1
|
Lunev EA, Klementieva NV, Vassilieva SG, Volovikov EA, Jappy D, Savchenko IM, Svetlova EA, Polikarpova AV, Shubina MY, Spirin DM, Anufrieva KS, Lebedev PR, Pokrovsky VM, Utkina MV, Krut' VG, Sintsov M, Popov S, Deykin AV, Rozov A, Egorova TV, Bardina MV. Development of an AAV-RNAi strategy to silence the dominant variant GNAO1 c.607G>A linked to encephalopathy. Gene Ther 2025:10.1038/s41434-025-00532-x. [PMID: 40229422 DOI: 10.1038/s41434-025-00532-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 03/24/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Heterozygous mutations in GNAO1 cause an ultra-rare neurodevelopmental disease called GNAO1 encephalopathy, characterized by infantile epilepsy and movement disorder. Here, we provide a functional characterization of the hotspot mutation GNAO1 c.607G>A (p.G203R) and conduct early-phase development of an adeno-associated virus (AAV)-mediated gene therapy approach. The GNAO1 gene encodes the Gαo protein that is involved in neuronal signaling. We showed that the Gαo-G203R lost its ability to enhance forskolin-stimulated cAMP synthesis in HEK293T cells. In primary neuronal culture, Gαo-G203R had a dominant-negative effect on neuronal activity and GABAB-dependent synaptic release. To ablate the mutant protein, we used selective silencing of the pathogenic variant using effectors of RNA interference (RNAi). We selected the short hairpin RNA (sh1500) that suppressed the c.607G>A transcripts, resulting in a 3.8-fold increase in the ratio of wild-type to mutant GNAO1 transcripts in patient-specific neurons. We also detected off-target effects of sh1500 as well as transcriptome changes associated with AAV transduction and RNAi activation. We improved the AAV construct by using an artificial miRNA (miR1500) and the neuron-specific hSyn promoter. Systemic administration of AAV9-hSyn-miR1500 did not cause pathological changes in Gnao1-GGA mice with a "humanized" target sequence. Importantly, AAV9 transduced Gαo-positive neurons in the striatum, thalamus, substantia nigra, and cerebellum, which we defined as primary targets for gene therapy. Our findings pave the road toward the development of AAV-RNAi approaches for dominant-negative GNAO1 variants.
Collapse
Affiliation(s)
- Evgenii A Lunev
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Natalia V Klementieva
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Svetlana G Vassilieva
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Egor A Volovikov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - Irina M Savchenko
- Marlin Biotech, Sochi, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina A Svetlova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Anna V Polikarpova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Maria Y Shubina
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Danil M Spirin
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, Russia
| | - Ksenia S Anufrieva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, Russia
| | - Petr R Lebedev
- Laboratory of Genetic Technologies and Genome Editing for Biomedicine and Animal Health, Joint Center for Genetic Technologies, Belgorod National Research University, Belgorod, Russia
| | - Vladimir M Pokrovsky
- Laboratory of Genetic Technologies and Genome Editing for Biomedicine and Animal Health, Joint Center for Genetic Technologies, Belgorod National Research University, Belgorod, Russia
| | | | - Viktoriya G Krut'
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - Mikhail Sintsov
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | | | - Alexey V Deykin
- Laboratory of Genetic Technologies and Genome Editing for Biomedicine and Animal Health, Joint Center for Genetic Technologies, Belgorod National Research University, Belgorod, Russia
| | - Andrei Rozov
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - Tatiana V Egorova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
- Marlin Biotech, Sochi, Russia
| | - Maryana V Bardina
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia.
- Marlin Biotech, Sochi, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
2
|
Skobeleva K, Wang G, Kaznacheyeva E. STIM Proteins: The Gas and Brake of Calcium Entry in Neurons. Neurosci Bull 2025; 41:305-325. [PMID: 39266936 PMCID: PMC11794855 DOI: 10.1007/s12264-024-01272-5] [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: 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
|
Pikor D, Hurła M, Słowikowski B, Szymanowicz O, Poszwa J, Banaszek N, Drelichowska A, Jagodziński PP, Kozubski W, Dorszewska J. Calcium Ions in the Physiology and Pathology of the Central Nervous System. Int J Mol Sci 2024; 25:13133. [PMID: 39684844 DOI: 10.3390/ijms252313133] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Calcium ions play a key role in the physiological processes of the central nervous system. The intracellular calcium signal, in nerve cells, is part of the neurotransmission mechanism. They are responsible for stabilizing membrane potential and controlling the excitability of neurons. Calcium ions are a universal second messenger that participates in depolarizing signal transduction and contributes to synaptic activity. These ions take an active part in the mechanisms related to memory and learning. As a result of depolarization of the plasma membrane or stimulation of receptors, there is an extracellular influx of calcium ions into the cytosol or mobilization of these cations inside the cell, which increases the concentration of these ions in neurons. The influx of calcium ions into neurons occurs via plasma membrane receptors and voltage-dependent ion channels. Calcium channels play a key role in the functioning of the nervous system, regulating, among others, neuronal depolarization and neurotransmitter release. Channelopathies are groups of diseases resulting from mutations in genes encoding ion channel subunits, observed including the pathophysiology of neurological diseases such as migraine. A disturbed ability of neurons to maintain an appropriate level of calcium ions is also observed in such neurodegenerative processes as Alzheimer's disease, Parkinson's disease, Huntington's disease, and epilepsy. This review focuses on the involvement of calcium ions in physiological and pathological processes of the central nervous system. We also consider the use of calcium ions as a target for pharmacotherapy in the future.
Collapse
Affiliation(s)
- Damian Pikor
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Joanna Poszwa
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Alicja Drelichowska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Paweł P Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
4
|
Maciąg F, Chhikara A, Heine M. Calcium channel signalling at neuronal endoplasmic reticulum-plasma membrane junctions. Biochem Soc Trans 2024; 52:1617-1629. [PMID: 38934485 PMCID: PMC11668288 DOI: 10.1042/bst20230819] [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: 03/27/2024] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Neurons are highly specialised cells that need to relay information over long distances and integrate signals from thousands of synaptic inputs. The complexity of neuronal function is evident in the morphology of their plasma membrane (PM), by far the most intricate of all cell types. Yet, within the neuron lies an organelle whose architecture adds another level to this morphological sophistication - the endoplasmic reticulum (ER). Neuronal ER is abundant in the cell body and extends to distant axonal terminals and postsynaptic dendritic spines. It also adopts specialised structures like the spine apparatus in the postsynapse and the cisternal organelle in the axon initial segment. At membrane contact sites (MCSs) between the ER and the PM, the two membranes come in close proximity to create hubs of lipid exchange and Ca2+ signalling called ER-PM junctions. The development of electron and light microscopy techniques extended our knowledge on the physiological relevance of ER-PM MCSs. Equally important was the identification of ER and PM partners that interact in these junctions, most notably the STIM-ORAI and VAP-Kv2.1 pairs. The physiological functions of ER-PM junctions in neurons are being increasingly explored, but their molecular composition and the role in the dynamics of Ca2+ signalling are less clear. This review aims to outline the current state of research on the topic of neuronal ER-PM contacts. Specifically, we will summarise the involvement of different classes of Ca2+ channels in these junctions, discuss their role in neuronal development and neuropathology and propose directions for further research.
Collapse
Affiliation(s)
- Filip Maciąg
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Arun Chhikara
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| |
Collapse
|
5
|
Oshkolova AA, Grekhnev DA, Kruchinina AA, Belikova LD, Volovikov EA, Lebedeva OS, Bogomazova AN, Vigont VA, Lagarkova MA, Kaznacheyeva EV. Comparison of the calcium signaling alterations in GABA-ergic medium spiny neurons produced from iPSCs of different origins. Biochimie 2024; 222:63-71. [PMID: 38163516 DOI: 10.1016/j.biochi.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
Disease models based on induced pluripotent stem cells (iPSCs) are in high demand because of their physiological adequacy and well-reproducibility of the pathological phenotype. Nowadays, the most common approach to generate iPSCs is the reprogramming of somatic cells using vectors based on lentivirus or Sendai virus. We have previously shown impairments of calcium signaling including store-operated calcium entry in Huntington's disease-specific iPSCs-based GABA-ergic medium spiny neurons. However, different approaches for iPSCs generation make it difficult to compare the models since the mechanism of reprogramming may influence the electrophysiological properties of the terminally differentiated neurons. Here, we have studied the features of calcium homeostasis in GABA-ergic medium spiny neurons differentiated from iPSCs obtained from fibroblasts of the same donor using different methods. Our data demonstrated that there were no significant differences neither in calcium influx through the store-operated channels, nor in the levels of proteins activating this type of calcium entry in neurons differentiated from iPSCs generated with lenti- and Sendai viruses-based approaches. We also found no differences in voltage-gated calcium entry for these neurons. Thus, we clearly showed that various methods of cell reprogramming result in similar deregulations in neuronal calcium signaling which substantiates the ability to combine the experimental data on functional studies of ion channels in models based on iPSCs obtained by different methods and expands the prospects for the use of biobanking.
Collapse
Affiliation(s)
- Arina A Oshkolova
- Institute of Cytology RAS, 194064, Tikhoretsky Ave 4, St. Petersburg, Russia
| | - Dmitriy A Grekhnev
- Institute of Cytology RAS, 194064, Tikhoretsky Ave 4, St. Petersburg, Russia
| | - Anna A Kruchinina
- Institute of Cytology RAS, 194064, Tikhoretsky Ave 4, St. Petersburg, Russia
| | - Lilia D Belikova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, St. Malaya Pirogovskaya, 1a, Moscow, Russia
| | - Egor A Volovikov
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, St. Malaya Pirogovskaya, 1a, Moscow, Russia
| | - Olga S Lebedeva
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, St. Malaya Pirogovskaya, 1a, Moscow, Russia
| | - Alexandra N Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, St. Malaya Pirogovskaya, 1a, Moscow, Russia
| | - Vladimir A Vigont
- Institute of Cytology RAS, 194064, Tikhoretsky Ave 4, St. Petersburg, Russia
| | - Maria A Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, St. Malaya Pirogovskaya, 1a, Moscow, Russia
| | | |
Collapse
|
6
|
Djordjevic S, Itzykson R, Hague F, Lebon D, Legrand J, Ouled‐Haddou H, Jedraszak G, Harbonnier J, Collet L, Paubelle E, Marolleau J, Garçon L, Boyer T. STIM2 is involved in the regulation of apoptosis and the cell cycle in normal and malignant monocytic cells. Mol Oncol 2024; 18:1571-1592. [PMID: 38234211 PMCID: PMC11161727 DOI: 10.1002/1878-0261.13584] [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: 09/27/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Calcium is a ubiquitous messenger that regulates a wide range of cellular functions, but its involvement in the pathophysiology of acute myeloid leukemia (AML) is not widely investigated. Here, we identified, from an analysis of The Cancer Genome Atlas and genotype-tissue expression databases, stromal interaction molecule 2 (STIM2) as being highly expressed in AML with monocytic differentiation and negatively correlated with overall survival. This was confirmed on a validation cohort of 407 AML patients. We then investigated the role of STIM2 in cell proliferation, differentiation, and survival in two leukemic cell lines with monocytic potential and in normal hematopoietic stem cells. STIM2 expression increased at the RNA and protein levels upon monocyte differentiation. Phenotypically, STIM2 knockdown drastically inhibited cell proliferation and induced genomic stress with DNA double-strand breaks, as shown by increased levels of phosphorylate histone H2AXγ (p-H2AXγ), followed by activation of the cellular tumor antigen p53 pathway, decreased expression of cell cycle regulators such as cyclin-dependent kinase 1 (CDK1)-cyclin B1 and M-phase inducer phosphatase 3 (CDC25c), and a decreased apoptosis threshold with a low antiapoptotic/proapoptotic protein ratio. Our study reports STIM2 as a new actor regulating genomic stability and p53 response in terms of cell cycle and apoptosis of human normal and malignant monocytic cells.
Collapse
Affiliation(s)
| | - Raphaël Itzykson
- Département Hématologie et ImmunologieHôpital Saint‐Louis, Assistance Publique‐Hôpitaux de ParisFrance
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRSUniversité Paris CitéFrance
| | - Frédéric Hague
- Laboratoire de Physiologie Cellulaire et Moléculaire UR4667Université Picardie Jules VerneAmiensFrance
| | - Delphine Lebon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Julien Legrand
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | | | - Guillaume Jedraszak
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Laboratoire de Génétique ConstitutionnelleCHU Amiens‐PicardieFrance
| | | | - Louison Collet
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | - Etienne Paubelle
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Jean‐Pierre Marolleau
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Loïc Garçon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| | - Thomas Boyer
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| |
Collapse
|
7
|
Luo R, Gourriérec PL, Antigny F, Bedouet K, Domenichini S, Gomez AM, Benitah JP, Sabourin J. STIM2 variants regulate Orai1/TRPC1/TRPC4-mediated store-operated Ca 2+ entry and mitochondrial Ca 2+ homeostasis in cardiomyocytes. Cell Calcium 2024; 119:102871. [PMID: 38537434 DOI: 10.1016/j.ceca.2024.102871] [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: 10/22/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
The stromal interaction molecules (STIMs) are the sarcoplasmic reticulum (SR) Ca2+ sensors that trigger store-operated Ca2+ entry (SOCE) in a variety of cell types. While STIM1 isoform has been the focus of the research in cardiac pathophysiology, the function of the homolog STIM2 remains unknown. Using Ca2+ imaging and patch-clamp techniques, we showed that knockdown (KD) of STIM2 by siRNAs increased SOCE and the ISOC current in neonatal rat ventricular cardiomyocytes (NRVMs). Within this cardiomyocyte model, we identified the transcript expression of Stim2.1 and Stim2.2 splice variants, with predominance for Stim2.2. Using conventional and super-resolution confocal microscopy (STED), we found that exogenous STIM2.1 and STIM2.2 formed pre-clusters with a reticular organization at rest. Following SR Ca2+ store depletion, some STIM2.1 and STIM2.2 clusters were translocated to SR-plasma membrane (PM) junctions and co-localized with Orai1. The overexpression strategy revealed that STIM2.1 suppressed Orai1-mediated SOCE and the ISOC current while STIM2.2 enhanced SOCE. STIM2.2-enhanced SOCE was also dependent on TRPC1 and TRPC4. Even if STIM2 KD or splice variants overexpression did not affect cytosolic Ca2+ cycling, we observed, using Rhod-2/AM Ca2+ imaging, that Orai1 inhibition or STIM2.1 overexpression abolished the mitochondrial Ca2+ (mCa2+) uptake, as opposed to STIM2 KD. We also found that STIM2 was present in the mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) by interacting with the inositol trisphosphate receptors (IP3Rs), voltage-dependent anion channel (VDAC), mitochondrial Ca2+ uniporter (MCU), and mitofusin-2 (MNF2). Our results suggested that, in NRVMs, STIM2.1 constitutes the predominant functional variant that negatively regulates Orai1-generated SOCE. It participates in the control of mCa2+ uptake capacity possibly via the STIM2-IP3Rs-VDAC-MCU and MNF2 complex.
Collapse
Affiliation(s)
- Rui Luo
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, 91400 Orsay, France
| | - Pauline Le Gourriérec
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, 91400 Orsay, France
| | - Fabrice Antigny
- Inserm, UMR-S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Kaveen Bedouet
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, 91400 Orsay, France
| | - Séverine Domenichini
- Université Paris-Saclay, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique-Plateforme MIPSIT, Orsay, France
| | - Ana-Maria Gomez
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, 91400 Orsay, France
| | - Jean-Pierre Benitah
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, 91400 Orsay, France
| | - Jessica Sabourin
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
8
|
Courjaret R, Prakriya M, Machaca K. SOCE as a regulator of neuronal activity. J Physiol 2024; 602:1449-1462. [PMID: 37029630 PMCID: PMC11963908 DOI: 10.1113/jp283826] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023] Open
Abstract
Store operated Ca2+ entry (SOCE) is a ubiquitous signalling module with established roles in the immune system, secretion and muscle development. Recent evidence supports a complex role for SOCE in the nervous system. In this review we present an update of the current knowledge on SOCE function in the brain with a focus on its role as a regulator of brain activity and excitability.
Collapse
Affiliation(s)
- Raphael Courjaret
- Calcium Signaling Group, Research Department, Weill Cornell Medicine Qatar, Qatar Foundation, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Khaled Machaca
- Calcium Signaling Group, Research Department, Weill Cornell Medicine Qatar, Qatar Foundation, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
9
|
Benedetti MC, D'andrea T, Colantoni A, Silachev D, de Turris V, Boussadia Z, Babenko VA, Volovikov EA, Belikova L, Bogomazova AN, Pepponi R, Whye D, Buttermore ED, Tartaglia GG, Lagarkova MA, Katanaev VL, Musayev I, Martinelli S, Fucile S, Rosa A. Cortical neurons obtained from patient-derived iPSCs with GNAO1 p.G203R variant show altered differentiation and functional properties. Heliyon 2024; 10:e26656. [PMID: 38434323 PMCID: PMC10907651 DOI: 10.1016/j.heliyon.2024.e26656] [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: 08/03/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Pathogenic variants in the GNAO1 gene, encoding the alpha subunit of an inhibitory heterotrimeric guanine nucleotide-binding protein (Go) highly expressed in the mammalian brain, have been linked to encephalopathy characterized by different combinations of neurological symptoms, including developmental delay, hypotonia, epilepsy and hyperkinetic movement disorder with life-threatening paroxysmal exacerbations. Currently, there are only symptomatic treatments, and little is known about the pathophysiology of GNAO1-related disorders. Here, we report the characterization of a new in vitro model system based on patient-derived induced pluripotent stem cells (hiPSCs) carrying the recurrent p.G203R amino acid substitution in Gαo, and a CRISPR-Cas9-genetically corrected isogenic control line. RNA-Seq analysis highlighted aberrant cell fate commitment in neuronal progenitor cells carrying the p.G203R pathogenic variant. Upon differentiation into cortical neurons, patients' cells showed reduced expression of early neural genes and increased expression of astrocyte markers, as well as premature and defective differentiation processes leading to aberrant formation of neuronal rosettes. Of note, comparable defects in gene expression and in the morphology of neural rosettes were observed in hiPSCs from an unrelated individual harboring the same GNAO1 variant. Functional characterization showed lower basal intracellular free calcium concentration ([Ca2+]i), reduced frequency of spontaneous activity, and a smaller response to several neurotransmitters in 40- and 50-days differentiated p.G203R neurons compared to control cells. These findings suggest that the GNAO1 pathogenic variant causes a neurodevelopmental phenotype characterized by aberrant differentiation of both neuronal and glial populations leading to a significant alteration of neuronal communication and signal transduction.
Collapse
Affiliation(s)
- Maria Cristina Benedetti
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Tiziano D'andrea
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Alessio Colantoni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Denis Silachev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690090, Vladivostok, Russia
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Valeria de Turris
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Zaira Boussadia
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina A. Babenko
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | - Egor A. Volovikov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Lilia Belikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Alexandra N. Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Rita Pepponi
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center and F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Elizabeth D. Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center and F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Gian Gaetano Tartaglia
- Center for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Maria A. Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435, Moscow, Russia
| | - Vladimir L. Katanaev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690090, Vladivostok, Russia
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva, 1211, Geneva, Switzerland
| | | | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sergio Fucile
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Alessandro Rosa
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| |
Collapse
|
10
|
Novikova IV, Grekhnev DA, Oshkolova A, Nomerovskaya MA, Kolesnikov DO, Krisanova AV, Yuskovets VN, Chernov NM, Yakovlev IP, Kaznacheyeva EV, Vigont VA. 1,2,3,4-dithiadiazole derivatives as a novel class of calcium signaling modulators. Biochem Biophys Res Commun 2024; 691:149333. [PMID: 38043197 DOI: 10.1016/j.bbrc.2023.149333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/08/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
Aberrant calcium signaling is associated with a diverse range of pathologies, including cardiovascular and neurodegenerative diseases, diabetes, cancer, etc… So, therapeutic strategies based on the correction of pathological calcium signaling are becoming extremely in demand. Thus, the development of novel calcium signaling modulators remains highly actual. Previously we found that 1,2,3,4-dithiadiazole derivative 3-(4-nitrophenyl)-5-phenyl-3H-1,2,3,4-dithiadiazole-2-oxide can strongly reduce calcium uptake through store-operated calcium (SOC) channels. Here we tested several structurally related compounds and found that most of them can effectively affect SOC channels and attenuate calcium content in the endoplasmic reticulum, thus, establishing 1,2,3,4-dithiadiazoles as a novel class of SOC channel inhibitors. Comparing different 1,2,3,4-dithiadiazole derivatives we showed that previously published 3-(4-nitrophenyl)-5-phenyl-3H-1,2,3,4-dithiadiazole-2-oxide and newly tested 3-(3,5-difluorophenyl)-5-phenyl-3H-1,2,3,4-dithiadiazole 2-oxide demonstrated the highest efficacy of SOC entry reduction, supposing the important role of electron-withdrawing substituents to realize the inhibitory activity of 1,2,3,4-dithiadiazoles.
Collapse
Affiliation(s)
- Iuliia V Novikova
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation
| | - Dmitriy A Grekhnev
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation
| | - Arina Oshkolova
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation
| | - Maria A Nomerovskaya
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation
| | - Dmitrii O Kolesnikov
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation
| | - Alena V Krisanova
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation
| | - Valeriy N Yuskovets
- Organic Chemistry Department, Saint-Petersburg State Chemical Pharmaceutical University, Prof. Popov st. 14, Saint-Petersburg, 197376, Russian Federation
| | - Nikita M Chernov
- Organic Chemistry Department, Saint-Petersburg State Chemical Pharmaceutical University, Prof. Popov st. 14, Saint-Petersburg, 197376, Russian Federation
| | - Igor P Yakovlev
- Organic Chemistry Department, Saint-Petersburg State Chemical Pharmaceutical University, Prof. Popov st. 14, Saint-Petersburg, 197376, Russian Federation
| | - Elena V Kaznacheyeva
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation
| | - Vladimir A Vigont
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russian Federation.
| |
Collapse
|
11
|
Lebedeva OS, Sharova EI, Grekhnev DA, Skorodumova LO, Kopylova IV, Vassina EM, Oshkolova A, Novikova IV, Krisanova AV, Olekhnovich EI, Vigont VA, Kaznacheyeva EV, Bogomazova AN, Lagarkova MA. An Efficient 2D Protocol for Differentiation of iPSCs into Mature Postmitotic Dopaminergic Neurons: Application for Modeling Parkinson's Disease. Int J Mol Sci 2023; 24:7297. [PMID: 37108456 PMCID: PMC10139404 DOI: 10.3390/ijms24087297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
About 15% of patients with parkinsonism have a hereditary form of Parkinson's disease (PD). Studies on the early stages of PD pathogenesis are challenging due to the lack of relevant models. The most promising ones are models based on dopaminergic neurons (DAns) differentiated from induced pluripotent stem cells (iPSCs) of patients with hereditary forms of PD. This work describes a highly efficient 2D protocol for obtaining DAns from iPSCs. The protocol is rather simple, comparable in efficiency with previously published protocols, and does not require viral vectors. The resulting neurons have a similar transcriptome profile to previously published data for neurons, and have a high level of maturity marker expression. The proportion of sensitive (SOX6+) DAns in the population calculated from the level of gene expression is higher than resistant (CALB+) DAns. Electrophysiological studies of the DAns confirmed their voltage sensitivity and showed that a mutation in the PARK8 gene is associated with enhanced store-operated calcium entry. The study of high-purity DAns differentiated from the iPSCs of patients with hereditary PD using this differentiation protocol will allow for investigators to combine various research methods, from patch clamp to omics technologies, and maximize information about cell function in normal and pathological conditions.
Collapse
Affiliation(s)
- Olga S. Lebedeva
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Elena I. Sharova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Dmitriy A. Grekhnev
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Liubov O. Skorodumova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Irina V. Kopylova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Ekaterina M. Vassina
- Vavilov Institute of General Genetics, GSP-1, Gubkina St., 3, 119991 Moscow, Russia
| | - Arina Oshkolova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Iuliia V. Novikova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Alena V. Krisanova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Evgenii I. Olekhnovich
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Vladimir A. Vigont
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Elena V. Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 St. Petersburg, Russia
| | - Alexandra N. Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| | - Maria A. Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, St. Malaya Pirogovskaya, 1a, 119435 Moscow, Russia
| |
Collapse
|
12
|
Bogomiakova ME, Sekretova EK, Anufrieva KS, Khabarova PO, Kazakova AN, Bobrovsky PA, Grigoryeva TV, Eremeev AV, Lebedeva OS, Bogomazova AN, Lagarkova MA. iPSC-derived cells lack immune tolerance to autologous NK-cells due to imbalance in ligands for activating and inhibitory NK-cell receptors. Stem Cell Res Ther 2023; 14:77. [PMID: 37038186 PMCID: PMC10088155 DOI: 10.1186/s13287-023-03308-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/28/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Dozens of transplants generated from pluripotent stem cells are currently in clinical trials. The creation of patient-specific iPSCs makes personalized therapy possible due to their main advantage of immunotolerance. However, some reports have claimed recently that aberrant gene expression followed by proteome alterations and neoantigen formation can result in iPSCs recognition by autologous T-cells. Meanwhile, the possibility of NK-cell activation has not been previously considered. This study focused on the comparison of autologous and allogeneic immune response to iPSC-derived cells and isogeneic parental somatic cells used for reprogramming. METHODS We established an isogeneic cell model consisting of parental dermal fibroblasts, fibroblast-like iPSC-derivatives (iPS-fibro) and iPS-fibro lacking beta-2-microglobulin (B2M). Using the cells obtained from two patients, we analyzed the activation of autologous and allogeneic T-lymphocytes and NK-cells co-cultured with target cells. RESULTS Here we report that cells differentiated from iPSCs can be recognized by NK-cells rather than by autologous T-cells. We observed that iPS-fibro elicited a high level of NK-cell degranulation and cytotoxicity, while isogeneic parental skin fibroblasts used to obtain iPSCs barely triggered an NK-cell response. iPSC-derivatives with B2M knockout did not cause an additional increase in NK-cell activation, although they were devoid of HLA-I, the major inhibitory molecules for NK-cells. Transcriptome analysis revealed a significant imbalance of ligands for activating and inhibitory NK-cell receptors in iPS-fibro. Compared to parental fibroblasts, iPSC-derivatives had a reduced expression of HLA-I simultaneously with an increased gene expression of major activating ligands, such as MICA, NECTIN2, and PVR. The lack of inhibitory signals might be due to insufficient maturity of cells differentiated from iPSCs. In addition, we showed that pretreatment of iPS-fibro with proinflammatory cytokine IFNγ restored the ligand imbalance, thereby reducing the degranulation and cytotoxicity of NK-cells. CONCLUSION In summary, we showed that iPSC-derived cells can be sensitive to the cytotoxic potential of autologous NK-cells regardless of HLA-I status. Thus, the balance of ligands for NK-cell receptors should be considered prior to iPSC-based cell therapies. Trial registration Not applicable.
Collapse
Affiliation(s)
- Margarita E Bogomiakova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435.
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, Russia, 119991.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435.
| | - Elizaveta K Sekretova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, Russia, 119991
| | - Ksenia S Anufrieva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
| | - Polina O Khabarova
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, Russia, 119991
| | - Anastasia N Kazakova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
| | - Pavel A Bobrovsky
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
| | | | - Artem V Eremeev
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
| | - Olga S Lebedeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
| | - Alexandra N Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya, Moscow, Russia, 119435
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, Russia, 119991
| |
Collapse
|
13
|
Correa BH, Moreira CR, Hildebrand ME, Vieira LB. The Role of Voltage-Gated Calcium Channels in Basal Ganglia Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:183-201. [PMID: 35339179 PMCID: PMC10190140 DOI: 10.2174/1570159x20666220327211156] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/11/2022] [Accepted: 03/14/2022] [Indexed: 11/22/2022] Open
Abstract
Calcium (Ca2+) plays a central role in regulating many cellular processes and influences cell survival. Several mechanisms can disrupt Ca2+ homeostasis to trigger cell death, including oxidative stress, mitochondrial damage, excitotoxicity, neuroinflammation, autophagy, and apoptosis. Voltage-gated Ca2+ channels (VGCCs) act as the main source of Ca2+ entry into electrically excitable cells, such as neurons, and they are also expressed in glial cells such as astrocytes and oligodendrocytes. The dysregulation of VGCC activity has been reported in both Parkinson's disease (PD) and Huntington's (HD). PD and HD are progressive neurodegenerative disorders (NDs) of the basal ganglia characterized by motor impairment as well as cognitive and psychiatric dysfunctions. This review will examine the putative role of neuronal VGCCs in the pathogenesis and treatment of central movement disorders, focusing on PD and HD. The link between basal ganglia disorders and VGCC physiology will provide a framework for understanding the neurodegenerative processes that occur in PD and HD, as well as a possible path towards identifying new therapeutic targets for the treatment of these debilitating disorders.
Collapse
Affiliation(s)
- Bernardo H.M. Correa
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlos Roberto Moreira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
14
|
Grekhnev DA, Kruchinina AA, Vigont VA, Kaznacheyeva EV. The Mystery of EVP4593: Perspectives of the Quinazoline-Derived Compound in the Treatment of Huntington's Disease and Other Human Pathologies. Int J Mol Sci 2022; 23:15724. [PMID: 36555369 PMCID: PMC9778905 DOI: 10.3390/ijms232415724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Quinazoline derivatives have various pharmacological activities and are widely used in clinical practice. Here, we reviewed the proposed mechanisms of the physiological activity of the quinazoline derivative EVP4593 and perspectives for its clinical implication. We summarized the accumulated data about EVP4593 and focused on its activities in different models of Huntington's disease (HD), including patient-specific iPSCs-based neurons. To make a deeper insight into its neuroprotective role in HD treatment, we discussed the ability of EVP4593 to modulate calcium signaling and reduce the level of the huntingtin protein. Moreover, we described possible protective effects of EVP4593 in other pathologies, such as oncology, cardiovascular diseases and parasite invasion. We hope that comprehensive analyses of the molecular mechanisms of EVP4593 activity will allow for the expansion of the scope of the EVP4593 application.
Collapse
|
15
|
The STIM1/2-Regulated Calcium Homeostasis Is Impaired in Hippocampal Neurons of the 5xFAD Mouse Model of Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms232314810. [PMID: 36499137 PMCID: PMC9738900 DOI: 10.3390/ijms232314810] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of age-related dementia. Neuronal calcium homeostasis impairment may contribute to AD. Here we demonstrated that voltage-gated calcium (VGC) entry and store-operated calcium (SOC) entry regulated by calcium sensors of intracellular calcium stores STIM proteins are affected in hippocampal neurons of the 5xFAD transgenic mouse model. We observed excessive SOC entry in 5xFAD mouse neurons, mediated by STIM1 and STIM2 proteins with increased STIM1 contribution. There were no significant changes in cytoplasmic calcium level, endoplasmic reticulum (ER) bulk calcium levels, or expression levels of STIM1 or STIM2 proteins. The potent inhibitor BTP-2 and the FDA-approved drug leflunomide reduced SOC entry in 5xFAD neurons. In turn, excessive voltage-gated calcium entry was sensitive to the inhibitor of L-type calcium channels nifedipine but not to the T-type channels inhibitor ML218. Interestingly, the depolarization-induced calcium entry mediated by VGC channels in 5xFAD neurons was dependent on STIM2 but not STIM1 protein in cells with replete Ca2+ stores. The result gives new evidence on the VGC channel modulation by STIM2. Overall, the data demonstrate the changes in calcium signaling of hippocampal neurons of the AD mouse model, which precede amyloid plaque accumulation or other signs of pathology manifestation.
Collapse
|
16
|
Grekhnev DA, Novikova IV, Krisanova AV, Yuskovets VN, Chernov NM, Yakovlev IP, Kaznacheyeva EV, Vigont VA. Dithiadiazole derivative 3-(4-nitrophenyl)-5-phenyl-3H-1,2,3,4-dithiadiazole-2-oxide - Novel modulator of store-operated calcium entry. Biochem Biophys Res Commun 2022; 626:38-43. [PMID: 35981420 DOI: 10.1016/j.bbrc.2022.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 11/19/2022]
Abstract
Pathological calcium homeostasis accompanies the development of a large number of different diseases, therefore, the search for new modulators of calcium signaling remains highly actual. Last decades store-operated calcium channels have been repeatedly postulated as a therapeutic target, so the compounds acting on them can be considered promising drug prototypes. Here, we tested several derivatives of 1,2,3,4-dithiadiazole, 1,3-thiazine, pyrazolopyrimidine and thiohydrazides for the ability to affect the thapsigargin-induced calcium response. Using calcium imaging and the patch-clamp technique we found that dithiadiazole derivative3-(4-nitrophenyl)-5-phenyl-3H-1,2,3,4-dithiadiazole-2-oxidehad a strong inhibitory effect on store-operated calcium entry at the micromolar concentration in HEK293 cells. Moreover, incubation of the cells with this compound also resulted in the decrease of ER calcium content. Thus, we have postulated 3-(4-nitrophenyl)-5-phenyl-3H-1,2,3,4-dithiadiazole-2-oxide as a novel inhibitor of store-operated calcium entry and suggested the derivatives of 1,2,3,4-dithiadiazole as a prospective class of compounds for searching new calcium modulators.
Collapse
Affiliation(s)
- Dmitriy A Grekhnev
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russia
| | - Iuliia V Novikova
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russia
| | - Alena V Krisanova
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russia
| | - Valeriy N Yuskovets
- Organic Chemistry Department, Saint-Petersburg State Chemical Pharmaceutical University, Prof. Popov st. 14, Saint-Petersburg, 197376, Russia
| | - Nikita M Chernov
- Organic Chemistry Department, Saint-Petersburg State Chemical Pharmaceutical University, Prof. Popov st. 14, Saint-Petersburg, 197376, Russia
| | - Igor P Yakovlev
- Organic Chemistry Department, Saint-Petersburg State Chemical Pharmaceutical University, Prof. Popov st. 14, Saint-Petersburg, 197376, Russia
| | | | - Vladimir A Vigont
- Institute of Cytology RAS, Tikhoretsky Ave. 4, St. Petersburg, 194064, Russia.
| |
Collapse
|
17
|
Chen X, Sun Y, Chen L, Chen XS, Pan M, Zhang Y, Wang Q, Yang W, Yin P, He D, Guo X, Yang S, Zeng Y, Yan S, Li XJ, Li S. Differential expression and roles of Huntingtin and Huntingtin-associated protein 1 in the mouse and primate brains. Cell Mol Life Sci 2022; 79:554. [PMID: 36251080 PMCID: PMC11802989 DOI: 10.1007/s00018-022-04577-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/12/2022] [Accepted: 09/27/2022] [Indexed: 11/03/2022]
Abstract
Huntingtin-associated protein 1 (HAP1) is the first identified protein whose function is affected by its abnormal interaction with mutant huntingtin (mHTT), which causes Huntington disease. However, the expression patterns of Hap1 and Htt in the rodent brain are not correlated. Here we found that the primate HAP1, unlike the rodent Hap1, is correlatively expressed with HTT in the primate brains. CRISPR/Cas9 targeting revealed that HAP1 deficiency in the developing human neurons did not affect neuronal differentiation and gene expression as seen in the mouse neurons. However, deletion of HAP1 exacerbated neurotoxicity of mutant HTT in the organotypic brain slices of adult monkeys. These findings demonstrate differential HAP1 expression and function in the mouse and primate brains, and suggest that interaction of HAP1 with mutant HTT may be involved in mutant HTT-mediated neurotoxicity in adult primate neurons.
Collapse
Affiliation(s)
- Xingxing Chen
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, 430000, Hubei, China
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yize Sun
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Laiqiang Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiu-Sheng Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Mingtian Pan
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yiran Zhang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Qi Wang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Weili Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Peng Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Dajian He
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiangyu Guo
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Su Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
18
|
Kaye J, Reisine T, Finkbeiner S. Huntington's disease iPSC models-using human patient cells to understand the pathology caused by expanded CAG repeats. Fac Rev 2022; 11:16. [PMID: 35865413 PMCID: PMC9264339 DOI: 10.12703/r/11-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
A major advance in the study of Huntington's disease (HD) has been the development of human disease models employing induced pluripotent stem cells (iPSCs) derived from patients with HD. Because iPSCs provide an unlimited source of cells and can be obtained from large numbers of HD patients, they are a uniquely valuable tool for investigating disease mechanisms and for discovering potential disease-modifying therapeutics. Here, we summarize some of the important findings in HD pathophysiology that have emerged from studies of patient-derived iPSC lines. Because they retain the genome and actual disease mutations of the patient, they provide a cell source to investigate genetic contributions to the disease. iPSCs provide advantages over other disease models. While iPSC-based technology erases some epigenetic marks, newly developed transdifferentiation methods now let us investigate epigenetic factors that control expression of mutant huntingtin (mHTT). Human HD iPSC lines allow us to investigate how endogenous levels of mHTT affect cell health, in contrast to other models that often rely on overexpressing the protein. iPSCs can be differentiated into neurons and other disease-related cells such as astrocytes from different brain regions to study brain regional differences in the disease process, as well as the cell-cell dependencies involved in HD-associated neurodegeneration. They also serve as a tissue source to investigate factors that impact CAG repeat instability, which is involved in regional differences in neurodegeneration in the HD brain. Human iPSC models can serve as a powerful model system to identify genetic modifiers that may impact disease onset, progression, and symptomatology, providing novel molecular targets for drug discovery.
Collapse
Affiliation(s)
- Julia Kaye
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, CA, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA, USA
- Department of Neurology and Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
19
|
Collins HE, Zhang D, Chatham JC. STIM and Orai Mediated Regulation of Calcium Signaling in Age-Related Diseases. FRONTIERS IN AGING 2022; 3:876785. [PMID: 35821821 PMCID: PMC9261457 DOI: 10.3389/fragi.2022.876785] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/30/2022] [Indexed: 01/19/2023]
Abstract
Tight spatiotemporal regulation of intracellular Ca2+ plays a critical role in regulating diverse cellular functions including cell survival, metabolism, and transcription. As a result, eukaryotic cells have developed a wide variety of mechanisms for controlling Ca2+ influx and efflux across the plasma membrane as well as Ca2+ release and uptake from intracellular stores. The STIM and Orai protein families comprising of STIM1, STIM2, Orai1, Orai2, and Orai3, are evolutionarily highly conserved proteins that are core components of all mammalian Ca2+ signaling systems. STIM1 and Orai1 are considered key players in the regulation of Store Operated Calcium Entry (SOCE), where release of Ca2+ from intracellular stores such as the Endoplasmic/Sarcoplasmic reticulum (ER/SR) triggers Ca2+ influx across the plasma membrane. SOCE, which has been widely characterized in non-excitable cells, plays a central role in Ca2+-dependent transcriptional regulation. In addition to their role in Ca2+ signaling, STIM1 and Orai1 have been shown to contribute to the regulation of metabolism and mitochondrial function. STIM and Orai proteins are also subject to redox modifications, which influence their activities. Considering their ubiquitous expression, there has been increasing interest in the roles of STIM and Orai proteins in excitable cells such as neurons and myocytes. While controversy remains as to the importance of SOCE in excitable cells, STIM1 and Orai1 are essential for cellular homeostasis and their disruption is linked to various diseases associated with aging such as cardiovascular disease and neurodegeneration. The recent identification of splice variants for most STIM and Orai isoforms while complicating our understanding of their function, may also provide insight into some of the current contradictions on their roles. Therefore, the goal of this review is to describe our current understanding of the molecular regulation of STIM and Orai proteins and their roles in normal physiology and diseases of aging, with a particular focus on heart disease and neurodegeneration.
Collapse
Affiliation(s)
- Helen E. Collins
- Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Dingguo Zhang
- Division of Molecular and Cellular Pathology, Department of PathologyUniversity of Alabama at Birmingham, Birmingham, AL, United States
| | - John C. Chatham
- Division of Molecular and Cellular Pathology, Department of PathologyUniversity of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: John C. Chatham,
| |
Collapse
|
20
|
Grekhnev DA, Kaznacheyeva EV, Vigont VA. Patient-Specific iPSCs-Based Models of Neurodegenerative Diseases: Focus on Aberrant Calcium Signaling. Int J Mol Sci 2022; 23:624. [PMID: 35054808 PMCID: PMC8776084 DOI: 10.3390/ijms23020624] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
The development of cell reprogramming technologies became a breakthrough in the creation of new models of human diseases, including neurodegenerative pathologies. The iPSCs-based models allow for the studying of both hereditary and sporadic cases of pathologies and produce deep insight into the molecular mechanisms underlying neurodegeneration. The use of the cells most vulnerable to a particular pathology makes it possible to identify specific pathological mechanisms and greatly facilitates the task of selecting the most effective drugs. To date, a large number of studies on patient-specific models of neurodegenerative diseases has been accumulated. In this review, we focused on the alterations of such a ubiquitous and important intracellular regulatory pathway as calcium signaling. Here, we reviewed and analyzed the data obtained from iPSCs-based models of different neurodegenerative disorders that demonstrated aberrant calcium signaling.
Collapse
Affiliation(s)
| | | | - Vladimir A. Vigont
- Laboratory of Ionic Channels of Cell Membranes, Department of Molecular Physiology of the Cell, Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave., 194064 St. Petersburg, Russia; (D.A.G.); (E.V.K.)
| |
Collapse
|
21
|
Monk R, Connor B. Cell Reprogramming to Model Huntington's Disease: A Comprehensive Review. Cells 2021; 10:cells10071565. [PMID: 34206228 PMCID: PMC8306243 DOI: 10.3390/cells10071565] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder characterized by the progressive decline of motor, cognitive, and psychiatric functions. HD results from an autosomal dominant mutation that causes a trinucleotide CAG repeat expansion and the production of mutant Huntingtin protein (mHTT). This results in the initial selective and progressive loss of medium spiny neurons (MSNs) in the striatum before progressing to involve the whole brain. There are currently no effective treatments to prevent or delay the progression of HD as knowledge into the mechanisms driving the selective degeneration of MSNs has been hindered by a lack of access to live neurons from individuals with HD. The invention of cell reprogramming provides a revolutionary technique for the study, and potential treatment, of neurological conditions. Cell reprogramming technologies allow for the generation of live disease-affected neurons from patients with neurological conditions, becoming a primary technique for modelling these conditions in vitro. The ability to generate HD-affected neurons has widespread applications for investigating the pathogenesis of HD, the identification of new therapeutic targets, and for high-throughput drug screening. Cell reprogramming also offers a potential autologous source of cells for HD cell replacement therapy. This review provides a comprehensive analysis of the use of cell reprogramming to model HD and a discussion on recent advancements in cell reprogramming technologies that will benefit the HD field.
Collapse
|
22
|
Zhu X, Zhang Y, Yang X, Hao C, Duan H. Gene Therapy for Neurodegenerative Disease: Clinical Potential and Directions. Front Mol Neurosci 2021; 14:618171. [PMID: 34194298 PMCID: PMC8236824 DOI: 10.3389/fnmol.2021.618171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/07/2021] [Indexed: 12/21/2022] Open
Abstract
The pathogenesis of neurodegenerative diseases (NDDs) is complex and diverse. Over the decades, our understanding of NDD has been limited to pathological features. However, recent advances in gene sequencing have facilitated elucidation of NDD at a deeper level. Gene editing techniques have uncovered new genetic links to phenotypes, promoted the development of novel treatment strategies and equipped researchers with further means to construct effective cell and animal models. The current review describes the history of evolution of gene editing tools, with the aim of improving overall understanding of this technology, and focuses on the four most common NDD disorders to demonstrate the potential future applications and research directions of gene editing.
Collapse
Affiliation(s)
- Xiaolin Zhu
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yu Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xin Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunyan Hao
- Department of Geriatrics, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hubin Duan
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China.,Department of Neurosurgery, Lvliang People's Hospital, Lvliang, China
| |
Collapse
|
23
|
Joshi T, Kumar V, Kaznacheyeva EV, Jana NR. Withaferin A Induces Heat Shock Response and Ameliorates Disease Progression in a Mouse Model of Huntington's Disease. Mol Neurobiol 2021; 58:3992-4006. [PMID: 33904021 DOI: 10.1007/s12035-021-02397-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Impairment of proteostasis network is one of the characteristic features of many age-related neurodegenerative disorders including autosomal dominantly inherited Huntington's disease (HD). In HD, N-terminal portion of mutant huntingtin protein containing expanded polyglutamine repeats accumulates as inclusion bodies and leads to progressive deterioration of various cellular functioning including proteostasis network. Here we report that Withaferin A (a small bioactive molecule derived from Indian medicinal plant, Withania somnifera) partially rescues defective proteostasis by activating heat shock response (HSR) and delays the disease progression in a HD mouse model. Exposure of Withaferin A activates HSF1 and induces the expression of HSP70 chaperones in an in vitro cell culture system and also suppresses mutant huntingtin aggregation in a cellular model of HD. Withaferin A treatment to HD mice considerably increased their lifespan as well as restored progressive motor behavioral deficits and declined body weight. Biochemical studies confirmed the activation of HSR and global decrease in mutant huntingtin aggregates load accompanied with improvement of striatal function in Withaferin A-treated HD mouse brain. Withaferin A-treated HD mice also exhibit significant decrease in inflammatory processes as evident from the decreased microglial activation. These results indicate immense potential of Withaferin A for the treatment of HD and related neurodegenerative disorders involving protein misfolding and aggregation.
Collapse
Affiliation(s)
- Tripti Joshi
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon, 122 051, India
| | - Vipendra Kumar
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon, 122 051, India
| | - Elena V Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences, 194064, St. Petersburg, Russia
| | - Nihar Ranjan Jana
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon, 122 051, India. .,School of Bioscience, Indian Institute of Technology, Kharagpur, 721302, India.
| |
Collapse
|
24
|
Latoszek E, Czeredys M. Molecular Components of Store-Operated Calcium Channels in the Regulation of Neural Stem Cell Physiology, Neurogenesis, and the Pathology of Huntington's Disease. Front Cell Dev Biol 2021; 9:657337. [PMID: 33869222 PMCID: PMC8047111 DOI: 10.3389/fcell.2021.657337] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
One of the major Ca2+ signaling pathways is store-operated Ca2+ entry (SOCE), which is responsible for Ca2+ flow into cells in response to the depletion of endoplasmic reticulum Ca2+ stores. SOCE and its molecular components, including stromal interaction molecule proteins, Orai Ca2+ channels, and transient receptor potential canonical channels, are involved in the physiology of neural stem cells and play a role in their proliferation, differentiation, and neurogenesis. This suggests that Ca2+ signaling is an important player in brain development. Huntington’s disease (HD) is an incurable neurodegenerative disorder that is caused by polyglutamine expansion in the huntingtin (HTT) protein, characterized by the loss of γ-aminobutyric acid (GABA)-ergic medium spiny neurons (MSNs) in the striatum. However, recent research has shown that HD is also a neurodevelopmental disorder and Ca2+ signaling is dysregulated in HD. The relationship between HD pathology and elevations of SOCE was demonstrated in different cellular and mouse models of HD and in induced pluripotent stem cell-based GABAergic MSNs from juvenile- and adult-onset HD patient fibroblasts. The present review discusses the role of SOCE in the physiology of neural stem cells and its dysregulation in HD pathology. It has been shown that elevated expression of STIM2 underlying the excessive Ca2+ entry through store-operated calcium channels in induced pluripotent stem cell-based MSNs from juvenile-onset HD. In the light of the latest findings regarding the role of Ca2+ signaling in HD pathology we also summarize recent progress in the in vitro differentiation of MSNs that derive from different cell sources. We discuss advances in the application of established protocols to obtain MSNs from fetal neural stem cells/progenitor cells, embryonic stem cells, induced pluripotent stem cells, and induced neural stem cells and the application of transdifferentiation. We also present recent progress in establishing HD brain organoids and their potential use for examining HD pathology and its treatment. Moreover, the significance of stem cell therapy to restore normal neural cell function, including Ca2+ signaling in the central nervous system in HD patients will be considered. The transplantation of MSNs or their precursors remains a promising treatment strategy for HD.
Collapse
Affiliation(s)
- Ewelina Latoszek
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Magdalena Czeredys
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| |
Collapse
|