1
|
Tapia-González S, DeFelipe J. Secretagogin as a marker to distinguish between different neuron types in human frontal and temporal cortex. Front Neuroanat 2023; 17:1210502. [PMID: 38020216 PMCID: PMC10646422 DOI: 10.3389/fnana.2023.1210502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 09/28/2023] [Indexed: 12/01/2023] Open
Abstract
The principal aim of the present work was to chemically characterize the population of neurons labeled for the calcium binding protein secretagogin (SCGN) in the human frontal and temporal cortices (Brodmann's area 10 and 21, respectively). Both cortical regions are involved in many high cognitive functions that are especially well developed (or unique) in humans, but with different functional roles. The pattern of SCGN immunostaining was rather similar in BA10 and BA21, with all the labeled neurons displaying a non-pyramidal morphology (interneurons). Although SCGN cells were present throughout all layers, they were more frequently observed in layers II, III and IV, whereas in layer I they were found only occasionally. We examined the degree of colocalization of SCGN with parvalbumin (PV) and calretinin (CR), as well as with nitric oxide synthase (nNOS; the enzyme responsible for the synthesis of nitric oxide by neurons) by triple immunostaining. We looked for possible similarities or differences in the coexpression patterns of SCGN with PV, CR and nNOS between BA10 and BA21 throughout the different cortical layers (I-VI). The percentage of colocalization was estimated by counting the number of all labeled cells through columns (1,100-1,400 μm wide) across the entire thickness of the cortex (from the pial surface to the white matter) in 50 μm-thick sections. Several hundred neurons were examined in both cortical regions. We found that SCGN cells include multiple neurochemical subtypes, whose abundance varies according to the cortical area and layer. The present results further highlight the regional specialization of cortical neurons and underline the importance of performing additional experiments to characterize the subpopulation of SCGN cells in the human cerebral cortex in greater detail.
Collapse
Affiliation(s)
- Silvia Tapia-González
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Laboratorio de Neurofisiología Celular, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
2
|
Torres MV, Ortiz-Leal I, Ferreiro A, Rois JL, Sanchez-Quinteiro P. Immunohistological study of the unexplored vomeronasal organ of an endangered mammal, the dama gazelle (Nanger dama). Microsc Res Tech 2023; 86:1206-1233. [PMID: 37494657 DOI: 10.1002/jemt.24392] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 07/28/2023]
Abstract
Dama gazelle is a threatened and rarely studied species found primarily in northern Africa. Human pressure has depleted the dama gazelle population from tens of thousands to a few hundred individuals. Since 1970, a founder population consisting of the last 17 surviving individuals in Western Sahara has been maintained in captivity, reproducing naturally. In preparation for the future implementation of assisted reproductive technology, certain aspects of dama gazelle reproductive biology have been established. However, the role played by semiochemical-mediated communications in the sexual behavior of dama gazelle remains unknown due partially to a lack of a neuroanatomical or morphofunctional characterization of the dama gazelle vomeronasal organ (VNO), which is the sensory organ responsible for pheromone processing. The present study characterized the dama gazelle VNO, which appears fully equipped to perform neurosensory functions, contributing to current understanding of interspecies VNO variability among ruminants. By employing histological, lectin-histochemical, and immunohistochemical techniques, we conducted a detailed morphofunctional evaluation of the dama gazelle VNO along its entire longitudinal axis. Our findings of significant structural and neurochemical transformation along the entire VNO suggest that future studies of the VNO should take a similar approach. The present study contributes to current understanding of dama gazelle VNO, providing a basis for future studies of semiochemical-mediated communications and reproductive management in this species. RESEARCH HIGHLIGHTS: This exhaustive immunohistological study of the vomeronasal organ (VNO) of the dama gazelle provides the first evidence of notable differences in the expression of neuronal markers along the rostrocaudal axis of the VNO. This provides a morphological basis for the implementation of pheromones in captive populations of dama gazelle.
Collapse
Affiliation(s)
- Mateo V Torres
- Department of Anatomy, Animal Production and Clinical Veterinary Sciences, Faculty of Veterinary, University of Santiago de Compostela, Lugo, Spain
| | - Irene Ortiz-Leal
- Department of Anatomy, Animal Production and Clinical Veterinary Sciences, Faculty of Veterinary, University of Santiago de Compostela, Lugo, Spain
| | | | | | - Pablo Sanchez-Quinteiro
- Department of Anatomy, Animal Production and Clinical Veterinary Sciences, Faculty of Veterinary, University of Santiago de Compostela, Lugo, Spain
| |
Collapse
|
3
|
Téllez de Meneses PG, Pérez-Revuelta L, Canal-Alonso Á, Hernández-Pérez C, Cocho T, Valero J, Weruaga E, Díaz D, Alonso JR. Immunohistochemical distribution of secretagogin in the mouse brain. Front Neuroanat 2023; 17:1224342. [PMID: 37711587 PMCID: PMC10498459 DOI: 10.3389/fnana.2023.1224342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Calcium is essential for the correct functioning of the central nervous system, and calcium-binding proteins help to finely regulate its concentration. Whereas some calcium-binding proteins such as calmodulin are ubiquitous and are present in many cell types, others such as calbindin, calretinin, and parvalbumin are expressed in specific neuronal populations. Secretagogin belongs to this latter group and its distribution throughout the brain is only partially known. In the present work, the distribution of secretagogin-immunopositive cells was studied in the entire brain of healthy adult mice. Methods Adult male C57BL/DBA mice aged between 5 and 7 months were used. Their whole brain was sectioned and used for immunohistochemistry. Specific neural populations were observed in different zones and nuclei identified according to Paxinos mouse brain atlas. Results Labelled cells were found with a Golgi-like staining, allowing an excellent characterization of their dendritic and axonal arborizations. Many secretagogin-positive cells were observed along different encephalic regions, especially in the olfactory bulb, basal ganglia, and hypothalamus. Immunostained populations were very heterogenous in both size and distribution, as some nuclei presented labelling in their entire extension, but in others, only scattered cells were present. Discussion Secretagogin can provide a more complete vision of calcium-buffering mechanisms in the brain, and can be a useful neuronal marker in different brain areas for specific populations.
Collapse
Affiliation(s)
- Pablo G. Téllez de Meneses
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Laura Pérez-Revuelta
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Ángel Canal-Alonso
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Bioinformatics, Intelligent Systems and Educational Technology (BISITE) Research Group, Universidad de Salamanca, Salamanca, Spain
| | - Carlos Hernández-Pérez
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Teresa Cocho
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Jorge Valero
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Eduardo Weruaga
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - David Díaz
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - José R. Alonso
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
4
|
Ortiz-Leal I, Torres MV, Vargas-Barroso V, Fidalgo LE, López-Beceiro AM, Larriva-Sahd JA, Sánchez-Quinteiro P. The olfactory limbus of the red fox ( Vulpes vulpes). New insights regarding a noncanonical olfactory bulb pathway. Front Neuroanat 2023; 16:1097467. [PMID: 36704406 PMCID: PMC9871471 DOI: 10.3389/fnana.2022.1097467] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction: The olfactory system in most mammals is divided into several subsystems based on the anatomical locations of the neuroreceptor cells involved and the receptor families that are expressed. In addition to the main olfactory system and the vomeronasal system, a range of olfactory subsystems converge onto the transition zone located between the main olfactory bulb (MOB) and the accessory olfactory bulb (AOB), which has been termed the olfactory limbus (OL). The OL contains specialized glomeruli that receive noncanonical sensory afferences and which interact with the MOB and AOB. Little is known regarding the olfactory subsystems of mammals other than laboratory rodents. Methods: We have focused on characterizing the OL in the red fox by performing general and specific histological stainings on serial sections, using both single and double immunohistochemical and lectin-histochemical labeling techniques. Results: As a result, we have been able to determine that the OL of the red fox (Vulpes vulpes) displays an uncommonly high degree of development and complexity. Discussion: This makes this species a novel mammalian model, the study of which could improve our understanding of the noncanonical pathways involved in the processing of chemosensory cues.
Collapse
Affiliation(s)
- Irene Ortiz-Leal
- Faculty of Veterinary, University of Santiago de Compostela, Lugo, Spain
| | - Mateo V. Torres
- Faculty of Veterinary, University of Santiago de Compostela, Lugo, Spain
| | - Víctor Vargas-Barroso
- Cellular Neuroscience, IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria
| | | | | | - Jorge A. Larriva-Sahd
- Institute of Neurobiology, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Pablo Sánchez-Quinteiro
- Faculty of Veterinary, University of Santiago de Compostela, Lugo, Spain,*Correspondence: Pablo Sanchez-Quinteiro
| |
Collapse
|
5
|
Mitra S, Basu S, Singh O, Srivastava A, Singru PS. Calcium-binding proteins typify the dopaminergic neuronal subtypes in the ventral tegmental area of zebra finch, Taeniopygia guttata. J Comp Neurol 2022; 530:2562-2586. [PMID: 35715989 DOI: 10.1002/cne.25352] [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: 11/15/2021] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Calcium-binding proteins (CBPs) regulate neuronal function in midbrain dopamine (DA)-ergic neurons in mammals by buffering and sensing the intracellular Ca2+ , and vesicular release. In birds, the equivalent set of neurons are important in song learning, directed singing, courtship, and energy balance, yet the status of CBPs in these neurons is unknown. Herein, for the first time, we probe the nature of CBPs, namely, Calbindin-, Calretinin-, Parvalbumin-, and Secretagogin-expressing DA neurons in the ventral tegmental area (VTA) and substantia nigra (SN) in the midbrain of zebra finch, Taeniopygia guttata. qRT-PCR analysis of ventral midbrain tissue fragment revealed higher Calbindin- and Calretinin-mRNA levels compared to Parvalbumin and Secretagogin. Application of immunofluorescence showed CBP-immunoreactive (-i) neurons in VTA (anterior [VTAa], mid [VTAm], caudal [VTAc]), SN (compacta [SNc], and reticulata [SNr]). Compared to VTAa, higher Calbindin- and Parvalbumin-immunoreactivity (-ir), and lower Calretinin-ir were observed in VTAm and VTAc. Secretagogin-ir was highly localized to VTAa. In SN, Calbindin- and Calretinin-ir were higher in SNc, SNr was Parvalbumin enriched, and Secretagogin-ir was not detected. Weak, moderate, and intense tyrosine hydroxylase (TH)-i VTA neurons were demarcated as subtypes 1, 2, and 3, respectively. While subtype 1 TH-i neurons were neither Calbindin- nor Calretinin-i, ∼80 and ∼65% subtype 2 and ∼30 and ∼45% subtype 3 TH-i neurons co-expressed Calbindin and Calretinin, respectively. All TH-i neuronal subtypes co-expressed Parvalbumin with reciprocal relationship with TH-ir. We suggest that the CBPs may determine VTA DA neuronal heterogeneity and differentially regulate their activity in T. guttata.
Collapse
Affiliation(s)
- Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhinav Srivastava
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
6
|
Basu S, Mitra S, Singh O, Chandramohan B, Singru PS. Secretagogin in the brain and pituitary of the catfish, Clarias batrachus: Molecular characterization and regulation by insulin. J Comp Neurol 2022; 530:1743-1772. [PMID: 35322425 DOI: 10.1002/cne.25311] [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: 08/25/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 11/12/2022]
Abstract
Secretagogin (scgn), is a novel hexa EF-hand, phylogenetically conserved calcium-binding protein. It serves as Ca2+ sensor and participates in Ca2+ -signaling and neuroendocrine regulation in mammals. However, its relevance in the brain of non-mammalian vertebrates has largely remained unexplored. To address this issue, we studied the cDNA encoding scgn, scgn mRNA expression, and distribution of scgn-equipped elements in the brain and pituitary of a teleost, Clarias batrachus (cb). The cbscgn cDNA consists of three transcripts (T) variants: T1 (2185 bp), T2 (2151 bp) and T3 (2060 bp). While 816 bp ORF in T1 and T2 encodes highly conserved six EF-hand 272 aa protein fully capable of Ca2+ -binding, 726-bp ORF in T3 encodes 242 aa protein. The T1 showed >90% and >70% identity with scgn of catfishes, and other teleosts and mammals, respectively. The T1-mRNA was widely expressed in the brain and pituitary, while the expression of T3 was restricted to the telencephalon. Application of the anti-scgn antiserum revealed a ∼32 kDa scgn-immunoreactive (scgn-i) band (known molecular weight of scgn) in the forebrain tissue, and immunohistochemically labeled neurons in the olfactory epithelium and bulb, telencephalon, preoptic area, hypothalamus, thalamus, and hindbrain. In the pituitary, scgn-i cells were seen in the pars distalis and intermedia. Insulin is reported to regulate scgn mRNA in the mammalian hippocampus, and feeding-related neuropeptides in the telencephalon of teleost. Intracranial injection of insulin significantly increased T1-mRNA expression and scgn-immunoreactivity in the telencephalon. We suggest that scgn may be an important player in the regulation of olfactory, neuroendocrine system, and energy balance functions in C. batrachus.
Collapse
Affiliation(s)
- Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Bathrachalam Chandramohan
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
7
|
Kanu B, Kia GSN, Aimola IA, Korie GC, Tekki IS. Rabies virus infection is associated with alterations in the expression of parvalbumin and secretagogin in mice brain. Metab Brain Dis 2021; 36:1267-1275. [PMID: 33783673 PMCID: PMC8008021 DOI: 10.1007/s11011-021-00717-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/14/2021] [Indexed: 12/21/2022]
Abstract
Infection with the deadly rabies virus (RABV) leads to alteration of cellular gene expression. The RABV, similar to other neurodegenerative diseases may be implicated in neuronal death due to an imbalance in Ca2+ homeostasis. Parvalbumin (PV) and Secretagogin (Scgn), two members of the Calcium-Binding Proteins (CBPs) are useful neuronal markers responsible for calcium regulation and buffering with possible protective roles against infections. This study investigated whether infection with rabies virus causes variance in expression levels of PV and Scgn using the Challenge virus standard (CVS) and Nigerian Street Rabies virus (SRV) strains. Forty-eight, 4-week-old BALB/c mice strains were divided into two test groups and challenged with Rabies virus (RABV) infection and one control group. The presence of RABV antigen was verified by direct fluorescent antibody test (DFAT) and real-time quantitative PCR (qRT-PCR) was used to assess PV and Scgn gene expression. Infection with both virus strains resulted in significant (p < 0.05) increases in expression during early infection. Mid-infection phase caused reduced expression for both genes. However, as infection progressed to the terminal phase, a lower increase in expression was measured. Gene expression and viral load correlation indicated no positive relationship. Neurons with these CBPs may have a greater capacity to buffer calcium and be more resistant to degenerative changes caused by RABV. This implies that, when PV and Scgn expression levels are kept adequately high, the integrity of neurons may be maintained and degeneration caused by RABV infection may be prevented or stopped, hence, these are possible constituents of effective rabies therapy.
Collapse
Affiliation(s)
- Brenda Kanu
- Department of Biochemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria.
| | - Grace S N Kia
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria
- Department of Veterinary Public Health, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Idowu A Aimola
- Department of Biochemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria
| | - George C Korie
- Department of Biochemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria
| | - Ishaya S Tekki
- Central Diagnostics Laboratory, National Veterinary Research Institute, Vom, Plateau State, Nigeria
| |
Collapse
|
8
|
Secretagogin marks amygdaloid PKCδ interneurons and modulates NMDA receptor availability. Proc Natl Acad Sci U S A 2021; 118:1921123118. [PMID: 33558223 DOI: 10.1073/pnas.1921123118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The perception of and response to danger is critical for an individual's survival and is encoded by subcortical neurocircuits. The amygdaloid complex is the primary neuronal site that initiates bodily reactions upon external threat with local-circuit interneurons scaling output to effector pathways. Here, we categorize central amygdala neurons that express secretagogin (Scgn), a Ca2+-sensor protein, as a subset of protein kinase Cδ (PKCδ)+ interneurons, likely "off cells." Chemogenetic inactivation of Scgn+/PKCδ+ cells augmented conditioned response to perceived danger in vivo. While Ca2+-sensor proteins are typically implicated in shaping neurotransmitter release presynaptically, Scgn instead localized to postsynaptic compartments. Characterizing its role in the postsynapse, we found that Scgn regulates the cell-surface availability of NMDA receptor 2B subunits (GluN2B) with its genetic deletion leading to reduced cell membrane delivery of GluN2B, at least in vitro. Conclusively, we describe a select cell population, which gates danger avoidance behavior with secretagogin being both a selective marker and regulatory protein in their excitatory postsynaptic machinery.
Collapse
|
9
|
Kelemen K, Szilágyi T. New Approach for Untangling the Role of Uncommon Calcium-Binding Proteins in the Central Nervous System. Brain Sci 2021. [PMID: 34069107 DOI: 10.3390/brainsci11050634ht] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Although Ca2+ ion plays an essential role in cellular physiology, calcium-binding proteins (CaBPs) were long used for mainly as immunohistochemical markers of specific cell types in different regions of the central nervous system. They are a heterogeneous and wide-ranging group of proteins. Their function was studied intensively in the last two decades and a tremendous amount of information was gathered about them. Girard et al. compiled a comprehensive list of the gene-expression profiles of the entire EF-hand gene superfamily in the murine brain. We selected from this database those CaBPs which are related to information processing and/or neuronal signalling, have a Ca2+-buffer activity, Ca2+-sensor activity, modulator of Ca2+-channel activity, or a yet unknown function. In this way we created a gene function-based selection of the CaBPs. We cross-referenced these findings with publicly available, high-quality RNA-sequencing and in situ hybridization databases (Human Protein Atlas (HPA), Brain RNA-seq database and Allen Brain Atlas integrated into the HPA) and created gene expression heat maps of the regional and cell type-specific expression levels of the selected CaBPs. This represents a useful tool to predict and investigate different expression patterns and functions of the less-known CaBPs of the central nervous system.
Collapse
Affiliation(s)
- Krisztina Kelemen
- Department of Physiology, Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Tibor Szilágyi
- Department of Physiology, Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| |
Collapse
|
10
|
Kelemen K, Szilágyi T. New Approach for Untangling the Role of Uncommon Calcium-Binding Proteins in the Central Nervous System. Brain Sci 2021; 11:brainsci11050634. [PMID: 34069107 PMCID: PMC8156796 DOI: 10.3390/brainsci11050634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Although Ca2+ ion plays an essential role in cellular physiology, calcium-binding proteins (CaBPs) were long used for mainly as immunohistochemical markers of specific cell types in different regions of the central nervous system. They are a heterogeneous and wide-ranging group of proteins. Their function was studied intensively in the last two decades and a tremendous amount of information was gathered about them. Girard et al. compiled a comprehensive list of the gene-expression profiles of the entire EF-hand gene superfamily in the murine brain. We selected from this database those CaBPs which are related to information processing and/or neuronal signalling, have a Ca2+-buffer activity, Ca2+-sensor activity, modulator of Ca2+-channel activity, or a yet unknown function. In this way we created a gene function-based selection of the CaBPs. We cross-referenced these findings with publicly available, high-quality RNA-sequencing and in situ hybridization databases (Human Protein Atlas (HPA), Brain RNA-seq database and Allen Brain Atlas integrated into the HPA) and created gene expression heat maps of the regional and cell type-specific expression levels of the selected CaBPs. This represents a useful tool to predict and investigate different expression patterns and functions of the less-known CaBPs of the central nervous system.
Collapse
|
11
|
Miczán V, Kelemen K, Glavinics JR, László ZI, Barti B, Kenesei K, Kisfali M, Katona I. NECAB1 and NECAB2 are Prevalent Calcium-Binding Proteins of CB1/CCK-Positive GABAergic Interneurons. Cereb Cortex 2021; 31:1786-1806. [PMID: 33230531 PMCID: PMC7869086 DOI: 10.1093/cercor/bhaa326] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
The molecular repertoire of the "Ca2+-signaling toolkit" supports the specific kinetic requirements of Ca2+-dependent processes in different neuronal types. A well-known example is the unique expression pattern of calcium-binding proteins, such as parvalbumin, calbindin, and calretinin. These cytosolic Ca2+-buffers control presynaptic and somatodendritic processes in a cell-type-specific manner and have been used as neurochemical markers of GABAergic interneuron types for decades. Surprisingly, to date no typifying calcium-binding proteins have been found in CB1 cannabinoid receptor/cholecystokinin (CB1/CCK)-positive interneurons that represent a large population of GABAergic cells in cortical circuits. Because CB1/CCK-positive interneurons display disparate presynaptic and somatodendritic Ca2+-transients compared with other interneurons, we tested the hypothesis that they express alternative calcium-binding proteins. By in silico data mining in mouse single-cell RNA-seq databases, we identified high expression of Necab1 and Necab2 genes encoding N-terminal EF-hand calcium-binding proteins 1 and 2, respectively, in CB1/CCK-positive interneurons. Fluorescent in situ hybridization and immunostaining revealed cell-type-specific distribution of NECAB1 and NECAB2 throughout the isocortex, hippocampal formation, and basolateral amygdala complex. Combination of patch-clamp electrophysiology, confocal, and STORM super-resolution microscopy uncovered subcellular nanoscale differences indicating functional division of labor between the two calcium-binding proteins. These findings highlight NECAB1 and NECAB2 as predominant calcium-binding proteins in CB1/CCK-positive interneurons.
Collapse
Affiliation(s)
- Vivien Miczán
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Roska Tamás Doctoral School of Sciences and Technology, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest 1083, Hungary
| | - Krisztina Kelemen
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș 540142, Romania
| | - Judit R Glavinics
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Zsófia I László
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Benjámin Barti
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Kata Kenesei
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Máté Kisfali
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
12
|
Sanders M, Petrasch-Parwez E, Habbes HW, Düring MV, Förster E. Postnatal Developmental Expression Profile Classifies the Indusium Griseum as a Distinct Subfield of the Hippocampal Formation. Front Cell Dev Biol 2021; 8:615571. [PMID: 33511122 PMCID: PMC7835525 DOI: 10.3389/fcell.2020.615571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
The indusium griseum (IG) is a cortical structure overlying the corpus callosum along its anterior–posterior extent. It has been classified either as a vestige of the hippocampus or as an extension of the dentate gyrus via the fasciola cinerea, but its attribution to a specific hippocampal subregion is still under debate. To specify the identity of IG neurons more precisely, we investigated the spatiotemporal expression of calbindin, secretagogin, Necab2, PCP4, and Prox1 in the postnatal mouse IG, fasciola cinerea, and hippocampus. We identified the calcium-binding protein Necab2 as a first reliable marker for the IG and fasciola cinerea throughout postnatal development into adulthood. In contrast, calbindin, secretagogin, and PCP4 were expressed each with a different individual time course during maturation, and at no time point, IG or fasciola cinerea principal neurons expressed Prox1, a transcription factor known to define dentate granule cell fate. Concordantly, in a transgenic mouse line expressing enhanced green fluorescent protein (eGFP) in dentate granule cells, neurons of IG and fasciola cinerea were eGFP-negative. Our findings preclude that IG neurons represent dentate granule cells, as earlier hypothesized, and strongly support the view that the IG is an own hippocampal subfield composed of a distinct neuronal population.
Collapse
Affiliation(s)
- Marie Sanders
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | | | - Hans-Werner Habbes
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | - Monika V Düring
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
13
|
Pérez-Revuelta L, Téllez de Meneses PG, López M, Briñón JG, Weruaga E, Díaz D, Alonso JR. Secretagogin expression in the mouse olfactory bulb under sensory impairments. Sci Rep 2020; 10:21533. [PMID: 33299042 PMCID: PMC7726155 DOI: 10.1038/s41598-020-78499-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/24/2020] [Indexed: 12/04/2022] Open
Abstract
The interneurons of the olfactory bulb (OB) are characterized by the expression of different calcium-binding proteins, whose specific functions are not fully understood. This is the case of one of the most recently discovered, the secretagogin (SCGN), which is expressed in interneurons of the glomerular and the granule cell layers, but whose function in the olfactory pathway is still unknown. To address this question, we examined the distribution, generation and activity of SCGN-positive interneurons in the OB of two complementary models of olfactory impairments: Purkinje Cell Degeneration (PCD) and olfactory-deprived mice. Our results showed a significant increase in the density of SCGN-positive cells in the inframitral layers of olfactory-deprived mice as compared to control animals. Moreover, BrdU analyses revealed that these additional SCGN-positive cells are not newly formed. Finally, the neuronal activity, estimated by c-Fos expression, increased in preexisting SCGN-positive interneurons of both deprived and PCD mice -being higher in the later- in comparison with control animals. Altogether, our results suggest that the OB possesses different compensatory mechanisms depending on the type of alteration. Particularly, the SCGN expression is dependent of olfactory stimuli and its function may be related to a compensation against a reduction in sensory inputs.
Collapse
Affiliation(s)
- L Pérez-Revuelta
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - P G Téllez de Meneses
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - M López
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - J G Briñón
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - E Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - D Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain. .,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain.
| | - J R Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| |
Collapse
|
14
|
Attaai AH, Noreldin AE, Abdel-Maksoud FM, Hussein MT. An updated investigation on the dromedary camel cerebellum (Camelus dromedarius) with special insight into the distribution of calcium-binding proteins. Sci Rep 2020; 10:21157. [PMID: 33273572 PMCID: PMC7713137 DOI: 10.1038/s41598-020-78192-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/12/2020] [Indexed: 01/07/2023] Open
Abstract
Studying the cerebella of different animals is important to expand the knowledge about the cerebellum. Studying the camel cerebellum was neglected even though the recent research in the middle east and Asia. Therefore, the present study was designed to achieve a detailed description of the morphology and the cellular organization of the camel cerebellum. Because of the high importance of the calcium ions as a necessary moderator the current work also aimed to investigate the distribution of calcium binding proteins (CaBP) such as calbindin D-28K (CB), parvalbumin (PV) and calretinin (CR) in different cerebellar cells including the non-traditional neurons. The architecture of camel cerebellum, as different mammals, consists of the medulla and three layered-cortex. According to our observation the cells in the granular layer were not crowded and many spaces were observed. CB expression was the highest by Purkinje cells including their dendritic arborization. In addition to its expression by the inhibitory interneurons (basket, stellate and Golgi neurons), it is also expressed by the excitatory granule cells. PV was expressed by Purkinje cells, including their primary arborization, and by the molecular layer cells. CR immunoreactivity (-ir) was obvious in almost all cell layers with varying degrees, however a weak or any expression by the Purkinje cells. The molecular layer cells and the Golgi and the non traditional large neurons of the granular layer showed the strongest CR-ir. Granule neurons showed moderate immunoreactivity for CB and CR. In conclusion, the results of the current study achieved a complete map for the neurochemical organization of CaBP expression and distribution by different cells in the camel cerebellum.
Collapse
Affiliation(s)
- Abdelraheim H Attaai
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, 71526, Assiut, Egypt
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, 22511, Damanhour, Egypt
| | - Fatma M Abdel-Maksoud
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, 71526, Assiut, Egypt.
| | - Manal T Hussein
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, 71526, Assiut, Egypt
| |
Collapse
|
15
|
Expression of ArfGAP3 in Vaginal Anterior Wall of Patients With Pelvic Floor Organ Prolapse in Pelvic Organ Prolapse and Non-Pelvic Organ Prolapse Patients. Female Pelvic Med Reconstr Surg 2020; 27:e64-e69. [PMID: 31868832 PMCID: PMC7774809 DOI: 10.1097/spv.0000000000000808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The purpose of this study was to study the expression of adenosine diphosphate ribosylation factor GTPase-activating protein 3 (ArfGAP3) in the anterior vaginal wall of patients with pelvic organ prolapse (POP).
Collapse
|
16
|
Dong Y, Li Y, Liu R, Li Y, Zhang H, Liu H, Chen J. Secretagogin, a marker for neuroendocrine cells, is more sensitive and specific in large cell neuroendocrine carcinoma compared with the markers CD56, CgA, Syn and Napsin A. Oncol Lett 2020; 19:2223-2230. [PMID: 32194720 PMCID: PMC7039081 DOI: 10.3892/ol.2020.11336] [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: 10/02/2018] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
A common method to distinguish large cell neuroendocrine carcinoma (LCNEC) from non-neuroendocrine large cell carcinoma (non-NE LCC) is from using specific immunohistochemistry markers, such as CgA, Syn, CD56 and Napsin A, however, the results remain controversial using these markers. Secretagogin (SCGN) is a newly discovered biomarker of neuroendocrine cells. In the present study, the expression of SCGN in 33 cases of human lung large cell carcinoma (LCC), including 17 cases of LCNEC and 16 cases of non-neuroendocrine (NE) LCC and lung cancer cell lines (A549, H1650, H358, H292 and H661). The association between SCGN expression and the clinicopathological characteristics of patients, including sex, age, clinical stage and metastasis, was analyzed. The results revealed that the different lung cancer cell lines had different expression levels of SCGN, and the SCGN protein was localized in the nucleus and cytoplasm of A549 cells detected using immunofluorescence. A total of 54.5% (18/33) of specimens positively expressed the SCGN protein. Of the 17 patients with LCNEC, only 23.5% (4/17) of cases were CgA positive, 35.29% (6/17) were Syn positive, 41.2% (7/17) were CD56 positive, and 41.2% (7/17) were Napsin A positive. However, SCGN was positively detected in 94.1% (16/17) of patients with LCNEC, which was more frequent compared with that in CgA, Syn, CD56 and Napsin A. Analysis of the clinical characteristics indicated that SCGN expression was only significantly associated with pathological type in patients with lung cancer (P<0.001). Furthermore, a positive correlation was observed between SCGN expression and CgA, Syn, and CD56 expression in patients with LCNEC. SCGN was co-localized with the NE markers (CgA, Syn, and CD56) in A549 lung cancer cells and in LCNEC tissues. Thus, SCGN displayed more sensitivity and specificity in lung cancer cells with NE differentiation. A combined analysis of SCGN and other common NE markers may be a potential tool for diagnosing these tumors.
Collapse
Affiliation(s)
- Yunlong Dong
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Department of Thoracic Surgery, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin 301800, P.R. China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Renwang Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Ying Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
17
|
Secretagogin Immunoreactivity Reveals Lugaro Cells in the Pigeon Cerebellum. THE CEREBELLUM 2019; 18:544-555. [PMID: 30904983 DOI: 10.1007/s12311-019-01023-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Lugaro cells are inhibitory interneurons found in the upper granular layer of the cerebellar cortex, just below or within the Purkinje cell layer. They are characterized by (1) a fusiform soma oriented in the parasagittal plane, (2) two pairs of dendrites emanating from opposite ends of the soma, (3) innervation from Purkinje cell collaterals, and (4) an axon that projects into the molecular layer akin to granular cell parallel fibers. Lugaro cells have been described in mammals, but not in other vertebrate classes, save one report in teleost fish. Here, we propose the existence of Lugaro cells in the avian cerebellum based on the morphological characteristics and connectivity described above. Immunohistochemical staining against the calcium binding protein secretagogin (SCGN) revealed Lugaro-like cells in the pigeon cerebellum. Some SCGN-labeled cells exhibit fusiform somata and dendrites parallel to the Purkinje cell layer in the parasagittal plane, as well as long axons that project into the molecular layer and travel alongside parallel fibers in the coronal plane. While mammalian Lugaro cells are known to express calretinin, the SCGN-labeled cells in the pigeon do not. SCGN-labeled cells also express glutamic acid decarboxylase, confirming their inhibitory function. Calbindin labeling revealed Purkinje cell terminals surrounding the SCGN-expressing cells. Our results suggest that Lugaro cells are more widespread among vertebrates than previously thought and may be a characteristic of the cerebellum of all vertebrates.
Collapse
|
18
|
Erzin G, Topçuoğlu C, Bayram Ş, Karadağ H, Ozkaya G, Turhan T, Göka E. Secretagogin may not be a new neuroendocrine biomarker in schizophrenia while levels may reflect clinical severity. PSYCHIAT CLIN PSYCH 2019. [DOI: 10.1080/24750573.2019.1589175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Gamze Erzin
- Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Psychiatry Department, Ankara, Turkey
- Present/permanent work address: Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Psychiatry Department, Ankara, Turkey
| | - Canan Topçuoğlu
- Biochemistry Department, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Şenol Bayram
- Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Psychiatry Department, Ankara, Turkey
| | - Hasan Karadağ
- Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Psychiatry Department, Ankara, Turkey
- Present/permanent work address: Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Psychiatry Department, Ankara, Turkey
| | - Güven Ozkaya
- Faculty of Medicine, Biostatistic Department, Uludag University, Bursa, Turkey
| | - Turan Turhan
- Biochemistry Department, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Erol Göka
- Ankara Diskapi Yildirim Beyazit Training and Research Hospital, Psychiatry Department, Ankara, Turkey
| |
Collapse
|
19
|
Zahola P, Hanics J, Pintér A, Máté Z, Gáspárdy A, Hevesi Z, Echevarria D, Adori C, Barde S, Törőcsik B, Erdélyi F, Szabó G, Wagner L, Kovacs GG, Hökfelt T, Harkany T, Alpár A. Secretagogin expression in the vertebrate brainstem with focus on the noradrenergic system and implications for Alzheimer's disease. Brain Struct Funct 2019; 224:2061-2078. [PMID: 31144035 PMCID: PMC6591208 DOI: 10.1007/s00429-019-01886-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/03/2019] [Indexed: 12/04/2022]
Abstract
Calcium-binding proteins are widely used to distinguish neuronal subsets in the brain. This study focuses on secretagogin, an EF-hand calcium sensor, to identify distinct neuronal populations in the brainstem of several vertebrate species. By using neural tube whole mounts of mouse embryos, we show that secretagogin is already expressed during the early ontogeny of brainstem noradrenaline cells. In adults, secretagogin-expressing neurons typically populate relay centres of special senses and vegetative regulatory centres of the medulla oblongata, pons and midbrain. Notably, secretagogin expression overlapped with the brainstem column of noradrenergic cell bodies, including the locus coeruleus (A6) and the A1, A5 and A7 fields. Secretagogin expression in avian, mouse, rat and human samples showed quasi-equivalent patterns, suggesting conservation throughout vertebrate phylogeny. We found reduced secretagogin expression in locus coeruleus from subjects with Alzheimer’s disease, and this reduction paralleled the loss of tyrosine hydroxylase, the enzyme rate limiting noradrenaline synthesis. Residual secretagogin immunoreactivity was confined to small submembrane domains associated with initial aberrant tau phosphorylation. In conclusion, we provide evidence that secretagogin is a useful marker to distinguish neuronal subsets in the brainstem, conserved throughout several species, and its altered expression may reflect cellular dysfunction of locus coeruleus neurons in Alzheimer’s disease.
Collapse
Affiliation(s)
- Péter Zahola
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary.,Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - János Hanics
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary.,Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Anna Pintér
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Zoltán Máté
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anna Gáspárdy
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Zsófia Hevesi
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary.,Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Diego Echevarria
- Institute of Neuroscience, University of Miguel Hernandez de Elche, Alicante, Spain
| | - Csaba Adori
- Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Beáta Törőcsik
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Ferenc Erdélyi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Ludwig Wagner
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria.,Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Alán Alpár
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary. .,Department of Anatomy, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
20
|
Maj M, Wagner L, Tretter V. 20 Years of Secretagogin: Exocytosis and Beyond. Front Mol Neurosci 2019; 12:29. [PMID: 30853888 PMCID: PMC6396707 DOI: 10.3389/fnmol.2019.00029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/23/2019] [Indexed: 01/04/2023] Open
Abstract
Calcium is one of the most important signaling factors in mammalian cells. Specific temporal and spatial calcium signals underlie fundamental processes such as cell growth, development, circadian rhythms, neurotransmission, hormonal actions and apoptosis. In order to translate calcium signals into cellular processes a vast number of proteins bind this ion with affinities from the nanomolar to millimolar range. Using classical biochemical methods an impressing number of calcium binding proteins (CBPs) have been discovered since the late 1960s, some of which are expressed ubiquitously, others are more restricted to specific cell types. In the nervous system expression patterns of different CBPs have been used to discern different neuronal cell populations, especially before advanced methods like single-cell transcriptomics and activity recording were available to define neuronal identity. However, understanding CBPs and their interacting proteins is still of central interest. The post-genomic era has coined the term “calciomics,” to describe a whole new research field, that engages in the identification and characterization of CBPs and their interactome. Secretagogin is a CBP, that was discovered 20 years ago in the pancreas. Consecutively it was found also in other organs including the nervous system, with characteristic expression patterns mostly forming cell clusters. Its regional expression and subcellular location together with the identification of protein interaction partners implicated, that secretagogin has a central role in hormone secretion. Meanwhile, with the help of modern proteomics a large number of actual and putative interacting proteins has been identified, that allow to anticipate a much more complex role of secretagogin in developing and adult neuronal cells. Here, we review recent findings that appear like puzzle stones of a greater picture.
Collapse
Affiliation(s)
- Magdalena Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA, United States
| | - Ludwig Wagner
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medizinische Universität Wien, Vienna, Austria
| | - Verena Tretter
- Department of Anesthesia and General Intensive Care, Clinical Department of Anesthesia, Medizinische Universität Wien, Vienna, Austria
| |
Collapse
|
21
|
Alpár A, Zahola P, Hanics J, Hevesi Z, Korchynska S, Benevento M, Pifl C, Zachar G, Perugini J, Severi I, Leitgeb P, Bakker J, Miklosi AG, Tretiakov E, Keimpema E, Arque G, Tasan RO, Sperk G, Malenczyk K, Máté Z, Erdélyi F, Szabó G, Lubec G, Palkovits M, Giordano A, Hökfelt TG, Romanov RA, Horvath TL, Harkany T. Hypothalamic CNTF volume transmission shapes cortical noradrenergic excitability upon acute stress. EMBO J 2018; 37:e100087. [PMID: 30209240 PMCID: PMC6213283 DOI: 10.15252/embj.2018100087] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 02/06/2023] Open
Abstract
Stress-induced cortical alertness is maintained by a heightened excitability of noradrenergic neurons innervating, notably, the prefrontal cortex. However, neither the signaling axis linking hypothalamic activation to delayed and lasting noradrenergic excitability nor the molecular cascade gating noradrenaline synthesis is defined. Here, we show that hypothalamic corticotropin-releasing hormone-releasing neurons innervate ependymal cells of the 3rd ventricle to induce ciliary neurotrophic factor (CNTF) release for transport through the brain's aqueductal system. CNTF binding to its cognate receptors on norepinephrinergic neurons in the locus coeruleus then initiates sequential phosphorylation of extracellular signal-regulated kinase 1 and tyrosine hydroxylase with the Ca2+-sensor secretagogin ensuring activity dependence in both rodent and human brains. Both CNTF and secretagogin ablation occlude stress-induced cortical norepinephrine synthesis, ensuing neuronal excitation and behavioral stereotypes. Cumulatively, we identify a multimodal pathway that is rate-limited by CNTF volume transmission and poised to directly convert hypothalamic activation into long-lasting cortical excitability following acute stress.
Collapse
Affiliation(s)
- Alán Alpár
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - Péter Zahola
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - János Hanics
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - Zsófia Hevesi
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Solomiia Korchynska
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Christian Pifl
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gergely Zachar
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
| | - Jessica Perugini
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Ilenia Severi
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Patrick Leitgeb
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Joanne Bakker
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Andras G Miklosi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gloria Arque
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Ramon O Tasan
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Günther Sperk
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Katarzyna Malenczyk
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Zoltán Máté
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Ferenc Erdélyi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gert Lubec
- Paracelsus Medical University, Salzburg, Austria
| | - Miklós Palkovits
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, Hungary
| | - Antonio Giordano
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Tomas Gm Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Departments of Comparative Medicine and Neuroscience, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
22
|
Malenczyk K, Szodorai E, Schnell R, Lubec G, Szabó G, Hökfelt T, Harkany T. Secretagogin protects Pdx1 from proteasomal degradation to control a transcriptional program required for β cell specification. Mol Metab 2018; 14:108-120. [PMID: 29910119 PMCID: PMC6034064 DOI: 10.1016/j.molmet.2018.05.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Specification of endocrine cell lineages in the developing pancreas relies on extrinsic signals from non-pancreatic tissues, which initiate a cell-autonomous sequence of transcription factor activation and repression switches. The steps in this pathway share reliance on activity-dependent Ca2+ signals. However, the mechanisms by which phasic Ca2+ surges become converted into a dynamic, cell-state-specific and physiologically meaningful code made up by transcription factors constellations remain essentially unknown. METHODS We used high-resolution histochemistry to explore the coincident expression of secretagogin and transcription factors driving β cell differentiation. Secretagogin promoter activity was tested in response to genetically manipulating Pax6 and Pax4 expression. Secretagogin null mice were produced with their pancreatic islets morphologically and functionally characterized during fetal development. A proteomic approach was utilized to identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome and verified in vitro by focusing on Pdx1 retention. RESULTS Here, we show that secretagogin, a Ca2+ sensor protein that controls α and β cell turnover in adult, is in fact expressed in endocrine pancreas from the inception of lineage segregation in a Pax4-and Pax6-dependent fashion. By genetically and pharmacologically manipulating secretagogin expression and interactome engagement in vitro, we find secretagogin to gate excitation-driven Ca2+ signals for β cell differentiation and insulin production. Accordingly, secretagogin-/- fetuses retain a non-committed pool of endocrine progenitors that co-express both insulin and glucagon. We identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome complex to prevent Pdx1 degradation through proteasome inactivation. This coincides with retained Nkx6.1, Pax4 and insulin transcription in prospective β cells. CONCLUSIONS In sum, secretagogin scales the temporal availability of a Ca2+-dependent transcription factor network to define β cell identity.
Collapse
Affiliation(s)
- Katarzyna Malenczyk
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Paracelsus Medical University, Strubergasse 21, A-5020, Salzburg, Austria
| | - Robert Schnell
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, SE-17177, Stockholm, Sweden
| | - Gert Lubec
- Paracelsus Medical University, Strubergasse 21, A-5020, Salzburg, Austria
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony utca 43, H-1083, Budapest, Hungary
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden.
| |
Collapse
|
23
|
Raju CS, Spatazza J, Stanco A, Larimer P, Sorrells SF, Kelley KW, Nicholas CR, Paredes MF, Lui JH, Hasenstaub AR, Kriegstein AR, Alvarez-Buylla A, Rubenstein JL, Oldham MC. Secretagogin is Expressed by Developing Neocortical GABAergic Neurons in Humans but not Mice and Increases Neurite Arbor Size and Complexity. Cereb Cortex 2018; 28:1946-1958. [PMID: 28449024 PMCID: PMC6019052 DOI: 10.1093/cercor/bhx101] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/10/2017] [Indexed: 11/14/2022] Open
Abstract
The neocortex of primates, including humans, contains more abundant and diverse inhibitory neurons compared with rodents, but the molecular foundations of these observations are unknown. Through integrative gene coexpression analysis, we determined a consensus transcriptional profile of GABAergic neurons in mid-gestation human neocortex. By comparing this profile to genes expressed in GABAergic neurons purified from neonatal mouse neocortex, we identified conserved and distinct aspects of gene expression in these cells between the species. We show here that the calcium-binding protein secretagogin (SCGN) is robustly expressed by neocortical GABAergic neurons derived from caudal ganglionic eminences (CGE) and lateral ganglionic eminences during human but not mouse brain development. Through electrophysiological and morphometric analyses, we examined the effects of SCGN expression on GABAergic neuron function and form. Forced expression of SCGN in CGE-derived mouse GABAergic neurons significantly increased total neurite length and arbor complexity following transplantation into mouse neocortex, revealing a molecular pathway that contributes to morphological differences in these cells between rodents and primates.
Collapse
Affiliation(s)
- Chandrasekhar S Raju
- Department of Neurological Surgery, University of California, San Francisco, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
| | - Julien Spatazza
- Department of Neurological Surgery, University of California, San Francisco, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
- Neurona Therapeutics, South San Francisco, CA, USA
| | - Amelia Stanco
- Department of Psychiatry, University of California, San Francisco, USA
- EntroGen, Woodland Hills, CA, USA
| | - Phillip Larimer
- Center for Integrative Neuroscience, University of California, San Francisco, USA
- Department of Neurology, University of California, San Francisco, USA
| | - Shawn F Sorrells
- Department of Neurological Surgery, University of California, San Francisco, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
| | - Kevin W Kelley
- Department of Neurological Surgery, University of California, San Francisco, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
| | - Cory R Nicholas
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
- Department of Neurology, University of California, San Francisco, USA
- Neurona Therapeutics, South San Francisco, CA, USA
| | - Mercedes F Paredes
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
- Department of Neurology, University of California, San Francisco, USA
| | - Jan H Lui
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
- Department of Neurology, University of California, San Francisco, USA
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA, USA
| | - Andrea R Hasenstaub
- Center for Integrative Neuroscience, University of California, San Francisco, USA
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, USA
| | - Arnold R Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
- Department of Neurology, University of California, San Francisco, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, University of California, San Francisco, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
| | - John L Rubenstein
- Department of Psychiatry, University of California, San Francisco, USA
| | - Michael C Oldham
- Department of Neurological Surgery, University of California, San Francisco, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, USA
| |
Collapse
|
24
|
Najdzion J. Cocaine- and amphetamine–regulated transcript peptide and calcium binding proteins immunoreactivity in the deep layers of the superior colliculus of the guinea pig: Implications for multisensory and visuomotor processing. J Chem Neuroanat 2018; 88:55-69. [DOI: 10.1016/j.jchemneu.2017.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/20/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022]
|
25
|
Kosaka T, Kosaka K. Calcium-binding protein, secretagogin, specifies the microcellular tegmental nucleus and intermediate and ventral parts of the cuneiform nucleus of the mouse and rat. Neurosci Res 2018; 134:30-38. [PMID: 29366872 DOI: 10.1016/j.neures.2018.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 11/30/2022]
Abstract
Secretagogin (SCGN) is a recently discovered calcium binding protein of the EF hand family, cloned from β cells of pancreatic island of Langerhans and endocrine cells of the gastrointestinal gland. SCGN characterizes some particular neuron groups in various regions of the nervous system and is considered as one of the useful neuron subpopulation markers. In the present study we reported that SCGN specifically labelled a particular neuronal cluster in the brainstem of the mice and rats. The comparison of the SCGN immunostaining with the choline acetyltransferase immunostaining and acetylcholinesterase staining clearly indicated that the particular cluster of SCGN positive neurons corresponded to the microcellular tegmental nucleus (MiTg) and the ventral portion of the cuneiform nucleus (CnF), both of which are components of the isthmus. The analyses in mice indicated that SCGN positive neurons in the MiTg and CnF were homogeneous in size and shape, appearing to compose a single complex: their somata were small comparing with the adjacent cholinergic neurons in the pedunculotegmantal nucleus, 10.5 vs 16.0 μm in diameter, and extended 2-3 slender smooth processes. SCGN might be one of significant markers to reconsider the delineations of the structures of the mouse, and presumably rat, brainstem.
Collapse
Affiliation(s)
- Toshio Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan.
| | - Katsuko Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan
| |
Collapse
|
26
|
Dudczig S, Currie PD, Jusuf PR. Developmental and adult characterization of secretagogin expressing amacrine cells in zebrafish retina. PLoS One 2017; 12:e0185107. [PMID: 28949993 PMCID: PMC5614429 DOI: 10.1371/journal.pone.0185107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 09/06/2017] [Indexed: 12/19/2022] Open
Abstract
Calcium binding proteins show stereotypical expression patterns within diverse neuron types across the central nervous system. Here, we provide a characterization of developmental and adult secretagogin-immunolabelled neurons in the zebrafish retina with an emphasis on co-expression of multiple calcium binding proteins. Secretagogin is a recently identified and cloned member of the F-hand family of calcium binding proteins, which labels distinct neuron populations in the retinas of mammalian vertebrates. Both the adult distribution of secretagogin labeled retinal neurons as well as the developmental expression indicative of the stage of neurogenesis during which this calcium binding protein is expressed was quantified. Secretagogin expression was confined to an amacrine interneuron population in the inner nuclear layer, with monostratified neurites in the center of the inner plexiform layer and a relatively regular soma distribution (regularity index > 2.5 across central–peripheral areas). However, only a subpopulation (~60%) co-labeled with gamma-aminobutyric acid as their neurotransmitter, suggesting that possibly two amacrine subtypes are secretagogin immunoreactive. Quantitative co-labeling analysis with other known amacrine subtype markers including the three main calcium binding proteins parvalbumin, calbindin and calretinin identifies secretagogin immunoreactive neurons as a distinct neuron population. The highest density of secretagogin cells of ~1800 cells / mm2 remained relatively evenly along the horizontal meridian, whilst the density dropped of to 125 cells / mm2 towards the dorsal and ventral periphery. Thus, secretagogin represents a new amacrine label within the zebrafish retina. The developmental expression suggests a possible role in late stage differentiation. This characterization forms the basis of functional studies assessing how the expression of distinct calcium binding proteins might be regulated to compensate for the loss of one of the others.
Collapse
Affiliation(s)
- Stefanie Dudczig
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- School of Biosciences, University of Melbourne, Parkville, VIC, Australia
| | - Peter David Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Patricia Regina Jusuf
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- School of Biosciences, University of Melbourne, Parkville, VIC, Australia
- * E-mail:
| |
Collapse
|
27
|
Chandra AJ, Lee SCS, Grünert U. Thorny ganglion cells in marmoset retina: Morphological and neurochemical characterization with antibodies against calretinin. J Comp Neurol 2017; 525:3962-3974. [PMID: 28875500 DOI: 10.1002/cne.24319] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/18/2017] [Accepted: 08/26/2017] [Indexed: 11/06/2022]
Abstract
In primates, over 17 morphological types of retinal ganglion cell have been distinguished by their dendritic morphology and stratification, but reliable markers for specific ganglion cell populations are still rare. The calcium binding protein calretinin is known to be expressed in the inner nuclear and the ganglion cell layer of marmoset retina, however, the specific cell type(s) expressing calretinin in the ganglion cell layer are yet to be determined. Here, we identified calretinin positive retinal ganglion cells in the common marmoset Callithrix jacchus. Double labeling with the ganglion cell marker RBPMS demonstrated that the large majority (80%) of the calretinin positive cells in the ganglion cell layer are ganglion cells, and 20% are displaced amacrine cells. The calretinin positive ganglion cells made up on average 12% of the total ganglion cell population outside of the foveal region and their proportion increased with eccentricity. Prelabeling with antibodies against calretinin and subsequent intracellular injection with DiI revealed that the large majority of the injected cells (n = 74) were either narrow thorny or broad thorny ganglion cells, 14 cells were displaced amacrine cells. Narrow thorny cells were further distinguished into outer and inner stratifying cells. In addition, weakly labeled cells with a large soma were identified as parasol ganglion cells. Our results show that three types of thorny ganglion cells in marmoset retina can be identified with antibodies against calretinin. Our findings are also consistent with the idea that the proportion of wide-field ganglion cell types increases in peripheral retina.
Collapse
Affiliation(s)
- Ashleigh J Chandra
- Department of Clinical Ophthalmology, Save Sight Institute, The University of Sydney, Sydney, New South Wales, 2000, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, New South Wales, 2000, Australia
| | - Sammy C S Lee
- Department of Clinical Ophthalmology, Save Sight Institute, The University of Sydney, Sydney, New South Wales, 2000, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, New South Wales, 2000, Australia
| | - Ulrike Grünert
- Department of Clinical Ophthalmology, Save Sight Institute, The University of Sydney, Sydney, New South Wales, 2000, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, New South Wales, 2000, Australia.,Discipline of Anatomy & Histology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, 2000, Australia
| |
Collapse
|
28
|
Oveland E, Nystad A, Berven F, Myhr KM, Torkildsen Ø, Wergeland S. 1,25-Dihydroxyvitamin-D3 induces brain proteomic changes in cuprizone mice during remyelination involving calcium proteins. Neurochem Int 2017; 112:267-277. [PMID: 28818673 DOI: 10.1016/j.neuint.2017.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 07/20/2017] [Accepted: 08/11/2017] [Indexed: 01/15/2023]
Abstract
Dietary supplementation of vitamin D is commonly recommended to patients with multiple sclerosis. We recently found that high-dose of the hormonally active 1,25-dihydroxyvitamin-D3 (1,25D) promotes myelin repair in the cuprizone model for de- and remyelination. In the present study, we quantified 5062 proteins, of which 125 were differentially regulated in brain tissue from 1,25D treated mice during remyelination, compared to placebo. Proteins upregulated in the early remyelination phase were involved in calcium binding, e.g. calretinin (>1.3 fold, p < 0.005), S10A5 and secretagogin, and involved in mitochondrial function, e.g. NADH-ubiquinone oxidoreductase chain 3, and acyl-coenzyme A synthetase. Calretinin, S10A5 and secretagogin expression levels were characterized using immunohistochemistry. Calretinin immunoreactivity was significantly increased (>3 fold, p = 0.016) in the medial septal nuclei of 1,25D treated mice in the early remyelination phase. Our results indicate that vitamin D may influence remyelination by mechanisms involving an increase in calretinin expression and potentially other calcium binding proteins.
Collapse
Affiliation(s)
- Eystein Oveland
- Proteomics Unit at University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Agnes Nystad
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Frode Berven
- Proteomics Unit at University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Norway; Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kjell-Morten Myhr
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Øivind Torkildsen
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Stig Wergeland
- Kristian Gerhard Jebsen MS Research Centre, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
29
|
Malenczyk K, Girach F, Szodorai E, Storm P, Segerstolpe Å, Tortoriello G, Schnell R, Mulder J, Romanov RA, Borók E, Piscitelli F, Di Marzo V, Szabó G, Sandberg R, Kubicek S, Lubec G, Hökfelt T, Wagner L, Groop L, Harkany T. A TRPV1-to-secretagogin regulatory axis controls pancreatic β-cell survival by modulating protein turnover. EMBO J 2017; 36:2107-2125. [PMID: 28637794 PMCID: PMC5510001 DOI: 10.15252/embj.201695347] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 04/27/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022] Open
Abstract
Ca2+-sensor proteins are generally implicated in insulin release through SNARE interactions. Here, secretagogin, whose expression in human pancreatic islets correlates with their insulin content and the incidence of type 2 diabetes, is shown to orchestrate an unexpectedly distinct mechanism. Single-cell RNA-seq reveals retained expression of the TRP family members in β-cells from diabetic donors. Amongst these, pharmacological probing identifies Ca2+-permeable transient receptor potential vanilloid type 1 channels (TRPV1) as potent inducers of secretagogin expression through recruitment of Sp1 transcription factors. Accordingly, agonist stimulation of TRPV1s fails to rescue insulin release from pancreatic islets of glucose intolerant secretagogin knock-out(-/-) mice. However, instead of merely impinging on the SNARE machinery, reduced insulin availability in secretagogin-/- mice is due to β-cell loss, which is underpinned by the collapse of protein folding and deregulation of secretagogin-dependent USP9X deubiquitinase activity. Therefore, and considering the desensitization of TRPV1s in diabetic pancreata, a TRPV1-to-secretagogin regulatory axis seems critical to maintain the structural integrity and signal competence of β-cells.
Collapse
Affiliation(s)
- Katarzyna Malenczyk
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fatima Girach
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Petter Storm
- Department of Clinical Sciences, Diabetes and Endocrinology CRC, Skåne University Hospital Malmö, Malmö, Sweden
| | - Åsa Segerstolpe
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | | | - Robert Schnell
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jan Mulder
- Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Erzsébet Borók
- Department of Cognitive Neurobiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli Naples, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli Naples, Italy
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Rickard Sandberg
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | - Stefan Kubicek
- CeMM Research Centre for Molecular Medicine, Vienna, Austria
| | - Gert Lubec
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ludwig Wagner
- University Clinic for Internal Medicine III, General Hospital Vienna, Vienna, Austria
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology CRC, Skåne University Hospital Malmö, Malmö, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Romanov RA, Alpár A, Hökfelt T, Harkany T. Molecular diversity of corticotropin-releasing hormone mRNA-containing neurons in the hypothalamus. J Endocrinol 2017; 232:R161-R172. [PMID: 28057867 DOI: 10.1530/joe-16-0256] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/04/2017] [Indexed: 01/05/2023]
Abstract
Hormonal responses to acute stress rely on the rapid induction of corticotropin-releasing hormone (CRH) production in the mammalian hypothalamus, with subsequent instructive steps culminating in corticosterone release at the periphery. Hypothalamic CRH neurons in the paraventricular nucleus of the hypothalamus are therefore considered as 'stress neurons'. However, significant morphological and functional diversity among neurons that can transiently produce CRH in other hypothalamic nuclei has been proposed, particularly as histochemical and molecular biology evidence associates CRH to both GABA and glutamate neurotransmission. Here, we review recent advances through single-cell RNA sequencing and circuit mapping to suggest that CRH production reflects a state switch in hypothalamic neurons and thus confers functional competence rather than being an identity mark of phenotypically segregated neurons. We show that CRH mRNA transcripts can therefore be seen in GABAergic, glutamatergic and dopaminergic neuronal contingents in the hypothalamus. We then distinguish 'stress neurons' of the paraventricular nucleus that constitutively express secretagogin, a Ca2+ sensor critical for the stimulus-driven assembly of the molecular machinery underpinning the fast regulated exocytosis of CRH at the median eminence. Cumulatively, we infer that CRH neurons are functionally and molecularly more diverse than previously thought.
Collapse
Affiliation(s)
- Roman A Romanov
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Alán Alpár
- MTA-SE NAP Research Group of Experimental Neuroanatomy and Developmental BiologyHungarian Academy of Sciences, Budapest, Hungary
- Department of AnatomySemmelweis University, Budapest, Hungary
| | - Tomas Hökfelt
- Department of NeuroscienceKarolinska Institutet, Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of NeuroscienceKarolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Secretagogin-dependent matrix metalloprotease-2 release from neurons regulates neuroblast migration. Proc Natl Acad Sci U S A 2017; 114:E2006-E2015. [PMID: 28223495 DOI: 10.1073/pnas.1700662114] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The rostral migratory stream (RMS) is viewed as a glia-enriched conduit of forward-migrating neuroblasts in which chemorepulsive signals control the pace of forward migration. Here we demonstrate the existence of a scaffold of neurons that receive synaptic inputs within the rat, mouse, and human fetal RMS equivalents. These neurons express secretagogin, a Ca2+-sensor protein, to execute an annexin V-dependent externalization of matrix metalloprotease-2 (MMP-2) for reconfiguring the extracellular matrix locally. Mouse genetics combined with pharmacological probing in vivo and in vitro demonstrate that MMP-2 externalization occurs on demand and that its loss slows neuroblast migration. Loss of function is particularly remarkable upon injury to the olfactory bulb. Cumulatively, we identify a signaling cascade that provokes structural remodeling of the RMS through recruitment of MMP-2 by a previously unrecognized neuronal constituent. Given the life-long presence of secretagogin-containing neurons in human, this mechanism might be exploited for therapeutic benefit in rescue strategies.
Collapse
|
32
|
Kosaka T, Yasuda S, Kosaka K. Calcium-binding protein, secretagogin, characterizes novel groups of interneurons in the rat striatum. Neurosci Res 2017; 119:53-60. [PMID: 28193530 DOI: 10.1016/j.neures.2017.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/28/2016] [Accepted: 01/18/2017] [Indexed: 11/25/2022]
Abstract
In the rat striatum numerous secretagogin (SCGN) positive neurons were scattered. They were heterogeneous in their morphological and chemical properties. We examined the colocalization of SCGN with known four interneuron markers, parvalbumin (PV), calretinin (CR), nitric oxide synthase (NOS) and choline acetyl transferase (ChAT). 60-70% of SCGN positive striatal neurons contained either PV or CR or ChAT, but none contained NOS. On the other hand the remaining 30-40% expressed none of these markers, most of which were GAD positive. The present study indicates that there are hitherto unknown groups of striatal interneurons in the rat striatum.
Collapse
Affiliation(s)
- Toshio Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan.
| | - Seiko Yasuda
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan
| | - Katsuko Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan
| |
Collapse
|
33
|
Khandelwal R, Sharma AK, Chadalawada S, Sharma Y. Secretagogin Is a Redox-Responsive Ca2+ Sensor. Biochemistry 2017; 56:411-420. [DOI: 10.1021/acs.biochem.6b00761] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Radhika Khandelwal
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Anand Kumar Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | - Swathi Chadalawada
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | - Yogendra Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| |
Collapse
|
34
|
Garas FN, Shah RS, Kormann E, Doig NM, Vinciati F, Nakamura KC, Dorst MC, Smith Y, Magill PJ, Sharott A. Secretagogin expression delineates functionally-specialized populations of striatal parvalbumin-containing interneurons. eLife 2016; 5. [PMID: 27669410 PMCID: PMC5036963 DOI: 10.7554/elife.16088] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/25/2016] [Indexed: 11/13/2022] Open
Abstract
Corticostriatal afferents can engage parvalbumin-expressing (PV+) interneurons to rapidly curtail the activity of striatal projection neurons (SPNs), thus shaping striatal output. Schemes of basal ganglia circuit dynamics generally consider striatal PV+ interneurons to be homogenous, despite considerable heterogeneity in both form and function. We demonstrate that the selective co-expression of another calcium-binding protein, secretagogin (Scgn), separates PV+ interneurons in rat and primate striatum into two topographically-, physiologically- and structurally-distinct cell populations. In rats, these two interneuron populations differed in their firing rates, patterns and relationships with cortical oscillations in vivo. Moreover, the axons of identified PV+/Scgn+ interneurons preferentially targeted the somata of SPNs of the so-called 'direct pathway', whereas PV+/Scgn- interneurons preferentially targeted 'indirect pathway' SPNs. These two populations of interneurons could therefore provide a substrate through which either of the striatal output pathways can be rapidly and selectively inhibited to subsequently mediate the expression of behavioral routines.
Collapse
Affiliation(s)
- Farid N Garas
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Rahul S Shah
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Eszter Kormann
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Natalie M Doig
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Federica Vinciati
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kouichi C Nakamura
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Matthijs C Dorst
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yoland Smith
- Yerkes National Primate Research Center, Department of Neurology, Emory University, Atlanta, United States.,Udall Center of Excellence for Parkinson's Disease Research, Emory University, Atlanta, United States
| | - Peter J Magill
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Andrew Sharott
- Medical Research Council Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
35
|
Secretagogin affects insulin secretion in pancreatic β-cells by regulating actin dynamics and focal adhesion. Biochem J 2016; 473:1791-803. [PMID: 27095850 PMCID: PMC4901359 DOI: 10.1042/bcj20160137] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/18/2016] [Indexed: 01/03/2023]
Abstract
Secretagogin (SCGN), a Ca2+-binding protein having six EF-hands, is selectively expressed in pancreatic β-cells and neuroendocrine cells. Previous studies suggested that SCGN enhances insulin secretion by functioning as a Ca2+-sensor protein, but the underlying mechanism has not been elucidated. The present study explored the mechanism by which SCGN enhances glucose-induced insulin secretion in NIT-1 insulinoma cells. To determine whether SCGN influences the first or second phase of insulin secretion, we examined how SCGN affects the kinetics of insulin secretion in NIT-1 cells. We found that silencing SCGN suppressed the second phase of insulin secretion induced by glucose and H2O2, but not the first phase induced by KCl stimulation. Recruitment of insulin granules in the second phase of insulin secretion was significantly impaired by knocking down SCGN in NIT-1 cells. In addition, we found that SCGN interacts with the actin cytoskeleton in the plasma membrane and regulates actin remodelling in a glucose-dependent manner. Since actin dynamics are known to regulate focal adhesion, a critical step in the second phase of insulin secretion, we examined the effect of silencing SCGN on focal adhesion molecules, including FAK (focal adhesion kinase) and paxillin, and the cell survival molecules ERK1/2 (extracellular-signal-regulated kinase 1/2) and Akt. We found that glucose- and H2O2-induced activation of FAK, paxillin, ERK1/2 and Akt was significantly blocked by silencing SCGN. We conclude that SCGN controls glucose-stimulated insulin secretion and thus may be useful in the therapy of Type 2 diabetes.
Collapse
|
36
|
Tessier CR. Considering calcium-binding proteins in invertebrates: multi-functional proteins that shape neuronal growth. Neural Regen Res 2016; 11:208-9. [PMID: 27073357 PMCID: PMC4810968 DOI: 10.4103/1673-5374.177711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Charles R Tessier
- Department of Medical and Molecular Genetics, Indiana University School of Medicine-South Bend, South Bend, IN, USA
| |
Collapse
|
37
|
McMahon SM, Chang CW, Jackson MB. Multiple cytosolic calcium buffers in posterior pituitary nerve terminals. ACTA ACUST UNITED AC 2016; 147:243-54. [PMID: 26880753 PMCID: PMC4772375 DOI: 10.1085/jgp.201511525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/06/2016] [Indexed: 01/03/2023]
Abstract
Researchers have measured the ability of nerve terminals to buffer Ca2+ entering in response to electrical activity to better understand plasticity of hormone release. Cytosolic Ca2+ buffers bind to a large fraction of Ca2+ as it enters a cell, shaping Ca2+ signals both spatially and temporally. In this way, cytosolic Ca2+ buffers regulate excitation-secretion coupling and short-term plasticity of release. The posterior pituitary is composed of peptidergic nerve terminals, which release oxytocin and vasopressin in response to Ca2+ entry. Secretion of these hormones exhibits a complex dependence on the frequency and pattern of electrical activity, and the role of cytosolic Ca2+ buffers in controlling pituitary Ca2+ signaling is poorly understood. Here, cytosolic Ca2+ buffers were studied with two-photon imaging in patch-clamped nerve terminals of the rat posterior pituitary. Fluorescence of the Ca2+ indicator fluo-8 revealed stepwise increases in free Ca2+ after a series of brief depolarizing pulses in rapid succession. These Ca2+ increments grew larger as free Ca2+ rose to saturate the cytosolic buffers and reduce the availability of Ca2+ binding sites. These titration data revealed two endogenous buffers. All nerve terminals contained a buffer with a Kd of 1.5–4.7 µM, and approximately half contained an additional higher-affinity buffer with a Kd of 340 nM. Western blots identified calretinin and calbindin D28K in the posterior pituitary, and their in vitro binding properties correspond well with our fluorometric analysis. The high-affinity buffer washed out, but at a rate much slower than expected from diffusion; washout of the low-affinity buffer could not be detected. This work has revealed the functional impact of cytosolic Ca2+ buffers in situ in nerve terminals at a new level of detail. The saturation of these cytosolic buffers will amplify Ca2+ signals and may contribute to use-dependent facilitation of release. A difference in the buffer compositions of oxytocin and vasopressin nerve terminals could contribute to the differences in release plasticity of these two hormones.
Collapse
Affiliation(s)
- Shane M McMahon
- Biophysics PhD Program, Department of Neuroscience, and Physiology PhD Program, University of Wisconsin, Madison, WI 53705
| | - Che-Wei Chang
- Biophysics PhD Program, Department of Neuroscience, and Physiology PhD Program, University of Wisconsin, Madison, WI 53705 Biophysics PhD Program, Department of Neuroscience, and Physiology PhD Program, University of Wisconsin, Madison, WI 53705
| | - Meyer B Jackson
- Biophysics PhD Program, Department of Neuroscience, and Physiology PhD Program, University of Wisconsin, Madison, WI 53705
| |
Collapse
|
38
|
Lee SCS, Weltzien F, Madigan MC, Martin PR, Grünert U. Identification of AⅡ amacrine, displaced amacrine, and bistratified ganglion cell types in human retina with antibodies against calretinin. J Comp Neurol 2015; 524:39-53. [PMID: 26053777 DOI: 10.1002/cne.23821] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 05/29/2015] [Accepted: 06/01/2015] [Indexed: 11/10/2022]
Abstract
Antibodies against calretinin are markers for one type of rod pathway interneuron (AⅡ amacrine cell) in the retina of some but not all mammalian species. The AⅡ cells play a crucial role in night-time (scotopic) vision and have been proposed as a target for optogenetic restoration of vision in retinal disease. In the present study we aimed to characterize the AⅡ cells in human retina. Postmortem human donor eyes were obtained with ethical approval and processed for calretinin immunofluorescence. Calretinin-positive somas in the inner nuclear and the ganglion cell layer were filled with the lipophilic dye DiI. The large majority (over 80%) of calretinin-immunoreactive cells is located in the inner nuclear layer, is immunopositive for glycine transporter 1, and shows the typical morphology of AⅡ amacrine cells. In addition, a small proportion of calretinin-positive cells in the inner nuclear layer and in the ganglion cell layer is glutamic acid decarboxylase-positive and shows the morphology of widefield amacrine cells (stellate, semilunar, and thorny amacrine cells). About half of the calretinin cells in the ganglion cell layer are bistratified ganglion cells resembling the small bistratified (presumed blue-ON/yellow-OFF) and the G17 ganglion cell previously described in primates. We conclude that in human retina, antibodies against calretinin can be used to identify AⅡ amacrine cells in the inner nuclear layer as well as widefield amacrine and small bistratified ganglion cells in the ganglion cell layer.
Collapse
Affiliation(s)
- Sammy C S Lee
- Department of Ophthalmology and Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, University of Sydney, Sydney, NSW, Australia
| | - Felix Weltzien
- Department of Ophthalmology and Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Michele C Madigan
- Department of Ophthalmology and Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Paul R Martin
- Department of Ophthalmology and Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, University of Sydney, Australia
| | - Ulrike Grünert
- Department of Ophthalmology and Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, University of Sydney, Australia
| |
Collapse
|
39
|
Girard F, Venail J, Schwaller B, Celio M. The EF-hand Ca2+-binding protein super-family: A genome-wide analysis of gene expression patterns in the adult mouse brain. Neuroscience 2015; 294:116-55. [DOI: 10.1016/j.neuroscience.2015.02.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 01/13/2023]
|
40
|
Sharma AK, Khandelwal R, Sharma Y, Rajanikanth V. Secretagogin, a hexa EF-hand calcium-binding protein: high level bacterial overexpression, one-step purification and properties. Protein Expr Purif 2015; 109:113-9. [PMID: 25703053 DOI: 10.1016/j.pep.2015.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 12/22/2014] [Accepted: 02/11/2015] [Indexed: 12/20/2022]
Abstract
Secretagogin (SCGN), a hexa EF-hand calcium-binding protein, is highly expressed in the endocrine cells (especially in pancreatic islets) and in restricted neuronal sub-populations, albeit at comparatively low level. Since SCGN is predicted to be a potential neuroendocrine marker in carcinoid tumors of lung and gastrointestinal tract, it is of paramount importance to understand the features of this protein in different environment for assigning its crucial functions in different tissues and under pathophysiological conditions. To score out the limitation of protein for in vitro studies, we report a one-step, high purity and high level bacterial purification of secretagogin by refolding from the inclusion bodies yielding about 40mg protein per litre of bacterial culture. We also report previously undocumented Ca(2+)/Mg(2+) binding and hydrodynamic properties of secretagogin.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India.
| | - Radhika Khandelwal
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | - Yogendra Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
41
|
Wieder N, Fink R, von Wegner F. Exact stochastic simulation of a calcium microdomain reveals the impact of Ca²⁺ fluctuations on IP₃R gating. Biophys J 2015; 108:557-67. [PMID: 25650923 PMCID: PMC4317541 DOI: 10.1016/j.bpj.2014.11.3458] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/04/2014] [Accepted: 11/18/2014] [Indexed: 01/07/2023] Open
Abstract
In this study, we numerically analyzed the nonlinear Ca(2+)-dependent gating dynamics of a single, nonconducting inositol 1,4,5-trisphosphate receptor (IP₃R) channel, using an exact and fully stochastic simulation algorithm that includes channel gating, Ca(2+) buffering, and Ca(2+) diffusion. The IP₃R is a ubiquitous intracellular Ca(2+) release channel that plays an important role in the formation of complex spatiotemporal Ca(2+) signals such as waves and oscillations. Dynamic subfemtoliter Ca(2+) microdomains reveal low copy numbers of Ca(2+) ions, buffer molecules, and IP₃Rs, and stochastic fluctuations arising from molecular interactions and diffusion do not average out. In contrast to models treating calcium dynamics deterministically, the stochastic approach accounts for this molecular noise. We varied Ca(2+) diffusion coefficients and buffer reaction rates to tune the autocorrelation properties of Ca(2+) noise and found a distinct relation between the autocorrelation time τac, the mean channel open and close times, and the resulting IP₃R open probability PO. We observed an increased PO for shorter noise autocorrelation times, caused by increasing channel open times and decreasing close times. In a pure diffusion model the effects become apparent at elevated calcium concentrations, e.g., at [Ca(2+)] = 25 μM, τac = 0.082 ms, the IP₃R open probability increased by ≈20% and mean open times increased by ≈4 ms, compared to a zero noise model. We identified the inactivating Ca(2+) binding site of IP₃R subunits as the primarily noise-susceptible element of the De Young and Keizer model. Short Ca(2+) noise autocorrelation times decrease the probability of Ca(2+) association and consequently increase IPvR activity. These results suggest a functional role of local calcium noise properties on calcium-regulated target molecules such as the ubiquitous IP₃R. This finding may stimulate novel experimental approaches analyzing the role of calcium noise properties on microdomain behavior.
Collapse
Affiliation(s)
- Nicolas Wieder
- Medical Biophysics Unit, Department of Physiology and Pathophysiology, Universität Heidelberg, Heidelberg, Germany.
| | - Rainer Fink
- Medical Biophysics Unit, Department of Physiology and Pathophysiology, Universität Heidelberg, Heidelberg, Germany
| | - Frederic von Wegner
- Medical Biophysics Unit, Department of Physiology and Pathophysiology, Universität Heidelberg, Heidelberg, Germany
| |
Collapse
|
42
|
The Ca2+ sensor protein swiprosin-1/EFhd2 is present in neurites and involved in kinesin-mediated transport in neurons. PLoS One 2014; 9:e103976. [PMID: 25133820 PMCID: PMC4136728 DOI: 10.1371/journal.pone.0103976] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 07/08/2014] [Indexed: 01/10/2023] Open
Abstract
Swiprosin-1/EFhd2 (EFhd2) is a cytoskeletal Ca2+ sensor protein strongly expressed in the brain. It has been shown to interact with mutant tau, which can promote neurodegeneration, but nothing is known about the physiological function of EFhd2 in the nervous system. To elucidate this question, we analyzed EFhd2−/−/lacZ reporter mice and showed that lacZ was strongly expressed in the cortex, the dentate gyrus, the CA1 and CA2 regions of the hippocampus, the thalamus, and the olfactory bulb. Immunohistochemistry and western blotting confirmed this pattern and revealed expression of EFhd2 during neuronal maturation. In cortical neurons, EFhd2 was detected in neurites marked by MAP2 and co-localized with pre- and post-synaptic markers. Approximately one third of EFhd2 associated with a biochemically isolated synaptosome preparation. There, EFhd2 was mostly confined to the cytosolic and plasma membrane fractions. Both synaptic endocytosis and exocytosis in primary hippocampal EFhd2−/− neurons were unaltered but transport of synaptophysin-GFP containing vesicles was enhanced in EFhd2−/− primary hippocampal neurons, and notably, EFhd2 inhibited kinesin mediated microtubule gliding. Therefore, we found that EFhd2 is a neuronal protein that interferes with kinesin-mediated transport.
Collapse
|
43
|
Gáti G, Lendvai D, Hökfelt T, Harkany T, Alpár A. Revival of Calcium-Binding Proteins for Neuromorphology: Secretagogin Typifies Distinct Cell Populations in the Avian Brain. BRAIN, BEHAVIOR AND EVOLUTION 2014; 83:82-92. [DOI: 10.1159/000357834] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 11/19/2022]
|
44
|
Combinatorial analysis of calcium-binding proteins in larval and adult zebrafish primary olfactory system identifies differential olfactory bulb glomerular projection fields. Brain Struct Funct 2014; 220:1951-70. [PMID: 24728871 DOI: 10.1007/s00429-014-0765-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 03/25/2014] [Indexed: 01/05/2023]
Abstract
In the zebrafish (Danio rerio) olfactory epithelium, the calcium-binding proteins (CBPs) calretinin and S100/S100-like protein are mainly expressed in ciliated or crypt olfactory sensory neurons (OSNs), respectively. In contrast parvalbumin and calbindin1 have not been investigated. We present a combinatorial immunohistological analysis of all four CBPs, including their expression in OSNs and their axonal projections to the olfactory bulb in larval and adult zebrafish. A major expression of calretinin and S100 in ciliated and crypt cells, respectively, with some expression of S100 in microvillous cells is confirmed. Parvalbumin and calbindin1 are strongly expressed in ciliated and microvillous cells, but not in crypt cells. Moreover, detailed combinatorial double-label experiments indicate that there are eight subpopulations of zebrafish OSNs: S100-positive crypt cells (negative for all other three CBPs), parvalbumin only, S100 and parvalbumin, parvalbumin and calbindin1, and parvalbumin and calbindin1 and calretinin-positive microvillous OSNs, as well as a major parvalbumin and calbindin1 and calretinin, and minor parvalbumin and calbindin1 and calretinin-only-positive ciliated OSN populations. CBP-positive projections to olfactory bulb are consistent with previous reports of ciliated OSNs projecting to dorsal and ventromedial glomerular fields and microvillous OSNs to ventrolateral glomerular fields. We newly describe parvalbumin-positive fibers to the mediodorsal field which is calretinin free, with its anterior part showing additionally calbindin1-positive fibers, but absence thereof in the posterior part, indicating an origin from microvillous OSNs in both parts. One singular glomerulus (mdG2) exhibits S100 and parvalbumin-positive fibers, apparently originating from all crypt cells plus some microvillous OSNs. Arguments for various olfactory labeled lines are discussed.
Collapse
|
45
|
Neuronal calcium-binding proteins 1/2 localize to dorsal root ganglia and excitatory spinal neurons and are regulated by nerve injury. Proc Natl Acad Sci U S A 2014; 111:E1149-58. [PMID: 24616509 DOI: 10.1073/pnas.1402318111] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Neuronal calcium (Ca(2+))-binding proteins 1 and 2 (NECAB1/2) are members of the phylogenetically conserved EF-hand Ca(2+)-binding protein superfamily. To date, NECABs have been explored only to a limited extent and, so far, not at all at the spinal level. Here, we describe the distribution, phenotype, and nerve injury-induced regulation of NECAB1/NECAB2 in mouse dorsal root ganglia (DRGs) and spinal cord. In DRGs, NECAB1/2 are expressed in around 70% of mainly small- and medium-sized neurons. Many colocalize with calcitonin gene-related peptide and isolectin B4, and thus represent nociceptors. NECAB1/2 neurons are much more abundant in DRGs than the Ca(2+)-binding proteins (parvalbumin, calbindin, calretinin, and secretagogin) studied to date. In the spinal cord, the NECAB1/2 distribution is mainly complementary. NECAB1 labels interneurons and a plexus of processes in superficial layers of the dorsal horn, commissural neurons in the intermediate area, and motor neurons in the ventral horn. Using CLARITY, a novel, bilaterally connected neuronal system with dendrites that embrace the dorsal columns like palisades is observed. NECAB2 is present in cell bodies and presynaptic boutons across the spinal cord. In the dorsal horn, most NECAB1/2 neurons are glutamatergic. Both NECAB1/2 are transported into dorsal roots and peripheral nerves. Peripheral nerve injury reduces NECAB2, but not NECAB1, expression in DRG neurons. Our study identifies NECAB1/2 as abundant Ca(2+)-binding proteins in pain-related DRG neurons and a variety of spinal systems, providing molecular markers for known and unknown neuron populations of mechanosensory and pain circuits in the spinal cord.
Collapse
|
46
|
Kashevarova AA, Nazarenko LP, Skryabin NA, Salyukova OA, Chechetkina NN, Tolmacheva EN, Sazhenova EA, Magini P, Graziano C, Romeo G, Kučinskas V, Lebedev IN. Array CGH analysis of a cohort of Russian patients with intellectual disability. Gene 2014; 536:145-50. [PMID: 24291026 DOI: 10.1016/j.gene.2013.11.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/30/2013] [Accepted: 11/01/2013] [Indexed: 12/20/2022]
Abstract
The use of array comparative genomic hybridization (array CGH) as a diagnostic tool in molecular genetics has facilitated the identification of many new microdeletion/microduplication syndromes (MMSs). Furthermore, this method has allowed for the identification of copy number variations (CNVs) whose pathogenic role has yet to be uncovered. Here, we report on our application of array CGH for the identification of pathogenic CNVs in 79 Russian children with intellectual disability (ID). Twenty-six pathogenic or likely pathogenic changes in copy number were detected in 22 patients (28%): 8 CNVs corresponded to known MMSs, and 17 were not associated with previously described syndromes. In this report, we describe our findings and comment on genes potentially associated with ID that are located within the CNV regions.
Collapse
Key Words
- ABLIM3
- ACAD10
- ADHD
- AFAP1L1
- AGA
- ASTN1
- Array comparative genomic hybridization (array CGH)
- CASP3
- CGH
- CHERISH
- CNS
- CNTN6
- CNV
- Copy number variation (CNV)
- DDX10
- DEAD (Asp-Glu-Ala-Asp) box polypeptide 10
- DECIPHER
- EEG
- EU
- European Union
- FJX1
- GLRA3
- HAND2
- HFE
- ID
- IFN
- IL17B
- ING2
- IQ
- IRF1
- IRF2
- Intellectual disability
- LDLRAD3
- METTL4
- MMP14
- MMSs
- MRI
- NDC80
- NDC80 kinetochore complex component
- NEIL3
- NO
- OMIM
- PCR
- PL CNS
- PON1
- PON2
- PON3
- SBF
- SCGN
- SCRG1
- SET domain binding factor
- SLC1A2
- SLC5A7
- SLC7A7
- SMCHD1
- SUFU
- SWAP switching B-cell complex 70kDa subunit
- SWAP70
- Ski-related novel protein N
- SnoN
- TGFβ
- TNR
- TRIM44
- WAGR
- Wilms tumor, aniridia, genitourinary anomalies and mental retardation syndrome
- actin binding LIM protein family, member 3
- actin filament associated protein 1-like 1
- acyl-CoA dehydrogenase family, member 10
- aspartylglucosaminidase
- astrotactin 1
- attention-deficit hyperactivity disorder
- caspase 3, apoptosis-related cysteine peptidase
- central nervous system
- comparative genomic hybridization
- contactin 6
- copy number variation
- database of chromosomal imbalance and phenotype in humans using ensembl resources
- electroencephalogram
- four jointed box 1 (Drosophila)
- glycine receptor, alpha 3
- grant of European Community's Seventh Framework Programme
- heart and neural crest derivatives expressed 2
- hemochromatosis
- inhibitor of growth family, member 2
- intellectual disability
- intelligence quotient
- interferon
- interferon regulatory factor 1
- interferon regulatory factor 2
- interleukin 17B
- low density lipoprotein receptor class A domain containing 3
- magnetic resonance imaging
- matrix metallopeptidase 14 (membrane-inserted)
- methyltransferase like 4
- microdeletion/microduplication syndromes
- nei endonuclease VIII-like 3 (E. coli)
- nitrogen oxide
- online mendelian inheritance in man
- paraoxonase 1
- paraoxonase 2
- paraoxonase 3
- perinatal lesion of central nervous system
- polymerase chain reaction
- qPCR
- quantitative PCR
- secretagogin, EF-hand calcium binding protein
- solute carrier family 1 (glial high affinity glutamate transporter), member 2
- solute carrier family 5 (sodium/choline cotransporter), member 7
- solute carrier family 7 (amino acid transporter light chain, y+L system), member 7
- stimulator of chondrogenesis 1
- structural maintenance of chromosomes flexible hinge domain containing 1
- suppressor of fused homolog (Drosophila)
- tenascin R
- transforming growth factor beta
- tripartite motif containing 44
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Vaidutis Kučinskas
- Vilnius University, Department of Human and Medical Genetics, Vilnius, Lithuania
| | | |
Collapse
|
47
|
Schwaller B. Calretinin: from a "simple" Ca(2+) buffer to a multifunctional protein implicated in many biological processes. Front Neuroanat 2014; 8:3. [PMID: 24550787 PMCID: PMC3913827 DOI: 10.3389/fnana.2014.00003] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 01/19/2014] [Indexed: 12/30/2022] Open
Abstract
The hexa-EF-hand Ca(2+)-binding protein calretinin (CR) is predominantly expressed in specific neurons of the central and peripheral nervous system. However, CR expression is also observed in non-neuronal cells, e.g., during embryonic development and in mesothelioma cells. Of the 6 EF-hand domains, 5 are functional; the first 4 domains form 2 pairs showing high cooperativity within a pair that results in non-linear modulation of intracellular Ca(2+) signals by CR. EF-hand domain 5 has a low affinity and represents the identified interaction site with CR-binding partners present in mouse cerebellar granule cells. CR binding to other targets including the pore-forming α1 subunit of the Ca(2+) channel Ca V 2.1, as well as to huntingtin indicates additional Ca(2+) sensor functions besides the well-known Ca(2+)-buffering functions. The absence of CR in cerebellar granule cells of CR(-/-) mice results in increased excitability and altered firing of Purkinje cells and promotes cerebellar 160-Hz oscillations impairing motor coordination. The putative role of CR in neuroprotection is still highly discussed. Altogether, CR emerges as a multi-functional protein also associated with development, i.e., cell proliferation, differentiation, and cell death.
Collapse
Affiliation(s)
- Beat Schwaller
- Anatomy, Department of Medicine, University of FribourgFribourg, Switzerland
| |
Collapse
|
48
|
Weltzien F, Dimarco S, Protti DA, Daraio T, Martin PR, Grünert U. Characterization of secretagogin-immunoreactive amacrine cells in marmoset retina. J Comp Neurol 2013; 522:435-55. [DOI: 10.1002/cne.23420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 06/24/2013] [Accepted: 06/25/2013] [Indexed: 11/12/2022]
Affiliation(s)
- Felix Weltzien
- Department of Ophthalmology and Save Sight Institute; University of Sydney; Australia
- Australian Research Council Centre of Excellence in Vision Science; University of Sydney; Australia
| | | | | | - Teresa Daraio
- Department of Ophthalmology and Save Sight Institute; University of Sydney; Australia
| | - Paul R. Martin
- Department of Ophthalmology and Save Sight Institute; University of Sydney; Australia
- Australian Research Council Centre of Excellence in Vision Science; University of Sydney; Australia
- School of Medical Sciences; University of Sydney; Australia
| | - Ulrike Grünert
- Department of Ophthalmology and Save Sight Institute; University of Sydney; Australia
- Australian Research Council Centre of Excellence in Vision Science; University of Sydney; Australia
| |
Collapse
|
49
|
Yamada J, Jinno S. S100A6 (calcyclin) is a novel marker of neural stem cells and astrocyte precursors in the subgranular zone of the adult mouse hippocampus. Hippocampus 2013; 24:89-101. [PMID: 24115312 DOI: 10.1002/hipo.22207] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2013] [Indexed: 01/15/2023]
Abstract
S100A6 (calcyclin), an EF-hand calcium binding protein, is considered to play various roles in the brain, for example, cell proliferation and differentiation, calcium homeostasis, and neuronal degeneration. In addition to some limbic nuclei, S100A6 is distributed in the rostral migratory stream, one of the major neurogenic niches of the adult brain. However, the potential involvement of S100A6 in adult neurogenesis remains unclear. In this study, we aimed to elucidate the role of S100A6 in the other major neurogenic niche, the subgranular zone of the dentate gyrus in the adult mouse hippocampus. Immunofluorescent multiple labeling showed that S100A6 was highly expressed in neural stem cells labeled by sex determining region Y-box 2, brain lipid-binding protein protein and glial fibrillary acidic protein. S100A6+ cells often extended a long process typical of radial glial morphology. In addition, S100A6 was found in some S100β+ astrocyte lineage cells. Interestingly, proliferating cell nuclear antigen was detected in a fraction of S100A6+/S100β+ cells. These cells were considered to be lineage-restricted astrocyte precursors maintaining mitotic potential. On the other hand, S100A6 was rarely seen in neural lineage cells labeled by T-box brain protein 2, doublecortin, calretinin and calbindin D28K. Cell fate-tracing experiment using BrdU showed that the majority of newly generated immature astrocytes were immunoreactive for S100A6, while mature astrocytes lacked S100A6 immunoreactivity. Administration of S100 protein inhibitor, trifluoperazine, caused a reduction in production of S100β+ astrocyte lineage cells, but had no impact on neurogenesis. Overall, our data provide the first evidence that S100A6 is a specific marker of neural stem cells and astrocyte precursors, and may be especially important for generation of astrocytes in the adult hippocampus.
Collapse
Affiliation(s)
- Jun Yamada
- Department of Developmental Molecular Anatomy, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
50
|
Kosaka K, Kosaka T. Secretagogin-containing neurons in the mouse main olfactory bulb. Neurosci Res 2013; 77:16-32. [PMID: 24008127 DOI: 10.1016/j.neures.2013.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 08/09/2013] [Accepted: 08/09/2013] [Indexed: 11/24/2022]
Abstract
Secretagogin (SCGN) is a recently discovered calcium binding protein of the EF hand family. We studied the structural features of SCGN-positive neurons in the mouse main olfactory bulb (MOB). SCGN-positive neurons were localized throughout layers but clustered in the glomerular layer (GL), mitral cell layer (MCL) and granule cell layer (GCL). They were heterogeneous, including numerous juxtaglomerular neurons, granule cells, small to medium-sized neurons in the external plexiform layer (EPL), and a few small cells in the ependymal/subependymal layer. Calretinin and/or tyrosine hydroxylase occasionally colocalized in SCGN-positive juxtaglomerular neurons. Calretinin also frequently colocalized in SCGN-positive EPL and GCL neurons. Morphologically some of juxtaglomerular SCGN-positive neurons were classical periglomerular cells, whereas others were apparently different from those periglomerular cells, although they were further heterogeneous. Some extended one slender process into a glomerulus which passed the glomerulus and further penetrated into another nearby glomeruli, and thus their dendritic processes spanned two or three or more glomeruli. We named this type of juxtaglomerular neurons "transglomerular cells." With the stereological analysis we estimated total number of juxtaglomerular SCGN-positive neurons at about 80,000/single MOB. The present study revealed the diversity of SCGN-positive neurons in the mouse MOB and their particular structural properties hitherto unknown.
Collapse
Affiliation(s)
- Katsuko Kosaka
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | |
Collapse
|