3 parameters in cell lines expressing MAAT and cell lines expressing ZAAT and NHK AAT mutants. Thus, in our cell-based models of α1-antitrypsin (AAT) deficiency, unlike the case for FENIB, we were unable to detect defects in calcium signalling.
Collapse
|
21
|
Calcium signaling regulates fundamental processes involved in Neuroblastoma progression. Cell Calcium 2019; 82:102052. [DOI: 10.1016/j.ceca.2019.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022]
|
22
|
Halakos EG, Connell AJ, Glazewski L, Wei S, Mason RW. Bottom up proteomics reveals novel differentiation proteins in neuroblastoma cells treated with 13-cis retinoic acid. J Proteomics 2019; 209:103491. [PMID: 31472280 DOI: 10.1016/j.jprot.2019.103491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/15/2019] [Accepted: 08/15/2019] [Indexed: 12/19/2022]
Abstract
Neuroblastoma, a cancer of the sympathetic nervous system, is the second most common pediatric cancer. A unique feature of neuroblastoma is remission in some patients due to spontaneous differentiation of metastatic tumors. 13-cis retinoic acid (13-cis RA) is currently used in the clinic to treat neuroblastoma due to its differentiation inducing effects. In this study, we used shotgun proteomics to identify proteins affected by 13-cis RA treatment in neuroblastoma SK-N-SH cells. Our results showed that 13-cis RA reduced proteins involved in extracellular matrix synthesis and organization and increased proteins involved in cell adhesion and neurofilament formation. These changes indicate that 13-cis RA induces tumor cell differentiation by decreasing extracellular matrix rigidity and increasing neurite overgrowth. Differentially-affected proteins identified in this study may be novel biomarkers of drug efficacy in the treatment of neuroblastoma. SIGNIFICANCE: As neuroblastoma can spontaneously differentiate, determining which proteins are involved in differentiation can guide development of novel treatments. 13-cis retinoic acid is currently used in the clinic as a differentiation inducer. Here we have established a proteome map of SK-N-SH cells treated with 13-cis retinoic acid. Bioinformatic analysis revealed the involvement of development, differentiation, extracellular matrix assembly, collagen biosynthesis, and neurofilament bundle association. This proteome map provides information as to which proteins are important for differentiation and identifies networks that can be targeted by drugs to treat neuroblastoma [1].
Collapse
Affiliation(s)
- Effie G Halakos
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Andrew J Connell
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Lisa Glazewski
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Robert W Mason
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
23
|
Babayan N, Tadevosyan G, Khondkaryan L, Grigoryan R, Sarkisyan N, Haroutiounian R, Stopper H. Ochratoxin A induces global DNA hypomethylation and oxidative stress in neuronal cells in vitro. Mycotoxin Res 2019; 36:73-81. [PMID: 31441013 DOI: 10.1007/s12550-019-00370-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 08/07/2019] [Accepted: 08/13/2019] [Indexed: 12/24/2022]
Abstract
Recently, it was reported that ochratoxin A (OTA) mycotoxin, produced by a number of Aspergillus and Penicillium fungal species, may cause neuropsychological impairment or mental and emotional disorders but the mechanism of neurotoxicity remains unknown. Adverse effects of OTA in human (SHSY5Y) and mouse (HT22) neuronal cell lines were studied in vitro. OTA was found to be non-cytotoxic in both cell lines at concentrations 2.5-30 μmol/l, which are above the levels reported for human and animal plasma. OTA led to slightly elevated chromosomal instability in HT22 cells at concentrations of 15-30 μmol/l after 48 h, while in SHSY5Y cells, no evidence for genotoxic effects was observed at concentrations of 2.5-30 μmol/l. OTA treatment at 10 μmol/l resulted in elevated levels of unmethylated cytosines in CpG dinucleotides (up to 1.4-fold), elevated levels of intracellular reactive oxygen species (up to 1.6-fold), and in elevated levels of oxidized DNA purines (up to 2.2-fold) in both cell lines. Detected global DNA hypomethylation and oxidative stress were found to be reversible in 96 h and 24-72 h, respectively. In general, the observed pattern of OTA-induced effects in both cell lines was similar, but HT22 cells exhibited higher sensitivity, as well as better repair capacity in response to OTA toxicity. In conclusion, the results suggest that oxidative stress and epigenetic changes are directly involved in OTA-induced neurotoxicity, while cytotoxicity and genotoxicity cannot be considered as primary cause of toxicity in neuronal cells in vitro.
Collapse
Affiliation(s)
- Nelly Babayan
- Institute of Molecular Biology, National Academy of Sciences of Armenia, Hasratyan 7, 0014, Yerevan, Armenia. .,Yerevan State University, A. Manoogian 1, 0025, Yerevan, Armenia.
| | - Gohar Tadevosyan
- Institute of Molecular Biology, National Academy of Sciences of Armenia, Hasratyan 7, 0014, Yerevan, Armenia
| | - Lusine Khondkaryan
- Institute of Molecular Biology, National Academy of Sciences of Armenia, Hasratyan 7, 0014, Yerevan, Armenia
| | - Ruzanna Grigoryan
- Institute of Molecular Biology, National Academy of Sciences of Armenia, Hasratyan 7, 0014, Yerevan, Armenia
| | - Natalya Sarkisyan
- Institute of Molecular Biology, National Academy of Sciences of Armenia, Hasratyan 7, 0014, Yerevan, Armenia
| | | | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher 9, 997078, Wuerzburg, Germany
| |
Collapse
|
24
|
Litowczenko J, Maciejewska BM, Wychowaniec JK, Kościński M, Jurga S, Warowicka A. Groove‐patterned surfaces induce morphological changes in cells of neuronal origin. J Biomed Mater Res A 2019; 107:2244-2256. [DOI: 10.1002/jbm.a.36733] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Jagoda Litowczenko
- NanoBioMedical CentreAdam Mickiewicz University Poznań Poland
- Department of Molecular Virology, Faculty of BiologyAdam Mickiewicz University Poznań Poland
| | | | - Jacek K. Wychowaniec
- NanoBioMedical CentreAdam Mickiewicz University Poznań Poland
- School of ChemistryUniversity College Dublin Dublin Ireland
| | - Mikołaj Kościński
- NanoBioMedical CentreAdam Mickiewicz University Poznań Poland
- Department of Physics and Biophysics, Faculty of Food Science and NutritionPoznań University of Life Sciences Poznań Poland
| | - Stefan Jurga
- NanoBioMedical CentreAdam Mickiewicz University Poznań Poland
| | - Alicja Warowicka
- NanoBioMedical CentreAdam Mickiewicz University Poznań Poland
- Department of Animal Physiology and Development, Institute of Experimental BiologyAdam Mickiewicz University Poznań Poland
| |
Collapse
|
25
|
Chalatsa I, Arvanitis DA, Koulakiotis NS, Giagini A, Skaltsounis AL, Papadopoulou-Daifoti Z, Tsarbopoulos A, Sanoudou D. The Crocus sativus Compounds trans-Crocin 4 and trans-Crocetin Modulate the Amyloidogenic Pathway and Tau Misprocessing in Alzheimer Disease Neuronal Cell Culture Models. Front Neurosci 2019; 13:249. [PMID: 30971876 PMCID: PMC6443833 DOI: 10.3389/fnins.2019.00249] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/04/2019] [Indexed: 11/23/2022] Open
Abstract
Crocus sativus L. natural compounds have been extensively used in traditional medicine for thousands of years. Recent research evidence is now emerging in support of its therapeutic potential for different pathologies including neurodegenerative diseases. Herein, the C. sativus L. natural compounds trans-crocin 4 and trans-crocetin were selected for in depth molecular characterization of their potentially protective effects against Alzheimer’s Disease (AD), utilizing two AD neuronal cell culture models (SH-SY5Y overexpressing APP and PC12 expressing hyperphosphorylated tau). Biologically relevant concentrations, ranging from 0.1 μM to 1 mM, applied for 24 h or 72 h, were well tolerated by differentiated wild type SH-SY5Y and PC12 cells. When tested on neuronally differentiated SH-SY5Y-APP both trans-crocin 4 and trans-crocetin had significant effects against amyloidogenic pathways. Trans-crocin 4 significantly decreased of β-secretase, a key enzyme of the amyloidogenic pathway, and APP-C99, while it decreased γ-secretases that generate toxic beta-amyloid peptides. Similarly, trans-crocetin treatment led to a reduction in β- and γ-secretases, as well as to accumulation of cellular AβPP. When tested on the neuronally differentiated PC12-htau cells, both compounds proved effective in suppressing the active forms of GSK3β and ERK1/2 kinases, as well as significantly reducing total tau and tau phosphorylation. Collectively, our data demonstrate a potent effect of trans-crocin 4 and trans-crocetin in suppressing key molecular pathways of AD pathogenesis, rendering them a promising tool in the prevention and potentially the treatment of AD.
Collapse
Affiliation(s)
- Ioanna Chalatsa
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Demetrios A Arvanitis
- Molecular Biology Division, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Athina Giagini
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexios Leandros Skaltsounis
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Zeta Papadopoulou-Daifoti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anthony Tsarbopoulos
- GAIA Research Center, Bioanalytical Department, The Goulandris Natural History Museum, Athens, Greece.,Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Molecular Biology Division, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
26
|
Maklad A, Sharma A, Azimi I. Calcium Signaling in Brain Cancers: Roles and Therapeutic Targeting. Cancers (Basel) 2019; 11:cancers11020145. [PMID: 30691160 PMCID: PMC6406375 DOI: 10.3390/cancers11020145] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Calcium signaling, in addition to its numerous physiological roles, is also implicated in several pathological conditions including cancer. An increasing body of evidence suggest critical roles of calcium signaling in the promotion of different aspects of cancer, including cell proliferation, therapy resistance and metastatic-related processes. In many cases, this is associated with altered expression and/or activity of some calcium channels and pumps. Brain cancers have also been the subject of many of these studies. In addition to diverse roles of calcium signals in normal brain function, a number of proteins involved in calcium transport are implicated to have specific roles in some brain cancers including gliomas, medulloblastoma, neuroblastoma and meningioma. This review discusses research that has been conducted so far to understand diverse roles of Ca2+-transporting proteins in the progression of brain cancers, as well as any attempts to target these proteins towards a therapeutic approach for the control of brain cancers. Finally, some knowledge gaps in the field that may need to be further considered are also discussed.
Collapse
Affiliation(s)
- Ahmed Maklad
- Division of Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7001, Australia.
| | - Anjana Sharma
- Division of Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7001, Australia.
| | - Iman Azimi
- Division of Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7001, Australia.
| |
Collapse
|
27
|
Ragucci S, Pacifico S, Ruocco MR, Crescente G, Nasso R, Simonetti M, Masullo M, Piccolella S, Pedone PV, Landi N, Di Maro A. Ageritin from poplar mushrooms: scale-up purification and cytotoxicity towards undifferentiated and differentiated SH-SY5Y cells. Food Funct 2019; 10:6342-6350. [DOI: 10.1039/c9fo01483g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ageritin from poplar mushroom is a valuable selective neurotoxin towards undifferentiated neuroblastoma SH-SY5Y cells.
Collapse
|
28
|
Vulinovic F, Krajka V, Hausrat TJ, Seibler P, Alvarez-Fischer D, Madoev H, Park JS, Kumar KR, Sue CM, Lohmann K, Kneussel M, Klein C, Rakovic A. Motor protein binding and mitochondrial transport are altered by pathogenic TUBB4A variants. Hum Mutat 2018; 39:1901-1915. [PMID: 30079973 DOI: 10.1002/humu.23602] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/05/2018] [Accepted: 07/29/2018] [Indexed: 12/21/2022]
Abstract
Mutations in TUBB4A have been identified to cause a wide phenotypic spectrum of diseases ranging from hereditary generalized dystonia with whispering dysphonia (DYT-TUBB4A) and hereditary spastic paraplegia (HSP) to leukodystrophy hypomyelination with atrophy of the basal ganglia and cerebellum (H-ABC). TUBB4A encodes the brain-specific β-tubulin isotype, β-tubulin 4A. To elucidate the pathogenic mechanisms conferred by TUBB4A mutations leading to the different phenotypes, we functionally characterized three pathogenic TUBB4A variants (c.4C>G,p.R2G; c.745G>A,p.D249N; c.811G>A, p.A271T) as representatives of the mutational and disease spectrum) in human neuroblastoma cells and human induced pluripotent stem cell (iPSC)-derived neurons. We showed that mRNA stability was not affected by any of the TUBB4A variants. Although two mutations (p.R2G and p.D249N) are located at the α/β-tubulin interdimer interface, we confirmed incorporation of all TUBB4A mutants into the microtubule network. However, we showed that the mutations p.D249N and p.A271T interfered with motor protein binding to microtubules and impaired neurite outgrowth and microtubule dynamics. Finally, TUBB4A mutations, as well as heterozygous knockout of TUBB4A, disrupted mitochondrial transport in iPSC-derived neurons. Taken together, our findings suggest that functional impairment of microtubule-associated transport is a shared pathogenic mechanism by which the TUBB4A mutations studied here cause a spectrum of diseases.
Collapse
Affiliation(s)
- Franca Vulinovic
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Victor Krajka
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Torben J Hausrat
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | - Harutyun Madoev
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Jin-Sung Park
- Department of Neurogenetics, Kolling Institute, Royal North Shore Hospital and the University of Sydney, St. Leonards, New South Wales, Australia
| | - Kishore R Kumar
- Department of Neurogenetics, Kolling Institute, Royal North Shore Hospital and the University of Sydney, St. Leonards, New South Wales, Australia
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Carolyn M Sue
- Department of Neurogenetics, Kolling Institute, Royal North Shore Hospital and the University of Sydney, St. Leonards, New South Wales, Australia
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | |
Collapse
|
29
|
Whitworth CL, Redfern CPF, Cheek TR. Differentiation-Induced Remodelling of Store-Operated Calcium Entry Is Independent of Neuronal or Glial Phenotype but Modulated by Cellular Context. Mol Neurobiol 2018; 56:857-872. [PMID: 29802571 PMCID: PMC6400879 DOI: 10.1007/s12035-018-1112-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/09/2018] [Indexed: 01/20/2023]
Abstract
Neurogenesis is a complex process leading to the generation of neuronal networks and glial cell types from stem cells or intermediate progenitors. Mapping subcellular and molecular changes accompanying the switch from proliferation to differentiation is vital for developing therapeutic targets for neurological diseases. Neuronal (N-type) and glial (S-type) phenotypes within the SH-SY5Y neuroblastoma cell line have distinct differentiation responses to 9-cis-retinoic acid (9cRA). In both cell phenotypes, these were accompanied at the single cell level by an uncoupling of Ca2+ store release from store-operated Ca2+ entry (SOCE), mediated by changes in the expression of calcium release-activated calcium pore proteins. This remodelling of calcium signalling was moderated by the predominant cell phenotype within the population. N- and S-type cells differed markedly in their phenotypic stability after withdrawal of the differentiation inducer, with the phenotypic stability of S-type cells, both morphologically and with respect to SOCE properties, in marked contrast to the lability of the N-type phenotype. Furthermore, the SOCE response of I-type cells, a presumed precursor to both N- and S-type cells, varied markedly in different cell environments. These results demonstrate the unique biology of neuronal and glial derivatives of common precursors and suggest that direct or indirect interactions between cell types are vital components of neurogenesis that need to be considered in experimental models.
Collapse
Affiliation(s)
- Claire L Whitworth
- Institute for Cell and Molecular Biosciences, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Division of Biological Chemistry & Drug Discovery, School of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| | - Christopher P F Redfern
- Northern Institute for Cancer Research, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Timothy R Cheek
- Institute for Cell and Molecular Biosciences, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
30
|
Kemp K, Dey R, Cook A, Scolding N, Wilkins A. Mesenchymal Stem Cell-Derived Factors Restore Function to Human Frataxin-Deficient Cells. CEREBELLUM (LONDON, ENGLAND) 2017; 16:840-851. [PMID: 28456899 PMCID: PMC5498643 DOI: 10.1007/s12311-017-0860-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Friedreich's ataxia is an inherited neurological disorder characterised by mitochondrial dysfunction and increased susceptibility to oxidative stress. At present, no therapy has been shown to reduce disease progression. Strategies being trialled to treat Friedreich's ataxia include drugs that improve mitochondrial function and reduce oxidative injury. In addition, stem cells have been investigated as a potential therapeutic approach. We have used siRNA-induced knockdown of frataxin in SH-SY5Y cells as an in vitro cellular model for Friedreich's ataxia. Knockdown of frataxin protein expression to levels detected in patients with the disorder was achieved, leading to decreased cellular viability, increased susceptibility to hydrogen peroxide-induced oxidative stress, dysregulation of key anti-oxidant molecules and deficiencies in both cell proliferation and differentiation. Bone marrow stem cells are being investigated extensively as potential treatments for a wide range of neurological disorders, including Friedreich's ataxia. The potential neuroprotective effects of bone marrow-derived mesenchymal stem cells were therefore studied using our frataxin-deficient cell model. Soluble factors secreted by mesenchymal stem cells protected against cellular changes induced by frataxin deficiency, leading to restoration in frataxin levels and anti-oxidant defences, improved survival against oxidative stress and stimulated both cell proliferation and differentiation down the Schwann cell lineage. The demonstration that mesenchymal stem cell-derived factors can restore cellular homeostasis and function to frataxin-deficient cells further suggests that they may have potential therapeutic benefits for patients with Friedreich's ataxia.
Collapse
Affiliation(s)
- Kevin Kemp
- Multiple Sclerosis and Stem Cell Group, School of Clinical Sciences, Clinical Neurosciences office, University of Bristol, 1st floor, Learning and Research building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Rimi Dey
- Multiple Sclerosis and Stem Cell Group, School of Clinical Sciences, Clinical Neurosciences office, University of Bristol, 1st floor, Learning and Research building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Amelia Cook
- Multiple Sclerosis and Stem Cell Group, School of Clinical Sciences, Clinical Neurosciences office, University of Bristol, 1st floor, Learning and Research building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Neil Scolding
- Multiple Sclerosis and Stem Cell Group, School of Clinical Sciences, Clinical Neurosciences office, University of Bristol, 1st floor, Learning and Research building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Alastair Wilkins
- Multiple Sclerosis and Stem Cell Group, School of Clinical Sciences, Clinical Neurosciences office, University of Bristol, 1st floor, Learning and Research building, Southmead Hospital, Bristol, BS10 5NB, UK
| |
Collapse
|
31
|
Pezzini F, Bettinetti L, Di Leva F, Bianchi M, Zoratti E, Carrozzo R, Santorelli FM, Delledonne M, Lalowski M, Simonati A. Transcriptomic Profiling Discloses Molecular and Cellular Events Related to Neuronal Differentiation in SH-SY5Y Neuroblastoma Cells. Cell Mol Neurobiol 2017; 37:665-682. [PMID: 27422411 DOI: 10.1007/s10571-016-0403-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022]
Abstract
Human SH-SY5Y neuroblastoma cells are widely utilized in in vitro studies to dissect out pathogenetic mechanisms of neurodegenerative disorders. These cells are considered as neuronal precursors and differentiate into more mature neuronal phenotypes under selected growth conditions. In this study, in order to decipher the pathways and cellular processes underlying neuroblastoma cell differentiation in vitro, we performed systematic transcriptomic (RNA-seq) and bioinformatic analysis of SH-SY5Y cells differentiated according to a two-step paradigm: retinoic acid treatment followed by enriched neurobasal medium. Categorization of 1989 differentially expressed genes (DEGs) identified in differentiated cells functionally linked them to changes in cell morphology including remodelling of plasma membrane and cytoskeleton, and neuritogenesis. Seventy-three DEGs were assigned to axonal guidance signalling pathway, and the expression of selected gene products such as neurotrophin receptors, the functionally related SLITRK6, and semaphorins, was validated by immunoblotting. Along with these findings, the differentiated cells exhibited an ability to elongate longer axonal process as assessed by the neuronal cytoskeletal markers biochemical characterization and morphometric evaluation. Recognition of molecular events occurring in differentiated SH-SY5Y cells is critical to accurately interpret the cellular responses to specific stimuli in studies on disease pathogenesis.
Collapse
Affiliation(s)
- Francesco Pezzini
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Laura Bettinetti
- Department of Biotechnologies, University of Verona, Verona, Italy
| | | | - Marzia Bianchi
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Elisa Zoratti
- Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
- Aptuit s.r.l., Verona, Italy
| | - Rosalba Carrozzo
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Filippo M Santorelli
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, IRCCS Stella Maris, Calambrone-Pisa, Italy
| | | | - Maciej Lalowski
- Medicum, Biochemistry/Developmental Biology Meilahti Clinical Proteomics Core Facility, University of Helsinki, Helsinki, Finland.
| | - Alessandro Simonati
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy.
| |
Collapse
|
32
|
Jardin I, Rosado JA. STIM and calcium channel complexes in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1418-26. [DOI: 10.1016/j.bbamcr.2015.10.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/25/2015] [Accepted: 10/07/2015] [Indexed: 12/12/2022]
|
33
|
van de Willige D, Hoogenraad CC, Akhmanova A. Microtubule plus-end tracking proteins in neuronal development. Cell Mol Life Sci 2016; 73:2053-77. [PMID: 26969328 PMCID: PMC4834103 DOI: 10.1007/s00018-016-2168-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/04/2016] [Accepted: 02/22/2016] [Indexed: 11/28/2022]
Abstract
Regulation of the microtubule cytoskeleton is of pivotal importance for neuronal development and function. One such regulatory mechanism centers on microtubule plus-end tracking proteins (+TIPs): structurally and functionally diverse regulatory factors, which can form complex macromolecular assemblies at the growing microtubule plus-ends. +TIPs modulate important properties of microtubules including their dynamics and their ability to control cell polarity, membrane transport and signaling. Several neurodevelopmental and neurodegenerative diseases are associated with mutations in +TIPs or with misregulation of these proteins. In this review, we focus on the role and regulation of +TIPs in neuronal development and associated disorders.
Collapse
Affiliation(s)
- Dieudonnée van de Willige
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Vashisht A, Trebak M, Motiani RK. STIM and Orai proteins as novel targets for cancer therapy. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. Am J Physiol Cell Physiol 2015; 309:C457-69. [PMID: 26017146 DOI: 10.1152/ajpcell.00064.2015] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Calcium (Ca(2+)) regulates a plethora of cellular functions including hallmarks of cancer development such as cell cycle progression and cellular migration. Receptor-regulated calcium rise in nonexcitable cells occurs through store-dependent as well as store-independent Ca(2+) entry pathways. Stromal interaction molecules (STIM) and Orai proteins have been identified as critical constituents of both these Ca(2+) influx pathways. STIMs and Orais have emerged as targets for cancer therapeutics as their altered expression and function have been shown to contribute to tumorigenesis. Recent data demonstrate that they play a vital role in development and metastasis of a variety of tumor types including breast, prostate, cervical, colorectal, brain, and skin tumors. In this review, we will retrospect the data supporting a key role for STIM1, STIM2, Orai1, and Orai3 proteins in tumorigenesis and discuss the potential of targeting these proteins for cancer therapy.
Collapse
Affiliation(s)
- Ayushi Vashisht
- Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; and
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University School of Medicine, Hershey, Pennsylvania
| | - Rajender K Motiani
- Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; and
| |
Collapse
|
35
|
Satheesh NJ, Büsselberg D. The role of intracellular calcium for the development and treatment of neuroblastoma. Cancers (Basel) 2015; 7:823-48. [PMID: 26010602 PMCID: PMC4491686 DOI: 10.3390/cancers7020811] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 05/05/2015] [Indexed: 12/16/2022] Open
Abstract
Neuroblastoma is the second most common paediatric cancer. It developsfrom undifferentiated simpatico-adrenal lineage cells and is mostly sporadic; however, theaetiology behind the development of neuroblastoma is still not fully understood. Intracellularcalcium ([Ca2+]i) is a secondary messenger which regulates numerous cellular processesand, therefore, its concentration is tightly regulated. This review focuses on the role of[Ca2+]i in differentiation, apoptosis and proliferation in neuroblastoma. It describes themechanisms by which [Ca2+]i is regulated and how it modulates intracellular pathways.Furthermore, the importance of [Ca2+]i for the function of anti-cancer drugs is illuminatedin this review as [Ca2+]i could be a target to improve the outcome of anti-cancer treatmentin neuroblastoma. Overall, modulations of [Ca2+]i could be a key target to induce apoptosisin cancer cells leading to a more efficient and effective treatment of neuroblastoma.
Collapse
Affiliation(s)
- Noothan Jyothi Satheesh
- Weill Cornell Medical College in Qatar, Qatar Foundation-Education City, POB 24144, Doha, Qatar.
| | - Dietrich Büsselberg
- Weill Cornell Medical College in Qatar, Qatar Foundation-Education City, POB 24144, Doha, Qatar.
| |
Collapse
|
36
|
da Rocha JF, da Cruz e Silva OAB, Vieira SI. Analysis of the amyloid precursor protein role in neuritogenesis reveals a biphasic SH-SY5Y neuronal cell differentiation model. J Neurochem 2015; 134:288-301. [DOI: 10.1111/jnc.13133] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/17/2015] [Accepted: 04/16/2015] [Indexed: 01/04/2023]
Affiliation(s)
- Joana Fernandes da Rocha
- Laboratório de Neurociências e Sinalização Celular; CBC and iBiMED; Secção Autónoma de Ciências da Saúde; Universidade de Aveiro; Aveiro Portugal
| | - Odete A. B. da Cruz e Silva
- Laboratório de Neurociências e Sinalização Celular; CBC and iBiMED; Secção Autónoma de Ciências da Saúde; Universidade de Aveiro; Aveiro Portugal
| | - Sandra Isabel Vieira
- Laboratório de Neurociências e Sinalização Celular; CBC and iBiMED; Secção Autónoma de Ciências da Saúde; Universidade de Aveiro; Aveiro Portugal
| |
Collapse
|
37
|
Selli C, Erac Y, Kosova B, Erdal ES, Tosun M. Silencing of TRPC1 regulates store-operated calcium entry and proliferation in Huh7 hepatocellular carcinoma cells. Biomed Pharmacother 2015; 71:194-200. [DOI: 10.1016/j.biopha.2015.02.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/21/2015] [Indexed: 01/23/2023] Open
|
38
|
Zui PAN, JianJie MA. Open Sesame: treasure in store-operated calcium entry pathway for cancer therapy. SCIENCE CHINA-LIFE SCIENCES 2014; 58:48-53. [PMID: 25481035 PMCID: PMC4765918 DOI: 10.1007/s11427-014-4774-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 10/21/2014] [Indexed: 11/26/2022]
Abstract
Store-operated Ca2+ entry (SOCE) controls intracellular Ca2+ homeostasis and regulates a wide range of cellular events including proliferation, migration and invasion. The discovery of STIM proteins as Ca2+ sensors and Orai proteins as Ca2+ channel pore forming units provided molecular tools to understand the physiological function of SOCE. Many studies have revealed the pathophysiological roles of Orai and STIM in tumor cells. This review focuses on recent advances in SOCE and its contribution to tumorigenesis. Altered Orai and/or STIM functions may serve as biomarkers for cancer prognosis, and targeting the SOCE pathway may provide a novel means for cancer treatment.
Collapse
Affiliation(s)
- PAN Zui
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Corresponding author (; )
| | - MA JianJie
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Corresponding author (; )
| |
Collapse
|
39
|
Lange I, Koomoa DLT. MycN promotes TRPM7 expression and cell migration in neuroblastoma through a process that involves polyamines. FEBS Open Bio 2014; 4:966-75. [PMID: 25426416 PMCID: PMC4241534 DOI: 10.1016/j.fob.2014.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/13/2014] [Accepted: 10/24/2014] [Indexed: 12/22/2022] Open
Abstract
MycN expression correlates with TRPM7 expression in neuroblastoma (NB) tumors. Expression of the transmembrane protein TRPM7 correlates with lower overall survival in NB tumors. MycN promotes TRPM7 mRNA and protein expression and increases TRPM7 channel activity. TRPM7 regulates NB cell migration. Polyamines regulate TRPM7 expression.
Neuroblastoma is an extra-cranial solid cancer in children. MYCN gene amplification is a prognostic indicator of poor outcome in neuroblastoma. Recent studies have shown that the multiple steps involved in cell migration are dependent on the availability of intracellular calcium (Ca2+). Although significant advances have been made in understanding the role of Ca2+ during migration, little has been achieved towards understanding its impact on the progression of diseases such as cancer. Interestingly, previous studies showed that cancer cell migration is regulated by TRPM7, a calcium-permeable ion channel. The objective of the current study was to elucidate the mechanism by which MycN promotes NB cell migration and the mechanism regulating TRPM7 expression. The results showed that MycN increased TRPM7 expression, induced TRPM7 channel activity, increased intracellular Ca2+ signaling, and promoted cell migration in NB cells. The results also showed that inhibition or down-regulation of ornithine decarboxylase (ODC) inhibited TRPM7 expression, a process that was reversed by spermidine. Overall, this study provides evidence that MycN promotes TRPM7 expression and cell migration through a mechanism that involves ODC synthesis of polyamines.
Collapse
Affiliation(s)
- Ingo Lange
- University of Hawaii at Hilo, The Daniel K. Inouye College of Pharmacy, Hilo, HI 96720, USA
| | - Dana-Lynn T Koomoa
- University of Hawaii at Hilo, The Daniel K. Inouye College of Pharmacy, Hilo, HI 96720, USA
| |
Collapse
|
40
|
Vesprini ND, Dawson TF, Yuan Y, Bruce D, Spencer GE. Retinoic acid affects calcium signaling in adult molluscan neurons. J Neurophysiol 2014; 113:172-81. [PMID: 25343782 DOI: 10.1152/jn.00458.2014] [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] [Indexed: 11/22/2022] Open
Abstract
Retinoic acid, the active metabolite of vitamin A, is important for nervous system development, regeneration, as well as cognitive functions of the adult central nervous system. These central nervous system functions are all highly dependent on neuronal activity. Retinoic acid has previously been shown to induce changes in the firing properties and action potential waveforms of adult molluscan neurons in a dose- and isomer-dependent manner. In this study, we aimed to determine the cellular pathways by which retinoic acid might exert such effects, by testing the involvement of pathways previously shown to be affected by retinoic acid. We demonstrated that the ability of all-trans retinoic acid (atRA) to induce electrophysiological changes in cultured molluscan neurons was not prevented by inhibitors of protein synthesis, protein kinase A or phospholipase C. However, we showed that atRA was capable of rapidly reducing intracellular calcium levels in the same dose- and isomer-dependent manner as shown previously for changes in neuronal firing. Moreover, we also demonstrated that the transmembrane ion flux through voltage-gated calcium channels was rapidly modulated by retinoic acid. In particular, the peak current density was reduced and the inactivation rate was increased in the presence of atRA, over a similar time course as the changes in cell firing and reductions in intracellular calcium. These studies provide further evidence for the ability of atRA to induce rapid effects in mature neurons.
Collapse
Affiliation(s)
- Nicholas D Vesprini
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Taylor F Dawson
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Ye Yuan
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Doug Bruce
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
41
|
Stromal interaction molecules as important therapeutic targets in diseases with dysregulated calcium flux. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2307-14. [DOI: 10.1016/j.bbamcr.2014.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/15/2014] [Accepted: 03/18/2014] [Indexed: 12/29/2022]
|
42
|
Sobradillo D, Hernández-Morales M, Ubierna D, Moyer MP, Núñez L, Villalobos C. A reciprocal shift in transient receptor potential channel 1 (TRPC1) and stromal interaction molecule 2 (STIM2) contributes to Ca2+ remodeling and cancer hallmarks in colorectal carcinoma cells. J Biol Chem 2014; 289:28765-82. [PMID: 25143380 DOI: 10.1074/jbc.m114.581678] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We have investigated the molecular basis of intracellular Ca(2+) handling in human colon carcinoma cells (HT29) versus normal human mucosa cells (NCM460) and its contribution to cancer features. We found that Ca(2+) stores in colon carcinoma cells are partially depleted relative to normal cells. However, resting Ca(2+) levels, agonist-induced Ca(2+) increases, store-operated Ca(2+) entry (SOCE), and store-operated currents (ISOC) are largely enhanced in tumor cells. Enhanced SOCE and depleted Ca(2+) stores correlate with increased cell proliferation, invasion, and survival characteristic of tumor cells. Normal mucosa cells displayed small, inward Ca(2+) release-activated Ca(2+) currents (ICRAC) mediated by ORAI1. In contrast, colon carcinoma cells showed mixed currents composed of enhanced ICRAC plus a nonselective ISOC mediated by TRPC1. Tumor cells display increased expression of TRPC1, ORAI1, ORAI2, ORAI3, and STIM1. In contrast, STIM2 protein was nearly depleted in tumor cells. Silencing data suggest that enhanced ORAI1 and TRPC1 contribute to enhanced SOCE and differential store-operated currents in tumor cells, whereas ORAI2 and -3 are seemingly less important. In addition, STIM2 knockdown decreases SOCE and Ca(2+) store content in normal cells while promoting apoptosis resistance. These data suggest that loss of STIM2 may underlie Ca(2+) store depletion and apoptosis resistance in tumor cells. We conclude that a reciprocal shift in TRPC1 and STIM2 contributes to Ca(2+) remodeling and tumor features in colon cancer.
Collapse
Affiliation(s)
- Diego Sobradillo
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Miriam Hernández-Morales
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Daniel Ubierna
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | | | - Lucía Núñez
- the Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, 47003 Valladolid, Spain
| | - Carlos Villalobos
- From the Institute of Molecular Biology and Genetics (IBGM), Spanish National Research Council (CSIC), 47003 Valladolid, Spain,
| |
Collapse
|
43
|
Stewart TA, Yapa KTDS, Monteith GR. Altered calcium signaling in cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2502-11. [PMID: 25150047 DOI: 10.1016/j.bbamem.2014.08.016] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/11/2014] [Indexed: 01/03/2023]
Abstract
It is the nature of the calcium signal, as determined by the coordinated activity of a suite of calcium channels, pumps, exchangers and binding proteins that ultimately guides a cell's fate. Deregulation of the calcium signal is often deleterious and has been linked to each of the 'cancer hallmarks'. Despite this, we do not yet have a full understanding of the remodeling of the calcium signal associated with cancer. Such an understanding could aid in guiding the development of therapies specifically targeting altered calcium signaling in cancer cells during tumorigenic progression. Findings from some of the studies that have assessed the remodeling of the calcium signal associated with tumorigenesis and/or processes important in invasion and metastasis are presented in this review. The potential of new methodologies is also discussed. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Teneale A Stewart
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Kunsala T D S Yapa
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|