Neuronal Cell Markers
Patima Tanapat (patima dot tanapat at gmail dot com)
Princeton, New Jersey, United States
DOI
//dx.doi.org/10.13070/mm.en.3.196
Date
last modified : 2024-06-27; original version : 2013-06-05
Cite as
MATER METHODS 2013;3:196
Abstract

A comprehensive review of immunohistochemical markers for CNS neuronal cell types.

Introduction

Neurons are the basic signaling components of the brain. Consequently, a fundamental part of any attempt to understand how the brain works as a whole is the investigation of functionally distinct neuronal cell types. Toward this end, immunohistochemical markers have emerged as one of the most valuable tools available to neuroscientists. Using antibodies against various cell components, investigators are able to identify cells expressing a neuronal phenotype and, moreover, collect information regarding their morphological characteristics and expression of specific proteins. Other approaches exists to define neuronal cell types. For example, physiological optical tagging sequencing (PhOTseq) relies on the initial GCaMP calcium imaging labeling of neurons through activation and then the permanent labeling of specific neurons with photoactivatable mCherry (PAmCherry) [6].

Table 1 lists the commonly used markers for all neurons and for neurons with specific neurotransmitters, many of which are discussed in more detail later. Separate articles address other cell types in the brain: glial cells, microglia, and glymphatic and meningeal lymphatic system.

Sym Protein Top three suppliers Reference
BCL11BB cell CLL/lymphoma 11BAbcam ab18465 (140), BioLegend 650601 (1), Cell Signaling Technology 12120 (1)
CALB1calbindin 1MilliporeSigma C9848 (139), SWant 300 (87), Abcam ab82812 (9)
CALB2calbindin 2SWant 6B3 (48), Invitrogen MA1-39562 (9), BD Biosciences 610908 (8)
CNP2',3'-cyclic nucleotide 3' phosphodiesteraseAbcam ab6319 (14), BioLegend 836404 (12), Cell Signaling Technology 5664 (5)
CTIP2 / BCL11BBAF chromatin remodeling complex subunit BCL11BAbcam ab18465 (140), BioLegend 650601 (1), Cell Signaling Technology 12120 (1)
DCXdoublecortinSanta Cruz Biotechnology sc-8066 (17), Abcam ab207175 (6), BD Biosciences 561505 (2) [7]
DLG4 / PSD95discs large MAGUK scaffold protein 4Invitrogen MA1-045 (222), Neuromab 75-028 (100), Cell Signaling Technology 3450 (43) [8, 9]
ELAVL3ELAV like RNA binding protein 3; HuC/DInvitrogen A-21271 (343), Santa Cruz Biotechnology sc-5261 (63) [7, 10]
ELAVL4ELAV like RNA binding protein 4; HuC/DInvitrogen A-21271 (343), Santa Cruz Biotechnology sc-5261 (63) [7, 10]
ENO2enolase 2Dako M0873 (21), Cell Signaling Technology 8171 (5), Santa Cruz Biotechnology sc-271384 (3)
EOMES / TBR2eomesoderminInvitrogen 14-4877-80 (23), Abcam ab183991 (9), Cell Signaling Technology 81493 (1) [11, 12]
FOSFos proto-oncogene, AP-1 transcription factor subunitCell Signaling Technology 2250 (44), Santa Cruz Biotechnology sc-166940 (10), Abcam ab134122 (7) [13, 14]
FOXG1forkhead box G1Abcam ab196868 (3)
GPHNgephyrinSynaptic Systems 147 011 (37), BD Biosciences 612632 (4), Santa Cruz Biotechnology sc-25311 (3) [8]
MAP2microtubule associated protein 2MilliporeSigma M4403 (97), Abcam ab11267 (25), Invitrogen MA5-12823 (23) [15, 16]
MAPTmicrotubule associated protein tauInvitrogen MN1020 (754), Abcam ab80579 (36), Cell Signaling Technology 9632 (17) [15]
NCAM1neural cell adhesion molecule 1BioLegend 318302 (59), BD Biosciences 564488 (56), Beckman Coulter A51078 (27) [17]
NEFLneurofilament lightDako M0762 (37), BioLegend 837904 (26), Cell Signaling Technology 2837 (12)
NEUROD1neuronal differentiation 1Abcam ab60704 (12), Cell Signaling Technology 4373 (3), Santa Cruz Biotechnology sc-46684 (1)
NSEgamma-enolase / enolase 2Dako M0873 (21), Cell Signaling Technology 8171 (5), Santa Cruz Biotechnology sc-271384 (3) [16, 18]
PAX6paired box 6Developmental Studies Hybridoma Bank PAX6 (55), BD Biosciences 561462 (10), Abcam ab78545 (8) [11, 19]
RBFOX3 / NEUNRNA binding fox-1 homolog 3Abcam ab177487 (69), BioLegend 834501 (5), Novus Biologicals NBP1-92693 (4) [14, 20]
S100BS100 calcium binding protein BMilliporeSigma S2532 (64), Invitrogen MA5-12969 (57), Abcam ab52642 (50)
SATB2SATB homeobox 2Abcam ab51502 (63), Santa Cruz Biotechnology sc-81376 (10), Cell Marque 384-R15 (2)
SLC32A1 / VGATsolute carrier family 32 member 1Synaptic Systems 131 011 (30), Invitrogen MA5-24643 (1), Santa Cruz Biotechnology sc-393373 (1) [14, 15]
SYPsynaptophysinMilliporeSigma S5768 (87), Abcam ab8049 (48), Invitrogen MA1-39558 (45) [21, 22]
SYNPRsynaptoporinAbcam ab175224 (1)
TBR1T-box brain transcription factor 1Abcam ab183032 (5)
TUBB3 / TuJ1tubulin beta 3 class IIIBioLegend 801201 (256), MilliporeSigma T8660 (89), Invitrogen 32-2600 (36) [15, 23]
VIMvimentinCell Signaling Technology 5741 (286), Invitrogen MA5-11883 (223), Abcam ab92547 (125)
Cholinergic neurons
ACHEacetylcholinesteraseInvitrogen MA3-042 (15), Abcam ab2803 (5)
CHATcholine acetyltransferaseAbcam ab178850 (3), Santa Cruz Biotechnology sc-55557 (2), LifeSpan Biosciences LS-C41821 (1)
SLC18A3vesicular acetylcholine transporterNeuromab 73-020 (3)
Dopaminergic neurons
DAT1 / SLC6A3dopamine transporterNovus Biologicals NBP2-22164 (13), Abcam ab5990 (4), Santa Cruz Biotechnology sc-32259 (3)
PITX3paired like homeodomain 3Enzo Life Sciences ALX-804-464-C100 (6) [16]
THtyrosine hydroxylaseImmunoStar 22941 (88), Pel-Freez P40101-0 (28), MilliporeSigma T1299 (24) [10, 24]
VMAT2 / SLC18A2solute carrier family 18 member A2OriGene TA500506 (1) [16, 22]
GABAergic neurons
GAD1 / GAD67glutamate decarboxylase 1Abcam ab26116 (20), MilliporeSigma G5419 (3), Cell Signaling Technology 63080 (2) [16]
GAD2glutamate decarboxylase 2Developmental Studies Hybridoma Bank GAD-6 (9), Abcam ab26113 (8), BD Biosciences 559931 (6)
GABBR1gamma-aminobutyric acid type B receptor subunit 1Abcam ab55051 (7) [19]
GABBR2gamma-aminobutyric acid type B receptor subunit 2Neuromab 75-124 (3), Abcam ab75838 (2) [19]
Glutamatergic neurons
GLSglutaminaseAbcam ab156876 (12), MilliporeSigma WH0002744M1 (2), Santa Cruz Biotechnology sc-100533 (1)
GRIN1glutamate ionotropic receptor NMDA type subunit 1Invitrogen 32-0500 (18), Synaptic Systems 114 011 (10), Cell Signaling Technology 5704 (10)
GRIN2Bglutamate ionotropic receptor NMDA type subunit 2BInvitrogen MA1-2014 (15), BD Biosciences 610416 (8), Cell Signaling Technology 4212 (7)
SLC17A6 / VGLUT2vesicular Glu transporter 2Invitrogen MA5-27613 (1), Frontier Institute VGluT2-GP-Af670-1 (1) [14, 25]
SLC17A7 / VGLUT1vesicular Glu transporter 1BioLegend 821301 (3), Abcam ab180188 (1), Santa Cruz Biotechnology sc-377425 (1) [15, 16]
Serotonergic neurons
TPH1tryptophan hydroxylase 1MilliporeSigma T0678 (8), Sino Biological 11931-R145 (6), Abcam ab52954 (3)
TPH2tryptophan hydroxylase 2MilliporeSigma T0678 (8)
Table 2. Neuronal cell markers and top cited antibodies against them among the over 60,000 formal publications in Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.
Neuronal Cell Markers figure 1
Figure 1. Adult rat C6 neuron immunolabeled for NSE (green) and counterstained with DAPI (blue). Bar = 20 µm. Reproduced from Figure 5A of [1].
General Markers of Neurons

Neurons are composed of four distinct morphological areas: a cell body (soma), dendrites, an axon, and presynaptic terminals. The highly specialized structure of these cells enables them to propagate electrical signals, or action potentials, that are the basis for communication between neurons. The soma is the metabolic center of the cell. It houses the nucleus containing the cell’s DNA as well as other organelles. From the soma, two types of processes arise. The first type, called a dendrite, receives incoming information whereas the second type, the axon, conveys outgoing information. Typically, a neuron will have several dendrites. Each of these dendrites can, in turn, contain hundreds to thousands of spines, which are the sites of input from the axons of other neurons. (It should be noted that axons may also synapse upon the soma or axon as well, although this is less frequent.) The axon arises out of a specialized region of the cell body called the axon hillock and is responsible for the propagation of the action potential. It eventually divides into several branches that terminate at synapses. Although the synapse is said to be the point of contact between neurons, the cells do not contact one another physically but are separated by a space termed the synaptic cleft. At the end of each axonal branch is the presynaptic terminal, which contains vesicles filled with neurotransmitter. When an action potential reaches the terminal, the vesicular contents are released into the synaptic cleft allowing chemical communication with the postsynaptic cell.

Neuron-specific enolase (NSE)

NSE, also referred to as gamma-enolase or enolase 2, is a cytosolic protein consistently expressed by mature neurons and cells of neuronal origin (Figure 1). It is a brain-specific glycolytic enzyme that plays an important role in intracellular energy metabolism [26]. During development, NSE levels are very low but then increase during the time frame associated with the morphological and functional maturation of neurons [27].

While NSE is widely used and accepted as a neuronal marker, it is important to note that it has been shown to be expressed in glial cells under certain conditions. Specifically, levels of NSE activity, protein, and mRNA have been detected in cultured oligodendrocytes similar to those observed in cultured neurons [28]. Its expression increases during oligodendrocyte differentiation but is repressed once the cells become mature. Under pathological conditions, NSE is detectable in glial neoplasms and reactive glial cells as well [29].

Neuronal nuclei (NeuN)

In 1992, Mullen et al reported the generation of monoclonal antibodies that recognized a neuronal specific nuclear protein in vertebrates (Figure 2) [30]. This protein, which they called Neuronal Nuclei (NeuN), was detected in most neuronal cell types throughout the central and peripheral nervous systems of adult mice [25]. NeuN, also called Fox-3 or RBFOX3, is involved in the regulation of mRNA splicing [31] and plays a role in regulating neural cell differentiation and nervous system development [31]. The appearance of the NeuN corresponds temporally to the withdrawal of neuronal cells from the cell cycle and/or with the initiation of terminal differentiation. The protein is detectable in both embryonic and adult neurons with the exception of cerebellar Purkinje cells, olfactory bulb mitral cells, retinal photoreceptor cells, and dopaminergic neurons in the substantia nigra [32, 33].

Neuronal Cell Markers figure 2
Figure 2. Example of NeuN-labeled neurons in mouse neocortex. Reproduced from Figure 3B of [2].

Fluorescence-activated nuclear sorting of NeuN-stained (and often counterstained by propidium iodide) can isolate neuronal nuclei [34, 35] and is commonly used in single-neuron studies like snmC-seq [36] or single-nucleus RNA-seq [37].

Microtubule-associated protein 2 (MAP-2)

MAP-2 is a neuron-specific cytoskeletal protein that is used as a marker of neuronal phenotype [38]. It is expressed in the vertebrate nervous system in both embryonic and adult tissues. Its expression is weak in neuronal precursors but becomes pronounced later (approximately one day after expression of neuron-specific tubulin isoform βIII) [39]. Studies in rats have indicated the existence of at least three isoforms of MAP-2 including 2a, 2b, and 2c. MAP-2c appears to be expressed exclusively during early development and only in axons. MAP-2c appears to be then replaced by MAP2a over the course of maturation. MAP2b, in contrast, has been found to be expressed throughout life.

MAP-2 protein is thought to be involved in microtubule assembly, acting to stabilize microtubule growth by crosslinking with intermediate filaments and other microtubules. Specifically, it may play a role in determining and stabilizing dendritic shape during neuronal development and is accordingly observed only within the dendritic arbor [40]. In general, its expression appears to be confined to neurons and reactive astrocytes [41]. Zullo JM et al used MAP2 as the marker of cortical neurons in the immunohistochemical labelling of coronal mouse brain sections [42]. Zeng Q et al identified differentiated neurons and astrocytes in primary cultures of mouse cortical neurons with MAP2 and GFAP labelling [25]. Velasco S et al identified neurons from brain organoids with a MAP-2 marker [9]. Dominy SS et al studied the co-localization of Porphyromonas gingivalis RgpB protein with hippocampal neurons from Alzheimer's disease patients using MAP2 as a neuronal marker [43].

ßIII Tubulin (TuJ1)

Tubulin beta III (TUBB3), also called TuJ1, is a tubulin thought to be involved specifically during differentiation of neuronal cell types [44]. Tubulins are a major building block of microtubules, which are structural components of the cytoskeleton attributed roles in cell structure maintenance, mitosis, meiosis, and intracellular transport among others. Accordingly, immunohistochemical staining of TuJ1 is found in the cell bodies, dendrites, axons, and axonal terminations of immature neurons. One of the significant advantages associated with the use of immunohistochemical detection of TuJ1 is the degree to which it reveals the fine details of axons and terminations (Figure 3). In fact, it has been found to be useful in the detection of injury-related alterations in the composition of the somatic cytoskeleton [45]. Although a form of ß-tubulin is also found in glial cells, it is not recognized by antibodies against TuJ1 [45]. Gur-Cohen S et al labelled dermal neuronal fibers with a TuJ1 antibody [46], so did Correa-Gallegos D et al in their wound healing study [47]. Maniatis S et al used TUBB3 staining to outline motor neuron somata [48].

Neuronal Cell Markers figure 3
Figure 3. Doublecortin-labeled cells in the dentate gyrus of adult mice. Reproduced from Figure 5 of [3].

Another tubulin gene TUBA4A has also been used as a neuronal marker. Hu CK et al identified neuronas in killifish embryos with Sigma antibody T7451 (clone 6-11B-1) against acetylated tubulin [49].

Doublecortin (DCX)

DCX is widely expressed by migrating neurons and is observed in the earliest stages of neuronal development [50] and is considered as a marker for developing and adult neurogenesis in the central nervous system [51, 52]. It is a microtubule-associated protein that is expressed in post-mitotic neurons and has a suggested role in the growth of neuronal processes at the leading edge of the cell [53]. Consistent with this role, DCX immunostaining appears most intense at extremities of neurites and continues into proximal regions of growth cones, but not the tips.

c-fos

Activation of c-fos has been used as a marker for neuronal activation [13, 14]. Bai L et al observed increased Fos expression in specific brain regions after the stimulation of various vagal sensory neurons [54].

Markers for Neurons with Specific Neurotransmitters
Glutamatergic neurons

Glutamatergic neurons can be distinguished by the vesicular glutamate transporterse, SLC17A7 / vGluT1 and SLC17A6 / vGluT2, membrane proteins responsible for the uptake of glutamate into synaptic vesicles at presynaptic nerve terminals. A third transporter, SLC17A8 / vGluT3, is expressed in cholinergic and serotoninergic neurons [55]. vGluT1 is identified in cerebral and cerebellar cortices and hippocampus and in the terminals of auditory nerves [56]. vGLUT2 is mostly in diencephalon and rhombencephalon neurons. Both vGluT1 and vGLUT2 are concentrated in the synaptic vesicles in the terminals of the axon of asymmetric synapses.

Members of the glutamate-gated ion channel superfamily such as NMDAR1 / GluN1 and NMDAR2B / GRIN2B / GluN2B also serve as markers for glutamatergic neurons. Eight isoforms of the GluN1 gene have been identified, which vary in the number of N-terminal and C-terminal domains, while their patterns of expression are distinct in time and space in human and rodents [57-59]. NMDAR1 is identified in glutamatergic neurons in cortical plate [60] and dispersed heterogeneously within the cytoplasm and scattered in apical dendrites.

Mitochondrial glutaminase protein, encoded by GLS gene, catalyzes the hydrolysis of glutamine to glutamate in brain and kidney.

GABAergic neurons

Transporters, receptors and enzymes for gamma-aminobutyric acid, a primary neurotransmitter, serve as markers for GABAergic neurons. Zeitler B et al used Darpp32, phosphodiesterase 10a (Pde10a), dopamine receptors D1

(Drd1a) and D2 (Drd2) as markers for medium spiny neurons [19].

GABA transporter 1

GABA in synaptic cleft is eliminated by sodium- and chloride-dependent GABA transporter 1 / GAT1 / solute carrier family 6 member 1 gene /SLC6A1. The protein consists of twelve transmembrane domains, and both C- and N-terminals are intracellularly situated [61]. It is highly expressed in brain and liver.

GABA B receptor 1

GABA B receptor subunit 1 (GABBR1) and 2 form a heterodimer, serving as the receptor for GABA. Multiple alternatively spliced isoforms for GABBR1 exist. It is highly expressed in brain and also expressed broadly in spleen and other tissues.

GABA B receptor 2

The second component of GABA type B receptor is gamma-aminobutyric acid type B receptor subunit 2 protein (GBR2 / GABAB2). Its expression is restricted to brain.

GAD67 / GAD1

There are two isoforms of functional GAD67 / GAD1, which catalyze the production of gamma-aminobutyric acid from L-glutamic acid. GAD67 is often used as a marker for GABAergic neurons along with GABA [62]. The gene is highly expressed in brain and also expressed in kidney, testis and other tissues.

At least eight more alternatively spliced variants are translated from this gene. Their distribution is region-dependent, though not all of them can be distinguished by immunohistochemistry [63].

GAD65 /GAD2

Glutamate decarboxylase 2 / GAD65 / GAD2 catalyzes the production of gamma-aminobutyric acid from L-glutamic acid. It expression is restricted to brain.

Dopaminergic neurons
Tyrosine hydroxylase / TH

Tyrosine hydroxylase is the rate-limiting enzyme during the synthesis of the catecholamines and is involved in the conversion of tyrosine to dopamine. The gene is highly expressed in adrenal glands. There are four major human TH isoforms; all are present in neurons [64].

Immunohistochemistry for TH has been an important tool for investigation of Parkinson’s disease, a condition characterized by movement disorder associated with the loss of the dopaminergic cells of the substantia nigra [24]. In post mortem studies, TH immunohistochemistry has been used to quantify the degree of dopaminergic cell loss in Parkinson’s patients [65]. To determine whether this type of loss might be mitigated by intervention, TH immunochemistry has been used as a means of examining the protective effects of various experimental manipulations in animal models of Parkinson’s disease [66], or confirm whether dopaminergic/adrenergic neurons were involved in specific processes [51, 67, 68]. Y Shwartz et al labelled sympathetic neurons innervating hair follicles with tyrosine hydroxylase [22].

Dopamine transporter / DAT1

Sodium-dependent dopamine transporter acts in presynaptic terminals, terminating dopamine effect by sodium-dependent reuptake. DAT-positive staining is predominantly associated with intracellular membranes (in perikarya and dendrites) or plasma membrane (in unmyelinated axons or medium- to small-diameter dendrites) [69]. Intracellular localization was variable, being detected in axons and varicosities or perycaria and dendrites [70].

GIRK2

GIRK2 (G protein-activated inward rectifier potassium channel 2 ) /KCNJ6 is a member of potassium inwardly rectifying channel subfamily J. It is highly expressed brain and also expressed in stomach and other tissues.

GIRK2 is considered as a mature DA neurons marker, as it is detected at day 50 in in vitro differentiation studies of human [71]. Almost all of TH-immunopositive neuronal cells are also GIRK2 positive in substantia nigra, but only 50-60% of these cells in ventral tegmental area are stained with GIRK2 [72].

Serotonergic neurons

This type of neurons is distinct phenotypically by proteins involved in serotonin (5-hydroxytryptamine) synthesis (tryptophan hydroxylase) and functioning (serononine transporter).

Tryptophan hydroxylases

Tryptophan hydroxylases (tryptophan hydroxylase 1 and 2) catalyze the first and rate limiting step in the biosynthesis of serotonin.

Serotonin transporter

Serotonin transporter recycles the neurotransmitter serotonin from synaptic spaces into presynaptic neurons and terminates its action of serotonin.

Pet1

The protein Pet1, also known as FEV transcription factor, is exclusively expressed in serotonergic neurons in the brain. It is also expressed in duodenum, stomach and other tissues.

Cholinergic neurons

Nerve cells which use acetylcholine as main neurotransmitter are called cholinergic neurons. Thus, proteins applied as their markers are involved acetylcholine metabolism.

Choline acetyltransferase / CHAT

Choline acetyltransferase catalyzes the biosynthesis of the neurotransmitter acetylcholine. It is expressed in the vast majority of cholinergic neurons, ChAT immunoreactivity is used as a biomarker of cognitive decline in various neurodegenerative disorders. Its detection has been a significant tool for investigating the neurological changes typical of Alzheimer’s Disease (AD), which is associated with a substantial loss of basal forebrain cholinergic neurons. Post-mortem studies have used ChAT immunohistochemistry to reveal decreases in the density of fibers and axon varicosities of ChAT-positive neurons in the superior frontal cortex and amygdala of cognitively impaired patients [73, 74]. Furthermore, in animal models, immunohistochemistry has been used to evaluate the capacity of experimental manipulations or transgenes to alter the number of ChAT-positive neurons in the medial septum and the Broca region [75, 76] or striatum [77]. In regions including the cingulate, motor, and sensory cortices, and the basal forebrain, immunolabeling has also been applied to evaluate cholinergic neurons for disruption of the ChAT-ir fiber network and for changes in overall morphology [78, 79].

Vesicular acetylcholine transporter

Vesicular acetylcholine transporter, also called solute carrier family 18 member A3, or SLC18A3, is the member of vesicular amine proton-dependent transporters. It transports acetylcholine into secretory vesicles for release into the extracellular space.

Acetylcholinesterase / ACHE

Acetylcholinesterase hydrolyzes acetylcholine at neuromuscular junctions and brain cholinergic synapses, and thus terminates signal transmission.

Additional Neuronal Markers

In addition to the markers that have been described above, there are a significant number of other neuron-associated markers (Table 1). Y Shwartz et al labeled the apposition between sympathetic nerve fibers and hair follicle stem cells with the pre-synaptic marker synaptotagmin, synaptophysin, or VMAT2 [22]. Zeng Q et al labeled presynaptic vesicles with synaptobrevin 1 in tissue sections of metastatic lesions and adjacent normal brain [25]. Abdo H et al detected unmyelinated nerve endings in the skin with PGP9.5 [80]. Brn2 [16, 81], Tbr1 [81, 82], and Ctip2 [81, 82] serve as cortical lineage neuronal markers. Velasco S et al marked forebrain progenitors with FOXG1, dorsal forebrain progenitors with EMX1 and PAX6, intermediate progenitor cells with TBR2, post-mitotic projection neurons with TBR1, corticofugal projection neurons with CTIP2, callosal projection neurons with SATB2 respectively, in brain organoids [9]. Kapogiannis D et al ascertained L1CAM-enriched plasma exosomes as of neuronal origin using synaptophysin and L1CAM [17]. Mauffrey P et al used polysialylated-neural cell adhesion molecule (PSA-NCAM) and internexin as markers of neural progenitors [51]. L Cantuti-Castelvetri et al detected olfactory neuronal progenitors with OLIG2 [23].

MarkerLocalizationFunctionAssociated Neurons
Polysialic acid-neural cell adhesion molecule (PSA-NCAM)plasma membraneneural cell adhesion molecule thought to play a role in regulating cell shape, growth or migration during development; thought to be involved in activity-induced synaptic plasticity in adulthood [83] expressed in populations of immature neurons
Neurogenic Differentiation 1 (NeuroD or Beta2)cytoplasm and nucleustranscription factor expressed in parts of the brain; involved in differentiation of nervous system; required for postnatally born microneurons to differentiate properly, absence results in cell death [84] expressed in populations of immature neurons
Taudistal portions of axons, not present in dendrite*highly soluble microtubule-associated protein mostly found in neurons compared to non-neuronal cells [85] ; modulates stability of the cytoskeleton and provides axonal flexibility [86] abundant in CNS neurons; also expressed at low levels in CNS astrocytes and oligodendrocytes
Calbindin-D28kcell bodies, dendrites, and spines; cytoplasm matrixCalcium-binding protein; plays a role in fast calcium buffering [87] Purkinje neurons [88] ; expressed in neuroendocrine cells
Calretinincytosol29kDa protein with 58% homology to calbindin-D28; first calcium-binding protein expressed in the vertebrate central nervous system [89] widely distributed in different neuronal populations including a subset of cortical interneurons [62, 77]
Neurofilament Protein (NFP)axonsneuronal cytoskeletal protein; intermediate filament; provides structural support for axons and regulates axon diameter [26, 90] widely distributed in different neuronal populations; levels are relatively low in some neurons
synaptoporin (SYNPR, SPO)mossy fiberpresynaptic channel protein [91-93] subpopulations of mouse hippocampal neurons [94]
Table 1. Additional commonly used immunohistochemical markers of CNS neurons.
Additional Discussion about Neuronal Cell Types

The first individual to recognize the vast diversity that exists across the neuron population was the founding father of neurobiology, Santiago Ramon y Cajal. Using Camillo Golgi’s method of silver impregnation, he observed the highly distinct morphological features of individual neurons. In doing so, he found that the cells comprising the brain ranged greatly in both size and shape. Whereas some neurons such as granule cells exhibited relatively simple processes arising from a small cell body, others like the cerebellar Purkinje cells were characterized by very florid and complex projections.

Neuronal Cell Markers figure 5
Figure 5. Confocal images of cells double-stained for nestin (green) and βIII-tubulin (red) in the adult rat brain. Reproduced from Figure 6A of [5].

Since Cajal’s initial discovery, scientists have been able to distinguish hundreds of different neuronal cell types. However, a unified system of classification remains elusive. This is, in part, due to the fact that the names assigned to different cells tend to vary in their degree of descriptive accuracy. Furthermore, categories of neurons often overlap, the result being that a particular cell type may be associated with several different names. In general, though, neurons can be classified according to criteria that include morphology, projection patterns, electrophysiological properties, and biochemical characteristics.

Examples of classifications based upon morphology (e.g., size, soma shape, and dendritic branch patterns) are arguably the most commonly found in the scientific literature. While morphology provides an obvious and accessible means of characterization, the deeper rationale for its application lies in its functional significance. Because the shape of a neuron is determined by the input it receives and the targets onto which it projects, the neuronal structure is a direct reflection of the cell’s underlying connectivity, and therefore also of its function. Take, for example, the shape of climbing fibers and ascending branches of granule cell axons in the cerebellum. Here, the axonal shape is determined by specific connections to target Purkinje cell dendrites in which the axonal arbor makes numerous contacts with a single dendrite [95]. Similarly, the type of information revealed by morphology can be illustrated by examining the classical pyramidal cell. The term pyramidal cell refers broadly to cells that are characterized by a large, triangular soma with distinct sets of apical and basal dendrites. This structural organization is suggestive of the role these cells play in the incorporation of varied inputs from different cellular layers into an integrated message that is communicated to another brain region [96].

Another important structural distinction often made is based upon the axonal projections of a neuron. Cells that extend an axon beyond the discrete population of neurons to which they belong (i.e., nucleus) are referred to as projection, or principal neurons, whereas those that make synaptic contact only within their nucleus are called interneurons, or intrinsic neurons. It should be emphasized that these terms refer only to the axonal projection, and not to the source of inputs. Thus, a projection neuron may receive only local inputs and, conversely, an intrinsic neuron may receive input from another nucleus. Identification of these patterns of connectivity is important as it provides insight regarding the flow of information processing that underlies the function associated with these cell populations. Indeed, this key concept underlies the significant effort that has recently been directed toward the compilation of comprehensive maps of the neural connections in the brain, called “connectomes” [97].

Neuronal cell types may also be distinguished by their electrophysiological properties. For example, neocortical neurons, which exhibit significant differences in their action potential response patterns, are typically classified as regular-spiking (RS), fast-spiking (FS), and intrinsically bursting (IB) cells. Regular-spiking cells adapt strongly in response to maintained stimuli whereas FS cells sustain high firing frequency with little or no adaptation and IB cells generate clusters of spikes singly or repetitively [98, 99]. The diversity in intrinsic firing patterns of these cells is significant in that it represents an additional level at which the response of these cells to stimuli can be modulated.

With regard to the distinguishing criteria covered thus far, immunohistochemistry can be used to provide information about morphology and, to some extent, axonal projections of different neuron populations. However, the real strength of the methodology in the context of neuronal phenotyping lies in its potential to help distinguish cells based upon biochemical characteristics. Indeed, as will be described in the following sections, the identification of neurons categorized by an association with a particular neurotransmitter system or the expression of a specific gene or protein is a frequently employed strategy.

Additional Discussion on Immunohistochemistry

While Golgi staining remains a useful tool for identifying neuronal cell types, the technique is associated with certain drawbacks. Probably the most significant of these is that, even when successful, the staining tends to be variable. Although complete impregnation of the dendritic tree can be achieved, axonal branches and fine distal terminal arborizations are difficult to thoroughly label [100]. Additionally, it is estimated that, for reasons that are unknown, impregnation only occurs in 1-10% of cells [101, 102]. While this aspect of the protocol is exactly what enabled Cajal’s original discovery that neurons were in fact independent entities, it makes targeting specific cells difficult and also increases the quantity of tissue required for processing to achieve appropriate sample sizes.

With the advent of immunohistochemical strategies for identifying cell types in the 1970s, investigators were able to circumvent some of the technical issues associated with older methods such as Golgi staining. By that time, researchers had begun using immunohistochemistry to identify neurons containing the monoamine-synthesizing enzymes tyrosine hydroxylase (TH), dopamine-beta-hydroxylase (DBH), and tryptophan hydroxylase (TrH) [103, 104, 104]. Nevertheless, it was subsequent to reports describing the use of the brain endolases: neuron-specific enolase (NSE) and non-neuronal enolase (NNE), as specific markers of neuronal and glial cells, respectively, that the use of immunohistochemistry for identifying neuronal cells really began to flourish [105]. Other studies followed describing immunolabeling of a range of different antigens differentially expressed among neuronal cell types including NGF-Inducible Large External glycoprotein (NILE-GF) [106], choline acetyltransferase [107], parvalbumin [10, 62], calretinin [62, 77], somatostatin [62], neuronal peptide Y [62] and neurofilament protein [108, 109] as markers. While not all of these have continued to be widely utilized in this context, the impact of the methodological strategy has remained important.

More recently, molecular approaches have been developed that are helping to elucidate neuronal characteristics. Methods that utilize transcriptional modulators and site-specific recombinases to label specific neuronal populations have become available to investigators [110]. However, immunohistochemistry has managed to maintain its status as a staple methodology for identifying neuronal cell types because it is relatively low in cost to perform, requires very little specialized equipment, and uses reagents are that are widely available. Moreover, routine microscopic methods can be used to collect data and, depending on the method of visualization, immunolabeled tissue may be preserved for reference.

References
  1. Portiansky E, Nishida F, Barbeito C, Gimeno E, Goya R. Increased number of neurons in the cervical spinal cord of aged female rats. PLoS ONE. 2011;6:e22537 pubmed publisher
  2. Peter M, Bathellier B, Fontinha B, Pliota P, Haubensak W, Rumpel S. Transgenic mouse models enabling photolabeling of individual neurons in vivo. PLoS ONE. 2013;8:e62132 pubmed publisher
  3. Merz K, Lie D. Evidence that Doublecortin is dispensable for the development of adult born neurons in mice. PLoS ONE. 2013;8:e62693 pubmed publisher
  4. Cerbai F, Lana D, Nosi D, Petkova Kirova P, Zecchi S, Brothers H, et al. The neuron-astrocyte-microglia triad in normal brain ageing and in a model of neuroinflammation in the rat hippocampus. PLoS ONE. 2012;7:e45250 pubmed publisher
  5. Hendrickson M, Rao A, Demerdash O, Kalil R. Expression of nestin by neural cells in the adult rat and human brain. PLoS ONE. 2011;6:e18535 pubmed publisher
  6. Lee D, Kume M, Holy T. Sensory coding mechanisms revealed by optical tagging of physiologically defined neuronal types. Science. 2019;366:1384-1389 pubmed publisher
  7. Pellegrini L, Bonfio C, Chadwick J, Begum F, Skehel M, Lancaster M. Human CNS barrier-forming organoids with cerebrospinal fluid production. Science. 2020;: pubmed publisher
  8. Yu K, Lin C, Hatcher A, Lozzi B, Kong K, Huang Hobbs E, et al. PIK3CA variants selectively initiate brain hyperactivity during gliomagenesis. Nature. 2020;578:166-171 pubmed publisher
  9. Velasco S, Kedaigle A, Simmons S, Nash A, Rocha M, Quadrato G, et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature. 2019;: pubmed publisher
  10. Spix T, Nanivadekar S, Toong N, Kaplow I, Isett B, Goksen Y, et al. Population-specific neuromodulation prolongs therapeutic benefits of deep brain stimulation. Science. 2021;374:201-206 pubmed publisher
  11. Barnat M, Capizzi M, Aparicio E, Boluda S, Wennagel D, Kacher R, et al. Huntington's disease alters human neurodevelopment. Science. 2020;369:787-793 pubmed publisher
  12. Boss B. Specialty nursing. J Neurosci Nurs. 1989;21:213-5 pubmed
  13. Gong R, Xu S, Hermundstad A, Yu Y, Sternson S. Hindbrain Double-Negative Feedback Mediates Palatability-Guided Food and Water Consumption. Cell. 2020;182:1589-1605.e22 pubmed publisher
  14. Tansley S, Gu N, Guzmán A, Cai W, Wong C, Lister K, et al. Microglia-mediated degradation of perineuronal nets promotes pain. Science. 2022;:eabl6773 pubmed publisher
  15. Haukedal H, Corsi G, Gadekar V, Doncheva N, Kedia S, de Haan N, et al. Golgi fragmentation - One of the earliest organelle phenotypes in Alzheimer's disease neurons. Front Neurosci. 2023;17:1120086 pubmed publisher
  16. Qian H, Kang X, Hu J, Zhang D, Liang Z, Meng F, et al. Reversing a model of Parkinson's disease with in situ converted nigral neurons. Nature. 2020;582:550-556 pubmed publisher
  17. Kapogiannis D, Mustapic M, Shardell M, Berkowitz S, Diehl T, Spangler R, et al. Association of Extracellular Vesicle Biomarkers With Alzheimer Disease in the Baltimore Longitudinal Study of Aging. JAMA Neurol. 2019;: pubmed publisher
  18. Lee J, Yang D, Goulbourne C, Im E, Stavrides P, Pensalfini A, et al. Faulty autolysosome acidification in Alzheimer's disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques. Nat Neurosci. 2022;: pubmed publisher
  19. Zeitler B, Froelich S, Marlen K, Shivak D, Yu Q, Li D, et al. Allele-selective transcriptional repression of mutant HTT for the treatment of Huntington's disease. Nat Med. 2019;25:1131-1142 pubmed publisher
  20. Choi C, Park J, Kim H, Chang K, Park J, MIN K. DSCR1 upregulation enhances dural meningeal lymphatic drainage to attenuate amyloid pathology of Alzheimer's disease. J Pathol. 2021;255:296-310 pubmed publisher
  21. Nakamura T, Oh C, Liao L, Zhang X, Lopez K, Gibbs D, et al. Noncanonical transnitrosylation network contributes to synapse loss in Alzheimer's disease. Science. 2021;371: pubmed publisher
  22. Shwartz Y, Gonzalez Celeiro M, Chen C, Pasolli H, Sheu S, Fan S, et al. Cell Types Promoting Goosebumps Form a Niche to Regulate Hair Follicle Stem Cells. Cell. 2020;: pubmed publisher
  23. Cantuti Castelvetri L, Ojha R, Pedro L, Djannatian M, Franz J, Kuivanen S, et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science. 2020;: pubmed publisher
  24. Cole T, Zhao H, Collier T, Sandoval I, Sortwell C, Steece Collier K, et al. α-Synuclein antisense oligonucleotides as a disease-modifying therapy for Parkinson's disease. JCI Insight. 2021;6: pubmed publisher
  25. Zeng Q, Michael I, Zhang P, Saghafinia S, Knott G, Jiao W, et al. Synaptic proximity enables NMDAR signalling to promote brain metastasis. Nature. 2019;573:526-531 pubmed publisher
  26. Iwanaga T, Takahashi Y, Fujita T. Immunohistochemistry of neuron-specific and glia-specific proteins. Arch Histol Cytol. 1989;52 Suppl:13-24 pubmed
  27. Marangos P, Schmechel D, Parma A, Goodwin F. Developmental profile of neuron-specific (NSE) and non-neuronal (NNE) enolase. Brain Res. 1980;190:185-93 pubmed
  28. Sensenbrenner M, Lucas M, Deloulme J. Expression of two neuronal markers, growth-associated protein 43 and neuron-specific enolase, in rat glial cells. J Mol Med (Berl). 1997;75:653-63 pubmed
  29. Vinores S, Marangos P, Bonnin J, Rubinstein L. Immunoradiometric and immunohistochemical demonstration of neuron-specific enolase in experimental rat gliomas. Cancer Res. 1984;44:2595-9 pubmed
  30. Mullen R, Buck C, Smith A. NeuN, a neuronal specific nuclear protein in vertebrates. Development. 1992;116:201-11 pubmed
  31. Kim K, Adelstein R, Kawamoto S. Identification of neuronal nuclei (NeuN) as Fox-3, a new member of the Fox-1 gene family of splicing factors. J Biol Chem. 2009;284:31052-61 pubmed publisher
  32. Cannon J, Greenamyre J. NeuN is not a reliable marker of dopamine neurons in rat substantia nigra. Neurosci Lett. 2009;464:14-7 pubmed publisher
  33. Kumar S, Buckmaster P. Neuron-specific nuclear antigen NeuN is not detectable in gerbil subtantia nigra pars reticulata. Brain Res. 2007;1142:54-60 pubmed
  34. Labonte B, Abdallah K, Maussion G, Yerko V, Yang J, Bittar T, et al. Regulation of impulsive and aggressive behaviours by a novel lncRNA. Mol Psychiatry. 2020;: pubmed publisher
  35. Lee M, Siddoway B, Kaeser G, Segota I, Rivera R, Romanow W, et al. Somatic APP gene recombination in Alzheimer's disease and normal neurons. Nature. 2018;563:639-645 pubmed publisher
  36. Luo C, Keown C, Kurihara L, Zhou J, He Y, Li J, et al. Single-cell methylomes identify neuronal subtypes and regulatory elements in mammalian cortex. Science. 2017;357:600-604 pubmed publisher
  37. Lake B, Ai R, Kaeser G, Salathia N, Yung Y, Liu R, et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science. 2016;352:1586-90 pubmed publisher
  38. Izant J, McIntosh J. Microtubule-associated proteins: a monoclonal antibody to MAP2 binds to differentiated neurons. Proc Natl Acad Sci U S A. 1980;77:4741-5 pubmed
  39. Dehmelt L, Halpain S. The MAP2/Tau family of microtubule-associated proteins. Genome Biol. 2005;6:204 pubmed
  40. Huber G, Matus A. Differences in the cellular distributions of two microtubule-associated proteins, MAP1 and MAP2, in rat brain. J Neurosci. 1984;4:151-60 pubmed
  41. Geisert E, Johnson H, Binder L. Expression of microtubule-associated protein 2 by reactive astrocytes. Proc Natl Acad Sci U S A. 1990;87:3967-71 pubmed
  42. Zullo J, Drake D, Aron L, O Hern P, Dhamne S, Davidsohn N, et al. Regulation of lifespan by neural excitation and REST. Nature. 2019;574:359-364 pubmed publisher
  43. Dominy S, LYNCH C, Ermini F, Benedyk M, Marczyk A, Konradi A, et al. Porphyromonas gingivalis in Alzheimer's disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv. 2019;5:eaau3333 pubmed publisher
  44. Lee M, Tuttle J, Rebhun L, Cleveland D, Frankfurter A. The expression and posttranslational modification of a neuron-specific beta-tubulin isotype during chick embryogenesis. Cell Motil Cytoskeleton. 1990;17:118-32 pubmed
  45. Geisert E, Frankfurter A. The neuronal response to injury as visualized by immunostaining of class III beta-tubulin in the rat. Neurosci Lett. 1989;102:137-41 pubmed
  46. Gur Cohen S, Yang H, Baksh S, Miao Y, Levorse J, Kataru R, et al. Stem cell-driven lymphatic remodeling coordinates tissue regeneration. Science. 2019;366:1218-1225 pubmed publisher
  47. Correa Gallegos D, Jiang D, Christ S, Ramesh P, Ye H, Wannemacher J, et al. Patch repair of deep wounds by mobilized fascia. Nature. 2019;576:287-292 pubmed publisher
  48. Maniatis S, Aijö T, Vicković S, Braine C, Kang K, Mollbrink A, et al. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science. 2019;364:89-93 pubmed publisher
  49. Hu C, Wang W, Brind Amour J, Singh P, Reeves G, Lorincz M, et al. Vertebrate diapause preserves organisms long term through Polycomb complex members. Science. 2020;367:870-874 pubmed publisher
  50. Francis F, Koulakoff A, Boucher D, Chafey P, Schaar B, Vinet M, et al. Doublecortin is a developmentally regulated, microtubule-associated protein expressed in migrating and differentiating neurons. Neuron. 1999;23:247-56 pubmed
  51. Mauffrey P, Tchitchek N, Barroca V, Bemelmans A, Firlej V, Allory Y, et al. Progenitors from the central nervous system drive neurogenesis in cancer. Nature. 2019;: pubmed publisher
  52. Choi S, Bylykbashi E, Chatila Z, Lee S, Pulli B, Clemenson G, et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer's mouse model. Science. 2018;361: pubmed publisher
  53. Friocourt G, Koulakoff A, Chafey P, Boucher D, Fauchereau F, Chelly J, et al. Doublecortin functions at the extremities of growing neuronal processes. Cereb Cortex. 2003;13:620-6 pubmed
  54. Bai L, Mesgarzadeh S, Ramesh K, Huey E, Liu Y, Gray L, et al. Genetic Identification of Vagal Sensory Neurons That Control Feeding. Cell. 2019;179:1129-1143.e23 pubmed publisher
  55. Gras C, Herzog E, Bellenchi G, Bernard V, Ravassard P, Pohl M, et al. A third vesicular glutamate transporter expressed by cholinergic and serotoninergic neurons. J Neurosci. 2002;22:5442-51 pubmed
  56. Oliveira A, Hydling F, Olsson E, Shi T, Edwards R, Fujiyama F, et al. Cellular localization of three vesicular glutamate transporter mRNAs and proteins in rat spinal cord and dorsal root ganglia. Synapse. 2003;50:117-29 pubmed
  57. Paoletti P, Bellone C, Zhou Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14:383-400 pubmed publisher
  58. Babenko V, Bragin A, Chadaeva I, Markel A, Orlov Y. [Differential alternative splicing in brain regions of rats selected for aggressive behavior]. Mol Biol (Mosk). 2017;51:870-880 pubmed publisher
  59. Liu H, Wang H, Peterson M, Zhang W, Hou G, Zhang Z. N-terminal alternative splicing of GluN1 regulates the maturation of excitatory synapses and seizure susceptibility. Proc Natl Acad Sci U S A. 2019;116:21207-21212 pubmed publisher
  60. Bagasrawala I, Memi F, V Radonjić N, Zecevic N. N-Methyl d-Aspartate Receptor Expression Patterns in the Human Fetal Cerebral Cortex. Cereb Cortex. 2017;27:5041-5053 pubmed publisher
  61. Guastella J, Brecha N, Weigmann C, Lester H, Davidson N. Cloning, expression, and localization of a rat brain high-affinity glycine transporter. Proc Natl Acad Sci U S A. 1992;89:7189-93 pubmed
  62. Wu Z, Parry M, Hou X, Liu M, Wang H, Cain R, et al. Gene therapy conversion of striatal astrocytes into GABAergic neurons in mouse models of Huntington's disease. Nat Commun. 2020;11:1105 pubmed publisher
  63. Trifonov S, Yamashita Y, Kase M, Maruyama M, Sugimoto T. Glutamic acid decarboxylase 1 alternative splicing isoforms: characterization, expression and quantification in the mouse brain. BMC Neurosci. 2014;15:114 pubmed publisher
  64. Lewis D, Melchitzky D, Haycock J. Four isoforms of tyrosine hydroxylase are expressed in human brain. Neuroscience. 1993;54:477-92 pubmed
  65. Kubis N, Faucheux B, Ransmayr G, Damier P, Duyckaerts C, Henin D, et al. Preservation of midbrain catecholaminergic neurons in very old human subjects. Brain. 2000;123 ( Pt 2):366-73 pubmed
  66. Taravini I, Chertoff M, Cafferata E, Courty J, Murer M, Pitossi F, et al. Pleiotrophin over-expression provides trophic support to dopaminergic neurons in parkinsonian rats. Mol Neurodegener. 2011;6:40 pubmed publisher
  67. Zhang X, Lei B, Yuan Y, Zhang L, Hu L, Jin S, et al. Brain control of humoral immune responses amenable to behavioural modulation. Nature. 2020;581:204-208 pubmed publisher
  68. Carta I, Chen C, Schott A, Dorizan S, Khodakhah K. Cerebellar modulation of the reward circuitry and social behavior. Science. 2019;363: pubmed publisher
  69. Nirenberg M, Chan J, Vaughan R, Uhl G, Kuhar M, Pickel V. Immunogold localization of the dopamine transporter: an ultrastructural study of the rat ventral tegmental area. J Neurosci. 1997;17:4037-44 pubmed
  70. Revay R, Vaughan R, Grant S, Kuhar M. Dopamine transporter immunohistochemistry in median eminence, amygdala, and other areas of the rat brain. Synapse. 1996;22:93-9 pubmed
  71. Xia N, Zhang P, Fang F, Wang Z, Rothstein M, Angulo B, et al. Transcriptional comparison of human induced and primary midbrain dopaminergic neurons. Sci Rep. 2016;6:20270 pubmed publisher
  72. Reyes S, Fu Y, Double K, Thompson L, Kirik D, Paxinos G, et al. GIRK2 expression in dopamine neurons of the substantia nigra and ventral tegmental area. J Comp Neurol. 2012;520:2591-607 pubmed publisher
  73. Ikonomovic M, Abrahamson E, Isanski B, Wuu J, Mufson E, DeKosky S. Superior frontal cortex cholinergic axon density in mild cognitive impairment and early Alzheimer disease. Arch Neurol. 2007;64:1312-7 pubmed
  74. Benzing W, Mufson E, Armstrong D. Immunocytochemical distribution of peptidergic and cholinergic fibers in the human amygdala: their depletion in Alzheimer's disease and morphologic alteration in non-demented elderly with numerous senile plaques. Brain Res. 1993;625:125-38 pubmed
  75. Yamada M, Chiba T, Sasabe J, Terashita K, Aiso S, Matsuoka M. Nasal Colivelin treatment ameliorates memory impairment related to Alzheimer's disease. Neuropsychopharmacology. 2008;33:2020-32 pubmed
  76. Engelhardt J, Le W, Siklos L, Obal I, Boda K, Appel S. Stereotaxic injection of IgG from patients with Alzheimer disease initiates injury of cholinergic neurons of the basal forebrain. Arch Neurol. 2000;57:681-6 pubmed
  77. Lallani S, Villalba R, Chen Y, Smith Y, Chan A. Striatal Interneurons in Transgenic Nonhuman Primate Model of Huntington's Disease. Sci Rep. 2019;9:3528 pubmed publisher
  78. Perez S, Dar S, Ikonomovic M, DeKosky S, Mufson E. Cholinergic forebrain degeneration in the APPswe/PS1DeltaE9 transgenic mouse. Neurobiol Dis. 2007;28:3-15 pubmed
  79. Fujishiro H, Umegaki H, Isojima D, Akatsu H, Iguchi A, Kosaka K. Depletion of cholinergic neurons in the nucleus of the medial septum and the vertical limb of the diagonal band in dementia with Lewy bodies. Acta Neuropathol. 2006;111:109-14 pubmed
  80. Abdo H, Calvo Enrique L, Lopez J, Song J, Zhang M, Usoskin D, et al. Specialized cutaneous Schwann cells initiate pain sensation. Science. 2019;365:695-699 pubmed publisher
  81. Trevino A, Sinnott Armstrong N, Andersen J, Yoon S, Huber N, Pritchard J, et al. Chromatin accessibility dynamics in a model of human forebrain development. Science. 2020;367: pubmed publisher
  82. Robbins J, Perfect L, Ribe E, Maresca M, Dangla Valls A, Foster E, et al. Clusterin Is Required for β-Amyloid Toxicity in Human iPSC-Derived Neurons. Front Neurosci. 2018;12:504 pubmed publisher
  83. Gascon E, Vutskits L, Kiss J. Polysialic acid-neural cell adhesion molecule in brain plasticity: from synapses to integration of new neurons. Brain Res Rev. 2007;56:101-18 pubmed
  84. Chae J, Stein G, Lee J. NeuroD: the predicted and the surprising. Mol Cells. 2004;18:271-88 pubmed
  85. Shin R, Iwaki T, Kitamoto T, Tateishi J. Hydrated autoclave pretreatment enhances tau immunoreactivity in formalin-fixed normal and Alzheimer's disease brain tissues. Lab Invest. 1991;64:693-702 pubmed
  86. Pooler A, Hanger D. Functional implications of the association of tau with the plasma membrane. Biochem Soc Trans. 2010;38:1012-5 pubmed publisher
  87. Bastianelli E. Distribution of calcium-binding proteins in the cerebellum. Cerebellum. 2003;2:242-62 pubmed
  88. Lammert C, Frost E, Bellinger C, Bolte A, McKee C, Hurt M, et al. AIM2 inflammasome surveillance of DNA damage shapes neurodevelopment. Nature. 2020;580:647-652 pubmed publisher
  89. Arenzana F, Santos Ledo A, Porteros A, Aijon J, Velasco A, Lara J, et al. Characterisation of neuronal and glial populations of the visual system during zebrafish lifespan. Int J Dev Neurosci. 2011;29:441-9 pubmed publisher
  90. Portier M, Escurat M, Landon F, Djabali K, Bousquet O. Peripherin and neurofilaments: expression and role during neural development. C R Acad Sci III. 1993;316:1124-40 pubmed
  91. Knaus P, Marquèze Pouey B, Scherer H, Betz H. Synaptoporin, a novel putative channel protein of synaptic vesicles. Neuron. 1990;5:453-62 pubmed
  92. Lee K, Queenan B, Rozeboom A, Bellmore R, Lim S, Vicini S, et al. Mossy fiber-CA3 synapses mediate homeostatic plasticity in mature hippocampal neurons. Neuron. 2013;77:99-114 pubmed publisher
  93. Ou Yang M, Kurz J, Nomura T, Popovic J, Rajapaksha T, Dong H, et al. Axonal organization defects in the hippocampus of adult conditional BACE1 knockout mice. Sci Transl Med. 2018;10: pubmed publisher
  94. Singec I, Knoth R, Ditter M, Hagemeyer C, Rosenbrock H, Frotscher M, et al. Synaptic vesicle protein synaptoporin is differently expressed by subpopulations of mouse hippocampal neurons. J Comp Neurol. 2002;452:139-53 pubmed
  95. Llinas RR, Walton KD, Lang EJ. Cerebellum. In: Shepherd GM, editors. The Synaptic Organization of the Brain. New York: Oxford University Press; 2004.
  96. Masland R. Neuronal cell types. Curr Biol. 2004;14:R497-500 pubmed
  97. The Human Connectome Project. Available from: www.humanconnectomeproject.org/
  98. Mountcastle V, Talbot W, Sakata H, Hyvarinen J. Cortical neuronal mechanisms in flutter-vibration studied in unanesthetized monkeys. Neuronal periodicity and frequency discrimination. J Neurophysiol. 1969;32:452-84 pubmed
  99. Connors B, Gutnick M. Intrinsic firing patterns of diverse neocortical neurons. Trends Neurosci. 1990;13:99-104 pubmed
  100. Luo L. Fly MARCM and mouse MADM: genetic methods of labeling and manipulating single neurons. Brain Res Rev. 2007;55:220-7 pubmed
  101. Shimono M, Tsuji N. Study of the selectivity of the impregnation of neurons by the Golgi method. J Comp Neurol. 1987;259:122-30 pubmed
  102. Pasternak J, Woolsey T. On the "selectivity" of the Golgi-Cox method. J Comp Neurol. 1975;160:307-12 pubmed
  103. Joh T, Shikimi T, Pickel V, Reis D. Brain tryptophan hydroxylase: purification of, production of antibodies to, and cellular and ultrastructural localization in serotonergic neurons of rat midbrain. Proc Natl Acad Sci U S A. 1975;72:3575-9 pubmed
  104. Pickel V. Immunocytochemical localization of neuronal antigens: tyrosine hydroxylase, substance P, [Met5]-enkephalin. Fed Proc. 1979;38:2374-80 pubmed
  105. Schmechel D, Marangos P, Zis A, Brightman M, Goodwin F. Brain endolases as specific markers of neuronal and glial cells. Science. 1978;199:313-5 pubmed
  106. Salton S, Richter Landsberg C, Greene L, Shelanski M. Nerve growth factor-inducible large external (NILE) glycoprotein: studies of a central and peripheral neuronal marker. J Neurosci. 1983;3:441-54 pubmed
  107. Houser C, Crawford G, Salvaterra P, Vaughn J. Immunocytochemical localization of choline acetyltransferase in rat cerebral cortex: a study of cholinergic neurons and synapses. J Comp Neurol. 1985;234:17-34 pubmed
  108. Borit A, Brooks T, Ordonez N, Kakulas B. Central neural antigens: detection and diagnostic application. Crit Rev Clin Lab Sci. 1986;23:219-43 pubmed
  109. Hayashi K, Motoi M, Nose S, Horie Y, Akagi T, Ogawa K, et al. An immunohistochemical study on the distribution of glial fibrillary acidic protein, S-100 protein, neuron-specific enolase, and neurofilament in medulloblastomas. Acta Pathol Jpn. 1987;37:85-96 pubmed
  110. Jefferis G, Livet J. Sparse and combinatorial neuron labelling. Curr Opin Neurobiol. 2012;22:101-10 pubmed publisher
ISSN : 2329-5139