Open access peer-reviewed chapter

Gap Junctions in the Dorsal Root Ganglia

Written By

Vishwajit Ravindra Deshmukh

Submitted: 25 September 2018 Reviewed: 19 October 2018 Published: 21 December 2018

DOI: 10.5772/intechopen.82128

From the Edited Volume

Neurons - Dendrites and Axons

Edited by Gonzalo Emiliano Aranda Abreu and María Elena Hernández Aguilar

Chapter metrics overview

1,402 Chapter Downloads

View Full Metrics

Abstract

Dorsal root ganglion (DRG) or spinal ganglia are present in relation to the dorsal ramus of the spinal nerves. The neurons in the dorsal root ganglion are pseudounipolar in type. The single process from the soma or body will divide into the central and peripheral processes. Dorsal root ganglion neurons constitute the first-order neurons for the pain pathways and can be categorized as small, medium and large varieties. Peripheral process collects the impulses from the peripheral receptors and the central process reaches out to the central nervous system. The neurons in the DRG were surrounded by the satellite glial cells (SGC). These cells ensheath the neurons from all the sides. Besides covering the neurons, they share features very much similar to the astrocytes such as expression of glutamine synthetase. Many quantitative studies have identified the different proportion of satellite glial cells for individual neurons. These cells have been identified to get activated when confronted by the noxious stimuli, injury or inflammation. Clinically, these cells were implied to be related to the many neurological disorders.

Keywords

  • neurons
  • satellite glial cells
  • communicating junctions
  • pain
  • connexin-43
  • glial fibrillary acidic protein
  • peripherin
  • Nissl stain
  • immunohistochemistry

1. Introduction

The human nervous system is an extremely efficient, compact, fast and reliable computing system, yet it weighs substantially less than most of the computers and performs at an incredibly greater capacity.

The nervous system is subdivided, morphologically into two components, the central nervous system (CNS) consisting of the brain and spinal cord and the peripheral nervous system (PNS) comprising of cranial and spinal nerves and ganglia.

Discrete collections of nerve cell bodies in the CNS are known as nuclei while in PNS, these are called ganglia. The nerve cell bodies are of varying sizes and shapes. Ganglia are present in the dorsal root of spinal nerves, the sensory root of the trigeminal nerve (Vth), Facial (VIIth), Glossopharyngeal (IXth), Vagus (Xth) nerves and in the autonomic nervous system [1]. Some of them have independent nomenclature like the “Gasserian ganglion” for the Vth nerve. Thus ganglia can be divided into two types somatic and autonomic (Figure 1). The nerve cell bodies in each of these differ in their size and shape. Somatic ganglia contain small to large pseudounipolar neurons while the autonomic ganglia contain small multipolar neurons.

Figure 1.

Differences in sensory and autonomic ganglia (courtesy: Cranial Nerves and Functional Anatomy, 1st ed. p. 12).

Depending on the number of processes, a neuron can be classified into various categories. Unipolar neurons (no dendrites only an axon) are rare in vertebrates, bipolar neurons (possesses an axon and a dendrite) present in olfactory mucosa and the retina and multipolar neurons (single axon and two or more dendrites) present in the central nervous system except the mesencephalic nucleus of the Vth cranial nerve. An additional type of neuron, the pseudounipolar neuron is present in sensory ganglia and the ganglia of Vth, VIIth, IXth and Xth cranial nerves. It divides into a central and peripheral process (Figure 2).

Figure 2.

Types of neurons in nervous system.

The neurons in sensory ganglia are at first bipolar, but the two neurites soon unite to form a single process during development. Structurally and electrophysiologically, both these processes show characteristic features of the axon [2]. Small satellite glial cells tightly wrap the cell bodies of the pseudounipolar neurons in the ganglion. The satellite cells that surround the pseudounipolar neuron are continuous with the Schwann cell sheath that surrounds the axon [3]. A distinctive feature of satellite glial cells by which they are distinguished from astrocytes is that they completely surround the individual sensory neuron. The neuron and its surrounding satellite glial cells form a distinct morphological and probably a functional unit [4]. The somatic ganglia of all the mammalian and avian species demonstrate this arrangement [5]. Satellite glial cells have been implicated in neuronal nutrition, homeostasis, and the process of apoptosis. It is known that astrocytes in the central nervous system perform ‘spatial buffering’ (regulation of K+) and it is presumed that SGCs also perform the same function [5]. Removing K+ from the perineuronal environment would reduce neuronal excitation and therefore contribute to the lowering of pain.

Advertisement

2. Morphology of Dorsal root ganglia (DRG)

Dorsal root ganglia (sensory ganglia) contain the cell bodies of primary afferent neurons that transmit the sensory information from the periphery into the central nervous system (CNS) [6]. Sensory ganglia were located near the entrance of dorsal root into the spinal cord, and are not a part of CNS. Sensory (somatic) ganglia lie outside the blood-brain barrier and are densely vascularized by fenestrated capillaries, making the neurons and SGCs easily accessible to compounds in the circulation, including chemotherapeutic drugs [7]. Chemotherapeutic drugs show greater accumulation in sensory ganglia than in peripheral nerves [8]. Dorsal root ganglia are more sensitive to heat than other nervous tissues [9]. It is known that pulsed radiofrequency can selectively block sensory nerves while minimizing the destruction of motor nerves. Sluijter et al. reported that the placement of a cannula 1–2 cm peripheral to the dorsal root ganglia could result in maximum effect when pulsed radiofrequency was applied on dorsal root ganglia of the spinal cord [10]. Kikuchi et al. [9] classified anatomical positions and variations of dorsal root ganglia into intraspinal (IS), intraforaminal (IF), and extraforaminal (EF) (Figure 3).

Figure 3.

Positions of dorsal root ganglia (DRG) were determined by two schematic lines and classified into three types. Line A: aligning the medial borders of L4 and L5 pedicles, Line B: aligning the centers of L4 and L5 pedicles, Intraspinal type (IS): DRG located proximal to line A, Intraforaminal type: DRG located between line A and B, Extraforaminal type: DRG located distal to line B [9].

Advertisement

3. Morphology and histology of sensory (somatic) ganglia

The segmental nature of the spinal cord is demonstrated by the presence of 31 pairs of spinal nerves, but there is little indication of segmentation in its internal structure. Each dorsal root is broken up into a series of rootlets that are attached to the spinal cord along the corresponding segment. The ventral root arises similarly as a series of rootlets. These rootlets join to form the ventral and dorsal roots. The dorsal and ventral roots traverse the subarachnoid space and pierce the arachnoid and dura mater. At this point, the dura mater becomes continuous with the epineurium. After passing through the epidural space, the roots reach the intervertebral foramina, where the dorsal root ganglia are located on the dorsal root.

Certain authors have put forward their views regarding the classification of the neurons in the dorsal root ganglia based upon their staining properties into two histological types called “large light” and “small dark”, visible under the light microscope. This has been confirmed by recent electron microscopic analysis that indicates [11] the existence of two basic types of DRG neurons usually termed as type A and type B rather than large light and small dark [12]. The neurons in the dorsal root ganglion can also be divided into three types (small, medium and large neurons) based upon the size of their cell bodies. This classification seems to be more appropriate because the size of the neuronal cell bodies determine their function. The large neurons are mainly concerned with the transmission of proprioception and discriminative touch while the medium-sized neurons transmit nerve impulses associated with sensations like light touch, pressure, pain and temperature. However, the small-sized neurons exclusively transmit action potentials related to pain and temperature.

Glial cells are involved in various pathological processes affecting the central nervous system [13]. There is strong evidence that CNS glial cells are involved (microglia and astrocytes) in the induction and maintenance of neuropathic pain [14]. Following injury of a peripheral nerve, satellite glial cells (SGCs) in the dorsal root ganglia undergo changes in cell number, structure and function, similar to those in the CNS [15]. Peripheral nerve transection increases gap junctions and intercellular coupling of SGCs. SGCs also upregulated the production of proinflammatory cytokines such as tumor necrosis factor-α after lumbar facet joint injury [16].

Thus it is well established that glial cells play a critical role in the genesis and persistence of pain [17]. This is particularly true for the sensory ganglia. Though there are far fewer satellite glial cells than astrocytes or Schwann cells, yet because of their unique location in sensory ganglia, SGCs can strongly influence the afferent sensation. They also respond to the nerve injury by upregulating glial fibrillary acidic protein (GFAP) [18]. One of the ways glial cells in the sensory ganglia transmit signals is through intercellular calcium waves (ICWs) via gap junctions and adenosine-5′-triphosphate (ATP) acting on purinergic type 2 (P2) receptors [19]. This signaling has been shown to be bi-directional between SGCs and neurons (Figure 4).

Figure 4.

Schematic diagram describing the structural and functional relations between SGCs and neurons in sensory ganglia, and the consequences of peripheral injury.

Advertisement

4. Classification of pseudounipolar neurons of dorsal root ganglia into small, medium and large

Older literature suggests that neurons in dorsal root ganglia can be divided into two histological types called “large light (LL)” and “small dark (SD)” on the basis of staining properties under the light microscope [20]. This population overlaps, but still, they show the several physiological, biochemical and functional differences. Small dark neurons transmit the sensation particularly carried by C fibers (non-myelinated, slow conducting) [21]. Whereas Large light transmits the sensation carried via a fiber (myelinated and fast conducting). Many of the small dark neurons contain substance P or calcitonin gene-related peptide, and they are concerned with thermo- and mechanoreception, and many of them are nociceptive. The terminals of Large light neurons are low threshold mechanoreceptors [22]. Neurons in the sensory ganglia have no dendrites and also do not receive synapses but are still endowed with receptors for numerous neurotransmitters. More recently depending upon the electron microscopic appearance neurons in the dorsal root ganglia were divided into Type A and Type B for large light and small dark neurons respectively. Various other electrophysiological classification depending upon conduction velocity, modality and adaptation rate serves to distinguished large number of functional types of sensory neurons, but it is not clear how these are related to the two basic histological types.

There are contradictions among the researchers regarding the classification of dorsal root ganglia neurons into small, medium and large categories.

One of the studies involving chronic constriction injury model of Bennet and Xie [23] that retains the connection with the original receptive field so that hyperalgesia and allodynia can be demonstrated, classify the neurons in DRG into small (23–30 μm), medium (31–40 μm) and large (41–53 μm), based on the optical measurement of the average diameter [23]. These grouping roughly correspond to those giving rise to C, Aδ and Aβ fibers, respectively [21].

More recently sensory neurons in dorsal root ganglia were classified depending upon the immunohistochemical staining such as Nav1.8 expression in sensory neurons isolated from dorsal root ganglia into small (27–31 μm), medium (31–40 μm) and large (40–50 μm) [24]. There are two factors, namely DNA content and transcriptional activity, that are determinants of cell size [25]. Differences in neuronal body size seem to be primarily determined by the transcriptional activity. A positive correlation between the cell body and total RNA synthesis has been demonstrated in frog neurons, indicating that large neurons need higher transcriptional activities to maintain their large size [26]. The neurons transcription rate is, in turn, positively related to the magnitude of interactions between neurons and their targets, which contributes to the regulation of the soma size and metabolic activity [27].

Sensory neurons of the dorsal root ganglia express multiple voltage-gated sodium channels that substantially differ in gating kinetics and pharmacology. Small diameter (less than 25 μm) neurons isolated from the rat DRG express a combination of fast tetrodotoxin-sensitive (TTX-S) and slow TTX-resistant (TTX-R) sodium channels while large diameter neurons (more than 30 μm) predominantly expresses TTX-S Na current [28].

Viral study including adeno-associated viral vectors (AAV) are increasingly used to deliver therapeutic genes to the central nervous system where they promote transgene expression in postmitotic neurons for long periods with little or no toxicity. In adult rat dorsal root ganglia authors investigated the cellular tropism of AAV8 containing green fluorescent protein gene (GFP) after intra-lumbar DRG injection. And after injection, 2% of small DRG neurons (less than 30 μm) were GFP (+) as compared to 32% large (more than 60 μm) DRG neurons [29].

Electron microscopic features of dorsal root a ganglion divides the neurons depending upon their size and the distribution of their organelles (Figure 5). They were further subdivided into six subtypes according to the arrangement and three-dimensional organization of the Nissl bodies and Golgi apparatus in the perikarya. Type A1 cells were large, clear neurons in which Nissl bodies, separated from each other by pale narrow strands of cytoplasm containing small stacks of Golgi saccules and rod-like mitochondria, were evenly distributed throughout the perikaryon. In type A2, the Nissl bodies assumed a similar distribution but were separated by much wider strands of cytoplasm. Type A3, the smallest of the type A category, displayed densely packed Nissl bodies and long stacks of Golgi saccules which formed a perinuclear ring in the midportion of the perikaryon. Type B cells were smaller and showed a concentric zonation of their organelles. In type B1, large Nissl bodies located in an outer cytoplasmic zone were made of long piles of parallel cisternae interrupted by curved Golgi stacks. Type B2 was characterized by a ring-like Golgi apparatus separating the perikaryon in a cortical zone composed mainly of Nissl substance and a juxtanuclear zone containing mitochondria and smooth endoplasmic reticulum. Type C cells were the smallest of the ganglion cells and contained small, poorly demarcated Nissl bodies and a juxtanuclear Golgi apparatus [30].

Figure 5.

Nissl’s staining showing the variety of neurons in the dorsal root ganglion. Black arrow represents the large neurons, red arrow represents the surrounding capsule and the asterisk showed the location of centrally placed collection of nerve fibers [33].

Neurotransmitter study involving tachykinin like substance P (SP) and neurokinin A, which are released by the C-type primary afferent terminals of the small DRG neurons, plays important role in spinal nociception. By means of non-radioactive in situ hybridization and whole-cell recording, authors showed that the small rat DRG neurons also express the NK-1 tachykinin receptor. In situ hybridization demonstrated that the positive neurons in rat DRG sections were mainly small with a diameter of less than 25 μm. And the remaining positive neurons were cells with a medium diameter between 26 and 40 μm. No positive large neurons (more than 40 μm) were observed [31].

Depending upon the molecular weight of neurofilaments and their expression in various categories of neurons in dorsal root ganglia, three different neurofilament subunits have been identified, i.e. light (NF-L), middle (NF-M) and high (NF-H). Previous data showed that all the dorsal root ganglia neurons express NF-M and NF-H while only NF-L defines a distinct group of neurons and significantly large-light neurons [32].

Advertisement

5. Peripherin: marker to differentiate the neurons in the DRG

Peripherin, a protein formerly called Y, was first identified by two-dimensional gel electrophoresis in the insoluble fraction of cellular extracts from mouse neuroblastoma cell lines [34]. Its presence has been previously established in the rodent peripheral nervous system mostly by biochemical studies; moreover, biochemical characterization following nerve transection also supports its localization in neurons within the peripheral nervous system [35]. This observation leads to coining of the term “Peripherin” to designate this particular protein entity. Peripherin is a 57-kDa-type III neuronal intermediate filament protein, which is capable of either self-assembling or co-assembling with all of the individual neurofilament subunits [36]. In particular, the small cells of the dorsal root ganglia neurons selectively contain peripherin [35] and thus becoming a useful marker to define the small ganglion cell subpopulation. The exact function of the peripherin is still unknown though it has been suggested to be a determinant of the shape and architecture of the peripheral nerve axons and also provides structural integrity to the cells [37]. Peripherin immunolabeling has seen to be an important marker especially for the study of peripheral nerve development and regeneration since this intermediate filament protein is highly over-expressed during axon elongation [38]. Previously this neurofilament were thought to be inert but in fact these are highly dynamic structures with many diverse function such as relaying the signals from the plasma membrane to the nucleus [39], maintaining the position and function of cellular organelles, and also regulating the protein synthesis [40]. This neurofilament is clinically relevant because of their association with the pathogenesis of some major neuronal disorders. Mainly, accumulation of neurofilament protein and peripherin in proximal axons are associated with amyotrophic lateral sclerosis [41] and also seen in other diseases such as Alzheimer’s disease [42]. Peripherin was used to identify the small to medium-sized neurons in the rat dorsal root ganglia in the present study as because these are associated with the transmission of pain from the periphery to the central nervous system. This would give an idea as to the actual number of neurons within the dorsal root ganglia involved in the transmission of pain (Figure 6).

Figure 6.

Immunohistochemical stained section with peripherin antibody of dorsal root ganglion representing the specific staining in small to medium sized neurons (white arrows). Larger neurons (black arrows) [33].

Advertisement

6. Satellite glial cells

Sensory neurons in the dorsal root ganglia are ensheathed by specialized glial cells termed ‘satellite glial cells’ (SGCs). Recently, there has been considerable interest in these cells as they are profoundly altered by peripheral injuries used to study pain behavior and appear to contribute to chronic pain [43]. Satellite glial cells are the peripheral glial cells, but share many properties with astrocytes in the central nervous system (CNS), including the expression of glutamine synthetase and transporters of amino acids neurotransmitters. However, satellite glial cells differ in some respects from astrocytes, particularly by the tight sheath they make around the neuronal cell bodies [44]. In the dorsal root ganglion, Schwann cells and the satellite cells are activated in response to ischemia, traumatic injury and inflammation [45]. Application of various cytokines to the exposed Dorsal root ganglia resulted in an increase in the discharge rate as well as increased mechanosensitivity of DRG and peripheral receptive fields [46]. Satellite glial cells are the consistent component of the DRG in all the species, yet their contribution to the basic neuronal function remains unknown, although these satellite cells were implicated in neuronal nutrition, homeostasis and the process of apoptosis [5].

Recent studies have demonstrated that a specific glial cell population, the satellite glial cells, has the ability to regulate ion concentration [47] and possess mechanisms for the release of cytokines [48], ATP [19] and other chemical messengers like calcium. Satellite glial cells influence neuronal excitability via the gap junctions [49]. The satellite glial cells undergo major changes as a result of injury to peripheral nerves and appear to contribute to chronic pain [4]. Quantitative studies on several species showed that a number of satellite glial cells per neuron increases in proportion to the neuron’s volume, consistent with the idea that these satellite glial cells support the neurons metabolically [50].

During pathological conditions, such as nerve injury or inflammation, SGCs demonstrate an altered phenotype similar to that seen in activated astrocytes, which includes increased expression of the glial fibrillary acidic protein (GFAP) and synthesis of cytokines [51]. SGCs are therefore said to undergo activation due to injury. Increased coupling by gap junctions between SGCs has been observed in several inflammatory pain and axotomy models [52].

Advertisement

7. Satellite glial cells as a structural unit

Satellite glial cells (SGCs) in sensory ganglia wraps completely around the neuron. Several investigators claimed that SGCs bear processes and are therefore structurally similar to astrocytes but recent researches are that SGCs are laminar and have no true processes. In general, each sensory neuron has its own SGCs sheath, which usually consists of several SGCs, and thus the neuron and its surrounding satellite glial cells form a distinct morphological and probably functional unit. The region containing connective tissue separates these units. In some cases (5.6% in rat DRG) neurons from a small group containing two to three cells that are enclosed in common connective tissue space [44]. The neurons in the clusters are in most cases separated from each other by SGC sheath. The SGCs envelope usually consists of flat processes that lie close to the neuronal plasma membrane. The distance between the glial cell and neuronal plasma membrane is about 20 nm [44]. The neurons send numerous fine processes (microvilli), some of which fit into the invaginations of SGCs thus increasing the neuronal surface area and may allow an extensive exchange of chemicals between two cell types. A study on cultured SGCs of embryonic and neonatal rats showed that SGCs could transform into astrocytes, Schwann cells and oligodendrocytes [53].

Quantitative studies on several species showed that the number of SGCs per neuron increases in proportion to the neuron volume [50] consistent with the idea that SGCs support the neurons metabolically. It was also found that the mean volume of the nerve cell body corresponding to an SGC was lower for small neurons than for large neurons, which implies that the metabolic needs of small neurons are better satisfied than those of large ones. Therefore, smaller neurons have a higher resistance to insults, which seems to be the case for mercury poisoning. However, there is experimental evidence that smaller neurons are more likely to die following axonal damage [54]. As sensory ganglia are not protected from substances circulating in the blood, SGCs may be important in the context of exposure to toxic substances. In several studies, SGCs were examined after poisoning with heavy metals and it was found that these cells take up organic mercury compounds [55], and lead [56]. Mercury poisoning also caused SGCs proliferation [57]. Nineteen days after the administration of organic mercury to rats, SGCs in DRG were heavily labeled for mercury, and their ability to take up GABA was greatly diminished. Interestingly, small neurons were considerably less labeled for mercury than large neurons, which could be attributed to a more effective protection by SGCs. Prolonged (3–18 months) administration of lead acetate to rats resulted in prominent changes in SGCs in DRG, which included proliferation and hypertrophy of these cells. Although a certain degree of neuronal damage was observed, it can be proposed that the changes in SGCs provide a better protection to the neurons during lead poisoning.

Advertisement

8. Satellite glial cells maintain ionic concentration

The satellite glial cells neighboring the pseudounipolar neurons have a highly negative resting membrane potential and noticeable potassium permeability. The primary means of limiting extracellular levels of potassium in the sensory ganglia occurs through the process commonly called spatial buffering or syphoning which is mediated by satellite glial cells. The maintenance of a low extracellular potassium concentration is crucial for controlling the neuronal resting membrane potential and neuronal excitability. In sensory ganglia increased neuronal excitability has been associated with the occurrence of altered sensation, including the development of the neuropathic pain [58]. In the CNS buffering of extracellular potassium ions is carried by astrocytes, which consist of uptake by inwardly rectifying potassium (Kir) channels and dissipation through other channels and gap junctions [59]. It is established that the Kir current and Kir4.1 expression occur in the satellite glial cells [60]. Voltage-gated potassium channels are one of the important physiological regulators of the membrane potentials in excitable cells including sensory ganglion neurons.

Advertisement

9. Neuron-glial interactions

Central nervous system glial cells are increasingly known to be important regulator of synaptic activity and the key functional unit of nervous system [61]. Even though many of the same voltage-sensitive ion channels and neurotransmitter receptors of neurons are found in glia; glial cells lack the membrane properties obligatory to fire action potentials. Nevertheless, these ion channels and electrogenic membrane transporters permit glia to sense indirectly the level of neuronal activity by monitoring activity-dependent changes in the chemical surroundings shared by these two cell types. Complex imaging methods, which allow observation of changes in intracellular and extracellular signaling molecules in real time, show that glia, communicate with one another and with neurons primarily through chemical signals rather than electrical signals. Many of these signaling systems overlap with the neurotransmitter signaling systems of neurons, but some are specialized for glial-glial and neuron-glial communication. Neuron-glia cell interaction through gap junctions and extracellular paracrine/autocrine processes are believed to be important in the development of peripheral sensitization within the trigeminal ganglia [62]. Peripheral sensitization, which is characterized by increased neuronal excitability and a lowered threshold for activation, may possibly trigger a migraine attack. Moreover, activation and sensitization of the trigeminovascular afferent fibers appear crucial for initiation of migraine pain and for subsequent central centralization, in which increased excitability of second-order neurons leads to pain and allodynia. Increased gap junction communication between neurons and satellite glial cells was observed in the trigeminal ganglion in response to chemical activation of sensory trigeminal nerves [62].

Increased neuronal-glial signaling by way of gap junctions is common in neuroinflammatory CNS disorders, such as cerebral ischemia and Alzheimer’s disease and may have underlying pathological significance [63]. Tonabersat (SB-220453), a compound that binds selectively and with high affinity to a unique stereoselective site i.e. the gap junctions and inhibits it in rats and human brains [64]. After an injury, the numbers of gap junctions that connect satellite glial cells increases [43] in a probable adjust to the greater release of potassium ions with intense neuronal activity. Injury to a peripheral nerve does not directly impact satellite glial cells integrity. However, changes in injured neurons can influence the ability of the surrounding SGCs to regulate K+ via neuromodulators such as adenosine triphosphate (ATP) and nitric oxide (NO) [65].

Satellite glial cells have unique proteins that include the inwardly rectifying K+ channel Kir4.1 [43], the connexin-43 (Cx43) subunit of gap junctions the purinergic receptor P2Y4 [66] and soluble guanylate cyclase. There is also evidence of the presence of small-conductance Ca2+−activated K+ channel SK3 that is present only in satellite glial cells. All the above proteins are involved, either directly or indirectly, in potassium ion (K+) buffering and, thus, can influence the level of neuronal excitability, which, in turn, has been associated with neuropathic pain conditions (Figure 7). They also used in vivo RNA interference to reduce the expression of Cx43 (present only in SGCs) in the rat trigeminal ganglion and showed that this resulted in the development of spontaneous pain behavior. The pain behavior is present only when Cx43 is reduced and returns to normal when Cx43 concentrations are restored [66, 67].

Figure 7.

Satellite glial cells involved in maintenance of potassium homeostasis [66].

Advertisement

10. Glial fibrillary acidic protein (GFAP): locator molecule for the satellite glial cells

Glial fibrillary acidic protein is principle intermediate filament in mature astrocytes of the central nervous system and satellite glial cells of sensory ganglia [4]. GFAP is strongly unregulated in response to CNS damage [68]. It is thought to be important in astrocyte neuronal interactions, astrocyte mobility and shape and for maintenance of homeostasis and vascular permeability at the blood-tissue interface [69]. GFAP is essential for normal white matter architecture and blood-brain barrier integrity and its absence leads to late-onset CNS dysmyelination [70]. Increased GFAP expression occurs in activated glial cells. Activated astrocytes are characterized by hypertrophy, the release of pro-inflammatory cytokines (IL-1, IL-6 and TNF-a), the release of nitric oxide and prostaglandins, and up-regulation of the intermediate filaments GFAP and vimentin [17]. Likewise, satellite glial cells (SGCs) display increased expression of GFAP after neuronal injury or inflammation and undergo a number of changes similar to those seen in astrocytes, such as synthesis of cytokines [71]. GFAP expression increases in the satellite glial cells of trigeminal ganglia after tooth pulp injury [72]. The present study also investigated the expression of GFAP in the satellite glial cells following acute pain (Figure 8).

Figure 8.

Immunohistochemical staining for the section of DRG using GFAP antibody. Black arrows representing the location of satellite glial cells. Red arrow showing the communication between two neurons [33].

GFAP is a marker of activated satellite glial cells and astrocytes [48]. These ropes like filaments are called intermediate filaments because their diameter of 8–10 nm is between those of actin filaments and microtubules. Nearly all-intermediate filaments consist of subunits with a molecular weight of about 50 kDa. Some evidence suggests that many of the stable structural proteins in intermediate filaments evolved from highly conserved enzymes, with only minor genetic modification. Intermediate filaments are formed from nonpolar and highly variable intermediate filament subunits. Unlike those of microfilaments and microtubules, the protein subunits of intermediate filaments show considerable diversity and tissue specificity. In addition, they do not possess enzymatic activity and form nonpolar filaments. Intermediate filaments also do not typically disappear and reform in the continuous manner characteristic of most microtubules and actin filaments. For these reasons, intermediate filaments are believed to play a primarily structural role within the cell and to compose the cytoplasmic link of a tissue-wide continuum of cytoplasmic, nuclear, and extracellular filaments. A highly variable central rod-shaped domain with strictly conserved globular domains at either end characterizes intermediate filament proteins. Although the various classes of intermediate filaments differ in the amino acid sequence of the rod-shaped domain and show some variation in molecular weight, they all share a homologous region that is important in filament self-assembly. Intermediate filaments are assembled from a pair of helical monomers that twist around each other to form coiled-coil dimers. Then, two coiled-coil dimers twist around each other in antiparallel fashion (parallel but pointing in opposite directions) to generate a staggered tetramer of two coiled-coil dimers, thus forming the nonpolarized unit of the intermediate filaments. Each tetramer, acting as an individual unit, is aligned along the axis of the filament. The ends of the tetramers are bound together to form the free ends of the filament. This assembly process provides a stable, staggered, helical array in which filaments are packed together and additionally stabilized by lateral binding interactions between adjacent tetramers [2].

Total six classes of intermediate filament are present in body, e.g., Class II and I include keratin and cytokeratin and class III include vimentin, glial acidic fibrillary protein (GFAP) and peripherin.

GFAP is the principal intermediate filament in mature astrocytes. GFAP is a soluble protein isolated from the multiple sclerosis plaques and presumably arising from the glial filaments [73]. The GFAP gene is located on the long (q) arm of chromosome 17 at position 21. Mutation in the GFAP results in Alexander disease characterized by rare leukoencephalopathy affecting predominantly the brainstem and cervical cord with insidious onset of clinical features and unified by the presence in astrocytes of Rosenthal fibers (protein aggregates mainly contain glial fibrillary acidic protein (GFAP) and small stress proteins) in the astrocytes especially in the subpial and subependymal in location. It is strongly upregulated in response to the CNS damage [68]. It is thought to be important in astrocyte-neuronal communication and is believed to modulate astrocyte motility and shape. Satellite glial cells (SGCs) responsible for the maintenance of homeostasis and vascular permeability at the blood-tissue interface [69]. In the peripheral nervous system, neurons located in sensory ganglia are tightly surrounded by SGCs, following injury these cells undergo modification in structure and function [15]. According to Feng et al., after ligation of the L5 spinal nerve, mechanical allodynia developed in the ipsilateral hind paw and expression of GFAP in the ipsilateral DRG increased significantly as early as 4 hours after surgery, and gradually increases up to peak level at day 7 and then stayed at high level till day 56 [74]. Significant change seen among the sizes of neurons means small to medium size neurons shows maximum GFAP immunoreactivity at 12 hours and on day 7, a number of larger neurons was surrounded by GFAP stained satellite cells.

11. Gap junctions in the nervous system

Gap junctions, tight junctions, adherens junctions, desmosomes, hemidesmosomes, focal adhesions, chemical synapses, and immunological synapses are complex multiunit plasma membrane structures that assemble in a localized spatial and temporal organization to maintain structural tissue organization and to provide the cell signaling functions. At least nine connexins (Cx26, Cx32, Cx33, Cx36, Cx37, Cx40, Cx43, Cx45, Cx46) are expressed to various degrees in the nervous system. Functional studies in diverse cell types and in various exogenous expression systems have revealed that gap junction channels formed by different connexins are regulated differently, both at the single channel level (gating controls such as voltage sensitivity and variations in unitary conductance) and at the level of synthesis (expression, altered for example by hormones, extracellular matrix). Some gap junction channels are more sensitive to various gating stimuli than others, some display some degree of ionic selectivity, and some will pair promiscuously with other connexins (heterologous channels) while others are quite selective in their interaction (homologous channels). Such differences are important from the standpoint of the physiological roles of gap junctions in different cell types, as well as in the establishment of communication compartments within the nervous system [75]. Connexins are differentially expressed in the brain during ontogeny. Most recently, tissue culture preparations from embryonic neural tissue have allowed manipulation of individual cells and evaluation of changes in junctional distribution and expression during maturation. Such studies have clarified the relationships between sequential changes in phenotypes of neural cells, with the extent of coupling mediated by Cx43 (which is abundant in neural precursor populations) and the appearance of other gap junction proteins. Expression pattern of Cx32, Cx43 and Cx30 during the development in rat brain indicates the Connexin-43 appears first at embryonic days 12-18 [76] and that Cx32 protein and mRNA appear during first or second postnatal week and increases during development. Immunohistochemical analysis of postnatal rat brain has shown that Cx43 first appears along radial glial cells and is most intense along cerebellar Bergmann glial cells [77]. Glia represents the major cell population in the CNS coupled by gap junctions. Indeed, compared to neurons, the level of connexin expression is high in these cells and persists until the adult stage [75]. For the two main types of macroglial cells, the astrocytes and the oligodendrocytes, several connexins have been detected [78]. Gap junctional communication is not limited to either astrocyte-to-astrocyte or oligodendrocyte-to-oligodendrocyte, but it also occurs in between both cell types. In the adult brains, the predominant connexin is Cx43, which is abundant in astrocytes and is also expressed in leptomeninges, endothelial cells and ependyma. The second type of microglia, the oligodendrocytes (and their peripheral counterparts, the Schwann cells), appear to express a different gap junction protein, Cx32, although to a lower extent in situ than the level of Cx43 expression exhibited by astrocytes. Astrocytes express Cx43 and are well coupled in vivo and under culture conditions. However, the strength of coupling and degree of Cx43 expression between astrocytes varies depending on brain regions being higher in the hypothalamus than in the striatum. Although glial gap junctions do not generate action potentials in normal conditions and are devoid of synaptic contacts, connexin channels provide a route that allows changes in membrane potential to be transmitted from one cell to its neighbors. Recently, the participation of astrocytic gap junction in neuroprotection has been investigated by comparing neuronal vulnerability in the presence of either communicating or non-communicating astrocytes [75].

12. Gap junctions and connexins

Gap junctions and their consistent connexin proteins have represented a new challenge in all tissues where they occur but no structure is more complex or more interconnected than the mammalian central and peripheral nervous systems (CNS and PNS). The term “Gap junctions” arose from the work of Revel and Karnovsky, who described the fine structure of the interconnections between mouse cardiomyocytes and between hepatocytes. Later development of specific antibodies to gap junction proteins and eventually the cloning of these connexin molecules have now led to the availability of a variety of techniques by which the distribution and expression patterns of specific types of gap junctions have been defined in a varied number of tissues, including the brain. Gap junctions are the clusters of intercellular channels that are composed of 12 subunits, 6 of which form a connexion or hemichannel contributed by each of the coupled cells [79]. Gap junctions are permeant to molecules up to 1 kDa and are found in virtually all cell types in mammals; few exceptions include circulating erythrocytes, spermatozoids and adult innervated skeletal muscle cells [80]. Gap junctional communication is essential for many physiological events, including cell synchronization, differentiation, cell growth, and metabolic coordination of avascular organ including epidermis and lens [81]. Connexin family members share a similar structural topology. Each connexin has four transmembrane domains that constitute the wall/pore of the channels. These domains are linked by two extracellular loops that play roles in the cell-cell recognition and docking processes. There are three unchanged cysteine residues in each loop, which solely form intraconnexin disulfide bonds [82]. The transmembrane domains and extracellular loops are highly conserved among the family members. Furthermore, connexin proteins have cytoplasmic N- and C-terminal and a cytoplasm loop linking the second and third transmembrane domains. Although the N-terminus is conserved, the cytoplasmic loop and C-terminus show great variation in terms of sequence and length. The cytoplasmic tail and loop are susceptible to various post-translational modifications (e.g., phosphorylation), which are believed to have regulatory roles [83]. Connexons (hemichannels) are then carried to the cell surface via vesicles transported through microtubules, which fuse to the plasma membrane. These hemichannels can either form nonjunctional channels in unopposed areas of the cell membrane or diffuse freely to regions of cell-to-cell contact to find a partner connexon from a neighboring cell to complete the formation of intercellular channels. Intercellular channels then cluster into gap junction plaques, a highly dynamic event involving removal of old channels from the center of the plaque, while adding new gap junction subunits to the periphery [84]. The intercellular channels from the middle of the plaque are internalized into vesicular structures called “annular junctions” [85], which either fuse with the lysosome for degradation by lysosomal enzymes or are targeted to the proteasomal pathway [86]. The continuous synthesis and degradation of connexins through these mechanisms may provide for the quick adaptation of tissues to changing environmental conditions. Unopposed hemichannels can also be functional under certain conditions, including mechanical and ischemic stress. Under these circumstances, open hemichannels are thought to facilitate the release of a variety of factors such as ATP, glutamate, and NAD+ into the extracellular space, generating different physiological responses [87].

Up to date, there were 20 proposed members of the connexin family of proteins that form gap junctional intercellular communication channels in mammalian tissues, and over half are reported to be present in the nervous system. Identification of the several connexin proteins at gap junctions between each neuronal and glial cell type is necessary for the sensible design of investigations into the functions of gap junctions between glial cells and into the functional contributions of electrical and “mixed” (chemical plus electrical) synapses to communication between neurons in the mammalian nervous system (Figure 9).

Figure 9.

Immunohistochemical staining using connexin-43 antibody. Black arrows represent the location of gap junctions between the satellite glial cells and the neuronal bodies [33].

13. Pathophysiology of connexins

Gap junction’s role has been well evaluated concerning cell-to-cell interaction. There are two effects derived from gap junction’s function that may determine life and death of the connected cells [89]. The bystander effect promotes the death of normal cells adjacent to an apoptotic cell by diffusing toxic metabolites through gap junctions. In the same way there is the Good Samaritan effect that allows a condemned cell to live by draining the toxic metabolites to adjacent cells and maintaining cells integrity and thus tissue homeostasis. In this way gap junctions perform a dual function either saving or killing interconnected cells [88]. Some pathological conditions are directly related to gap junctions or to their altered function. Some human diseases are caused by mutated connexins [89]. Mutations on Cx32 induce a peripheral neuropathy named Charcot-Marie-Tooth disease. The many conductivity changes observed in this disease may be caused by altered protein traffic to the junctions, altered channel permeability and, sometimes, altered conformation of heterotypic channels [78]. Mutations of Cx36 may lead to the most common hereditary non-syndromic deafness. Cx43 structure may be altered in some forms of human epilepsy where Cx43 mRNA expression may or may not be altered. High Cx43 levels have been detected in β-4 positive amyloid plaques of Alzheimer’s disease [77], indicating either astrocytes invasion of the plaques or increased Cx43 expression by astrocytes, as observed in PC12 cells (cells from a rat pheochromocytoma) with increased expression of carboxy-terminal portions of amyloid precursor protein [90]. However a higher Cx43 expression in that area may reflect the existence of many activated macrophages/microglia. The decrease of Cx43 within an inflammatory focus suggests that factors as IL-1 β are involved in astrocytic connectivity decrease as observed in autoimmune experimental encephalitis.

References

  1. 1. Standring S. Gray’s Anatomy: The Anatomical Basis of Clinical Practice. 40th edition. London, UK: Churchill Livingstone; 2008. p. 55
  2. 2. Ross M, Pawlina W. Histology: A Text and Atlas of Histology. 6th edition. Baltimore, MD: Lippincott Williams and Wilkins; 2006. pp. 62 and 354
  3. 3. Kiernan JA. Barr’s The Human Nervous System. Philadelphia: Wolters Kluwer; 2009. p. 44
  4. 4. Hanani M. Satellite glial cells in sensory ganglia: From form to function. Brain Research. Brain Research Reviews. 2005;48:457-476
  5. 5. Pannese E. Observation on the morphology, submicroscopic structure and biological properties of satellite cells in sensory ganglia of mammals. Zeitschrift für Zellforschung und Mikroskopische Anatomie. 1960;52:567-597
  6. 6. Aldskogius H, Elfvin LG, Forsman CA. Primary sensory afferents in the inferior mesenteric ganglion and related nerves of the guinea pig. An experimental study with anterogradely transported wheat germ agglutinin-horseradish peroxidase conjugate. Journal of the Autonomic Nervous System. 1986;15:179-190
  7. 7. Jimenez-Andrade J, Herrera M, Ghilardi J, Vardanyan M, Melemedjian O, Mantyh P. Vascularization of the dorsal root ganglia and peripheral nerve of the mouse: Implications for chemical-induced peripheral sensory neuropathies. Molecular Pain. 2008;4:10
  8. 8. Cavaletti G, Cavalletti E, Oggioni N, Sottani C, Minoia C, D’Incalci M, et al. Distribution of paclitaxel within the nervous system of the rat after repeated intravenous administration. Neurotoxicology. 2000;21:389-393
  9. 9. Kikuchi S, Sato K, Konno S, Hasue M. Anatomic and radiographic study of dorsal root ganglia. Spine (Phila Pa 1976). 1994;19:6-11
  10. 10. Sluijter ME. Radiofrequency, Part I: The Lumbosacral Region. Meggen: Flivopress SA; 2001. pp. 119-138
  11. 11. Lawson SN, Caddy KWT, Biscoe TJ. Development of rat dorsal root ganglia neurons. Cell and Tissue Research. 1974;153:399-414
  12. 12. Duce IR, Keen P. An ultrastructural classification of the neuronal cells bodies of the rat dorsal root ganglia using the zinc-iodide-osmium impregnation. Cell and Tissue Research. 1997;185:263-277
  13. 13. Allen NJ, Barres BA. Glia-more than just brain glue. Nature. 2009;457:675-677
  14. 14. Cao H, Zhang YQ. Spinal glial activation contributes to pathological pain states. Neuroscience and Biobehavioral Reviews. 2008;32:972-983
  15. 15. Cherkas PS, Huang TY, Pannicke T, Tal M, Reichenbach A, Hanani M. The effects of axotomy on neurons and satellite glial cells in mouse trigeminal ganglion. Pain. 2004;110:290-298
  16. 16. Miyagi M, Ohtori S, Ishikawa T, Aoki Y, Ozawa T, Doya H, et al. Up-regulation of TNFalpha in DRG satellite cells following lumbar facet joint injury in rats. European Spine Journal. 2006;15:953-958
  17. 17. Watkins LR, Milligan ED, Maier SF. Glial activation: A driving force for pathological pain. Trends in Neurosciences. 2001;24:450-455
  18. 18. Gungigake KK, Goto T, Nakao K, Kobayashi S, Yamaguchi K. Activation of satellite glial cells in rat trigeminal ganglia after upper molar extraction. Acta Histochemica et Cytochemica. 2009;42:143-149
  19. 19. Suadicani SO, Cherkas PS, Zuckerman J, Smith DN, Spray DC, Hanani M. Bidirectional calcium signaling between satellite glial cells and neurons in cultured mouse trigeminal ganglia. Neuron Glia Biology. 2010;6:43-51
  20. 20. Hatai S. Number and size of spinal ganglion cells and dorsal root fibres in the white rat at different ages. The Journal of Comparative Neurology. 1902;12:107-124
  21. 21. Harper AA, Lawson SN. Electrical properties of rat dorsal root ganglia neurons with different peripheral nerve conduction velocities. Journal of Physiology (London). 1985b;359:47-63
  22. 22. Lawson SN. Morphological and biochemical cell types of sensory neurons. In: Scott AS, editor. Sensory Neurons, Diversity, Development and Plasticity. New York: Oxford University Press; 1999. pp. 27-59
  23. 23. Bennet GJ, Xie YK. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain. 1988;33:87-107
  24. 24. Ramchandra R, Mc Grew S, Baxter J, Elmslie KS. Nav1.8 channels are expressed in large, as well as small, diameter sensory afferent neurons. Channels. 2013;7(1):34-37
  25. 25. Cavalier-Smith T. Nuclear volume control by nucleoskeletal DNA, selection for cell volume and cell growth rate, and the solution of the DNA C-value paradox. Journal of Cell Science. 1978;34:247-278
  26. 26. Sato S, Burgess SB, McIlwain DL. Transcription and motoneuron size. Journal of Neurochemistry. 1994;63:1609-1615
  27. 27. Goldschmidt RB, Steward O. Retrograde regulation of neuronal size in the enthorhinal cortex: Consequences of the destruction of dentate gyrus granule cells with colchicine. Restorative Neurology and Neuroscience. 1992;3:335-343
  28. 28. Ho C, O’Leary ME. Single-cell analysis of sodium channels expression in dorsal root ganglia neurons. Molecular and Cellular Neurosciences. 2011;46(1):159-166
  29. 29. Jacques SJ, Ahmed Z, Forbes A, Douglas MR, Vigenswara V, Berry M, et al. AAV8(gfp) preferentially targets large diameter dorsal root ganglion neurons after both intra-dorsal root ganglion and intrathecal injection. Molecular and Cellular Neurosciences. 2012;49(4):464-474
  30. 30. Rambourg A, Clermont Y, Beaudet A. Ultrastructural features of six types of neurons in rat dorsal root ganglia. Journal of Neurocytology. 1983;12:47-66
  31. 31. Yum SW, Zhang J, Mo K, Li J, Scherer SS. A novel recessive Nefl mutation causes a severe, early- onset axonal neuropathy. Annals of Neurology. 2009;66:759-770
  32. 32. Goldstein ME, House HB, Gainer H. NF-L and peripherin immunoreactivities define distinct class of rat sensory ganglion cells. Journal of Neuroscience Research. 1991;30:92-104
  33. 33. Deshmukh V, Prasoon P, Ray SB. Role of peripherin in defining specific populations of cell bodies in the dorsal root ganglia. International Journal of Medical Science and Public Health. 1 Aug 2016;5(8):1656-1661
  34. 34. Portier MM, de Nechaud B, Gros F. Peripherin, a new member of the intermediate filament protein family. Developmental Neuroscience. 1983b;6:335-344
  35. 35. Portier MM, Croizet B, Gros F. A sequence of changes in cytoskeletol componants during neuroblastoma differentiation. FEBS Letters. 1982;146:283-288
  36. 36. Troy CM, Brown K, Greene LA, Shelanski ML. Ontogeny of the neuronal intermediate filament protein, peripherin, in the mouse embryo. Neuroscience. 1990;36:217-237
  37. 37. Helfand BT, Chang L, Goldman RD. Intermediate filaments are dynamic and motile elements of cellular architecture. Journal of Cell Science. 2004;117:133-141
  38. 38. Oblinger MM, Wong J, Parysek LM. Axotomy-induced changes in the expression of a type III neuronal intermediate filament gene. The Journal of Neuroscience. 1989;9:3766-3775
  39. 39. Paramio JM, Jorcano JL. Beyond structure: Do intermediate filaments modulate cell signalling? BioEssays. 2002;24:836-844
  40. 40. Kim S, Wong P, Coulombe PA. A keratin cytoskeletal protein regulates protein synthesis and epithelial cell growth. Nature. 2006;441:362-365
  41. 41. Xiao S, McLean J, Robertson J. Neuronal intermediate filaments and ALS: A new look at an old question. Biochimica et Biophysica Acta. 2006;1762:1001-1012
  42. 42. Sternberger NH, Sternberger LA, Ulrich J. Aberrant neurofilament phosphorylation in Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America. 1985;82:4274-4276
  43. 43. Haung TY, Belzer V, Hanani M. Gap junctions in dorsal root ganglia: Possible contribution to visceral pain. European Journal of Pain. 2010;(14):49e1-4e11
  44. 44. Pannese E, Ledda M, Arcidiacono G, Rigamonti I. Clusters of nerve cells bodies enclosed within a common connective tissue envelope in the spinal ganglia of the lizard and rat. Cell and Tissue Research. 1991;264:209-214
  45. 45. Hashizume H, DeLeo JA, Colburn RW, et al. Spinal glial activation and cytokine expression after lumbar root injury in the rat. Spine. 2000;25:1206-1217
  46. 46. Ozaktay AC, Kallakuri S, Takebayashi T. Effects of interleukin-1 beta, interleukin-6, and tumor necrosis factor on sensitivity of dorsal root ganglion and peripheral receptive fields in rats. European Spine Journal. 2006;15:1529-1537
  47. 47. Pannese E, Ledda M, Cherkas PS, Huang TY, Hanani M. Satellite cell reactions to axon injury of sensory ganglion neurons: Increase in number of gap junctions and formation of bridges connecting previously separate perineuronal sheaths. Anatomy and Embryology (Berl). 2003;206:337-347
  48. 48. Takeda M, Tanimoto T, Kadoi J, Nasu M, Takahashi M, Kitagawa J, et al. Enhanced excitability of nociceptive trigeminal ganglion neurons by satellite glial cytokine following peripheral inflammation. Pain. 2007;129:155-166
  49. 49. Ohara TP, Vit JP, Bhargava A, Jasmin L. Evidence for a role of connexin 43 in trigeminal pain using RNA interference in vivo. Journal of Neurophysiology. 2008;100:3064-3073
  50. 50. Ledda M, De Palo S, Pannese E. Ratios between number of neuroglial cells and number and volume of nerve cells in the spinal ganglia of two species of reptiles and three species of mammals. Tissue & Cell. 2004;36:55-62
  51. 51. Woodham P, Anderson PN, Nadim W, Turmaine M. Satellite cells surrounding axotomised rat dorsal root ganglion cells increase expression of a GFAP-like protein. Neuroscience Letters. 1989;98:8-12
  52. 52. Hanani M, Haung TY, Cherkas PS, Ledda M, Pannese E. Glial cell plasticity in sensory ganglia induced by nrve damage. Neuroscience. 2002;114:279-283
  53. 53. Svennigsen AF, Colman DR, Pedraza L. Satellite cells of dorsal root ganglia are multipotential glial precursors. Neuron Glia Biology. 2004;1:85-93
  54. 54. Lieberman AR. Some factors affecting retrograde neuronal responses to axonal lesions. In: Bellairs R, Gray EG, editors. Essays on the Nervous System. Oxford: Clarendon; 1974. pp. 71-105
  55. 55. Kumamoto T, Fukuhara N, Miyatake T, Araki K, Takahashi Y, Araki S. Experimental neuropathy induced by methyl mercury com-pounds: Autoradiographic study of GABA uptake by dorsal root ganglia. European Neurology. 1986;25:269-277
  56. 56. Schlaepfer WW. Experimental lead neuropathy: A disease of the supporting cells in the peripheral nervous system. Journal of Neuropathology and Experimental Neurology. 1969;28:401-418
  57. 57. Schionning JD, Danscher G. Autometallographic mercury correlates with degnera tive changes in dorsal root ganglia of rats intoxicated with organic mercury. APMIS. 1999;107:303-310
  58. 58. Amir R, Devor M. Electrical excitability of the soma of sensory neurons is required for spike invasion of the soma, but not for through-conduction. Biophysical Journal. 2003;84(4):2181-2191
  59. 59. Konishi T. Developmental and activity-dependent changes in K+ currents in satellite glial cells in mouse superior cervical ganglion. Brain Research. 1996;708:7-15
  60. 60. Hibino H, Horio Y, Fujita A, Inanobe A, Doi K, Gotow T, et al. Expression of an inwardly rectifying K+ channel, Kir4.1, in satellite cells of rat cochlear ganglia. The American Journal of Physiology. 1999;277:C638-C644
  61. 61. Fields RD, Stevens-Graham B. New insights into neuron-glia communication. Science. 2002;298(5593):556-562
  62. 62. Thalakoti S, Patil VV, Damodaram S, Vause CV, Langford LE, Freeman SE, et al. Neuron-glia signaling in trigeminal ganglion: Implications for migraine pathology. Headache. 2007;47:1008-1023
  63. 63. Kielian T, Esen N. Effects of neuroinflammation on glia-glia gap junctional intercellular communication: A perspective. Neurochemistry International. 2004;45:429-436
  64. 64. Upton N, Thompson M. Benzo[b]pyranols and related novel antiepileptic agents. Progress in Medicinal Chemistry. 2000;37:177-200
  65. 65. Benfenati V, Caprini M, Nobile M, Rapisarda C, Ferroni S. Guanosine promotes the up-regulation of inward rectifier potassium current mediated by Kir4.1 in cultured rat cortical astrocytes. Journal of Neurochemistry. 2006 Jul;98(2):430-445
  66. 66. Vit JP, Jasmin L, Bhargava A, Ohara PT. Satellite glial cells in the trigeminal ganglion as a determinant of orofacial neuropathic pain. Neuron Glia Biology. 2006;2(4):247-257
  67. 67. Weick M, Cherkas P, S, Härtig W, Pannicke T, Uckermann O, Bringmann A, et al. P2 receptors in satellite glial cells in trigeminal ganglia of mice. Neuroscience. 2003;120:969-977
  68. 68. Lee Y, Su M, Messing A, et al. Astrocyte heterogeneity revealed by expression of a GFAP-Lac Z transgene. Glia. 2006;53:677-687
  69. 69. Danielyan L, Tolstonog G, Traub P, et al. Colocalization of glial fibrillary acidicprotein, metallothionein, and MHC II in human, rat, NOD/SCID, and nude mouse skin keratinocytes and fibroblasts. The Journal of Investigative Dermatology. 2007;127:555-563
  70. 70. Liedtke W, Edelmann W, Bieri PL, et al. GFAP is necessary for the integrity of CNS white matter architecture and long term maintainance of myelination. Neuron. 1996;17:607-615
  71. 71. Jasmin L, Vit JP, Bhargava A, Ohara PT. Can satellite glial cells be therapeutic targets for pain control? Neuron Glia Biology. 2010;6:63-71
  72. 72. Stephenson JL, Byers MR. GFAP immunoreactivity in trigeminal ganglion satellite cells after tooth injury in rats. Experimental Neurology. 1995;131:11-22
  73. 73. Bignami A, Eng LF, Dahl D, Uyeda CT. Localization of the glial acidic fibrillary protein in astrocyte by immunofluoroscence. Brain Research. 1972;43:429-435
  74. 74. Liu F-Y, Sun Y-N, Wang F-t, Li Q, Li S, Zhao Z-F. Activation of satellite glial cells in lumbar dorsal root ganglia contributes to neuropathic pain after spinal nerve ligation. Brain Research. 2011;1427:65-77
  75. 75. Andrade-Rozental AF, Rozental R, Hopperstad MG, Wu JK, Vrionis FD, Spray DC. Gap junctions: The "kiss of death" and the "kiss of life". Brain Research. Brain Research Reviews. 2000;32:308-315
  76. 76. Belliveau DJ, Naus CCJ. Cellular localization of gap junctions mRNAs in developing rat brain. Developmental Neuroscience. 1995;17:81-96
  77. 77. Yamamoto T, Vukelic J, Hertzberg EL, Nagy JI. Differential anatomical and cellular patterns of connexin 43 expression during postnatal development of rat brain. Developmental Brain Research. 1992;66:165-180
  78. 78. Dermietzel R, Spray DC. From neuro-glue ('Nervenkitt') to glia: A prologue. Glia. 1998;24:1-7
  79. 79. Spray DC, Scemes E, Rozental R. Introduction to cell-cell communication. In: Zigmond B, Landis S, editors. Fundamental neuroscience. New York, NY: Academic Press; 1998. pp. 317-343
  80. 80. Sohl G, Maxeiner S, Willecke K. Expression and functions of neuronal gap junctions. Nature Reviews. Neuroscience. 2005;6:191-200
  81. 81. Vinken M, Vanhaecke T, Papeleu P, Snykers S, Henkens T, Rogiers V. Connexins and their channels in cell growth and cell death. Cellular Signalling. 2006;18:592-600
  82. 82. Krutovskikh V, Yamasaki H. Connexin gene mutations in human genetic diseases. Mutation Research. 2000;462:197-207
  83. 83. Cruciani V, Mikalsen SO. Connexins, gap junctional intercellular communication and kinases. Biology of the Cell. 2002;94:433-443
  84. 84. Gaietta G, Deerinck TJ, Adams SR, Bouwer J, Tour O, Laird DW, et al. Multicolor and electron microscopic imaging of connexin trafficking. Science. 2002;296:503-507
  85. 85. Jordan K, Chodock R, Hand AR, Laird DW. The origin of annular junctions: A mechanism of gap junction internalization. Journal of Cell Science. 2001;114:763-773
  86. 86. Qin H, Shao Q, Igdoura SA, Alaoui-Jamali MA, Laird DW. Lysosomal and proteasomal degradation play distinct roles in the life cycle of Cx43 in gap junctional intercellular communication-deficient and -competent breast tumor cells. The Journal of Biological Chemistry. 2003;278:30005-30014
  87. 87. Evans WH, De VE, Leybaert L. The gap junction cellular internet: Connexin hemichannels enter the signalling limelight. The Biochemical Journal. 2006;397:1-14
  88. 88. Farahani R, Pina-Benabou MH, Kyrozis A, Siddiq A, Barradas PC, Chiu FC, et al. Alterations in metabolism and gap junction expression may determine the role of astrocytes as "good samaritans" or executioners. Glia. 2005;50:351-361
  89. 89. Rosenthal R, Giaume C, Spray DC. Gap junctions in the nervous system. Brain Research Reviews. 2000;32:11-15
  90. 90. Lynn BD, Marotta CA, Nagy JI. Propagation of intercellular calcium waves in Pc12 cells overexpressing a carboxy-terminal fragment of amyloid precursor protein. Neuroscience Letters. 1995;199:21-24

Written By

Vishwajit Ravindra Deshmukh

Submitted: 25 September 2018 Reviewed: 19 October 2018 Published: 21 December 2018