Next Article in Journal
Adaptation and Changes in Actin Dynamics and Cell Motility as Early Responses of Cultured Mammalian Cells to Altered Gravitational Vector
Next Article in Special Issue
TNF-α Plus IL-1β Induces Opposite Regulation of Cx43 Hemichannels and Gap Junctions in Mesangial Cells through a RhoA/ROCK-Dependent Pathway
Previous Article in Journal
Time-Course Transcriptomic Profiling of Floral Induction in Cultivated Strawberry
Previous Article in Special Issue
GABABR Modulation of Electrical Synapses and Plasticity in the Thalamic Reticular Nucleus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Direct Cell-Cell Communication via Membrane Pores, Gap Junction Channels, and Tunneling Nanotubes: Medical Relevance of Mitochondrial Exchange

1
Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), 105 11th Street, Galveston, TX 77555, USA
2
Department of Surgery and TEAM Health Anesthesia, University of South Florida, 2 Tampa General Circle, Tampa, FL 33606, USA
3
Department of Pharmacology and Physiology, School of Medicine and Dentistry, University Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 6133; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23116133
Submission received: 30 April 2022 / Revised: 28 May 2022 / Accepted: 28 May 2022 / Published: 30 May 2022
(This article belongs to the Special Issue Chemical Regulation of Gap Junction Channels and Hemichannels 2.0)

Abstract

:
The history of direct cell-cell communication has evolved in several small steps. First discovered in the 1930s in invertebrate nervous systems, it was thought at first to be an exception to the “cell theory”, restricted to invertebrates. Surprisingly, however, in the 1950s, electrical cell-cell communication was also reported in vertebrates. Once more, it was thought to be an exception restricted to excitable cells. In contrast, in the mid-1960s, two startling publications proved that virtually all cells freely exchange small neutral and charged molecules. Soon after, cell-cell communication by gap junction channels was reported. While gap junctions are the major means of cell-cell communication, in the early 1980s, evidence surfaced that some cells might also communicate via membrane pores. Questions were raised about the possible artifactual nature of the pores. However, early in this century, we learned that communication via membrane pores exists and plays a major role in medicine, as the structures involved, “tunneling nanotubes”, can rescue diseased cells by directly transferring healthy mitochondria into compromised cells and tissues. On the other hand, pathogens/cancer could also use these communication systems to amplify pathogenesis. Here, we describe the evolution of the discovery of these new communication systems and the potential therapeutic impact on several uncurable diseases.

1. The Identification of Membrane Channels

The “cell theory,” proposed by Matthias J. Schleiden and Theodor Schwann in the early 19th century, stated that plant and animal tissue are both made of independent units (later called “cells”) [1,2]; rev. in [3]. This theory implied the existence of a wall-like structure (now known as the plasma membrane) functioning as a barrier to prevent molecules from spreading into neighboring cells and the extracellular compartment or vice versa. However, although this theory did not consider that molecules might move across the plasma membrane, we learned that certain molecules freely cross the plasma membrane soon after publication. Indeed, evidence of the existence of membrane channels was finally reported in the mid-20th century primarily by the studies of Alan L. Hodgkin and coworkers [4,5,6,7,8]; rev. in [9,10].

2. Direct Cell-Cell Communication via Gap Junctions

Unequivocal evidence of membrane channels directly mediates cell-to-cell communication in virtually all cells, but circulating cells emerged in the 1960s [11,12,13]. However, knowledge of ionic cell-cell communication in some excitable cells was reported earlier in invertebrate nervous systems [14,15].
In the late 1800s, zoologists discovered that invertebrates such as annelids (earthworms), crustaceans (crayfish), and cephalopods (squid) have large tube-like structures that extend from rostral to caudal ends of the body. Most relevant was the early report of this structure in the squid [16], a structure that would become fundamentally important for studying the ionic basis of the action potential and the function of ion channels in general. Most scientists attributed different functions to these tube-like structures, but Leydig, in 1864, was the first to propose their nervous function [17]. However, his interpretation was ignored for almost a century until John Z. Young eventually proved it right in 1938 [18].
Early in the 20th century, scientists realized that some of these giant structures (axons) were not continuous. The first to report it was George E. Johnson, who discovered discontinuities in giant crustacean axons (Cambarus and Palaemonetes) [19]. Two years later, these discontinuities were named “septa” in earthworm axons by Howard B. Stough [14]—earthworms have three giant axons: a median and two lateral, all segmented.
In his 1930′s study [15], Stough found that when the median axon is cut, only the anterior part of the worm contracts; in contrast, when just the lateral axons are cut, posterior stimulation causes the posterior portion of the worm to contract up to the cut area. This induced him to conclude that: “… the median giant fiber conducts antero-posteriorly and the lateral giant fibers conduct postero-anteriorly”. This, however, was proven wrong by John C. Eccles and coworkers who recorded electrical impulses in isolated nerve cords elicited either in the head or the tail of the earthworm [20]; in their words: “… the transverse membranes do not influence the conduction of impulses, although the separation of the segments utilizing these transverse membranes appears to be as complete as that existing at vertebrate synaptic junctions”. Several research teams confirmed their findings a decade later [21,22,23,24]. Stough, nonetheless, deserves credit for demonstrating for the first time the direct cell-cell communication across septal barriers [14,15]. Three decades later, the detailed ultrastructure of earthworm junctions was described by Kiyoshi Hama as a close membrane apposition ~200 Å thick [25].
By noticing the apparent polarization of giant earthworm axons, Stough inadvertently discovered the capacity of injured cells to become independent from healthy cells [15]. This property, now known as cell-cell uncoupling, is mediated by the gating mechanism of gap junction channels. The unpolarized transmission of electrical impulses across septa was also reported in lateral giant axons of crayfish [26] and was later confirmed by intracellular recording [27,28,29,30]. Evidence of electrical cell-cell coupling was reported in other invertebrates, such as lobster cardiac ganglion [27,28,29,30] and muscle fibers [31], and leech segmental ganglia [32,33].

3. Direct Electrical Communication between Mammalian Cardiac Fibers

In the early 1950s, Silvio Weidmann made a major discovery while measuring by intracellular recording the myoplasm’s electrical resistance of kid heart’s Purkinje fibers [34]; in his words: “… The relatively low value of the specific d.c. resistance of myoplasm (twice that of Tyrode solution) suggests (i) that the smaller units making up the Purkinje fiber, the Purkinje cells, are not surrounded by ionic barriers of any importance, (ii) that transverse membranes do not subdivide Purkinje fibers, etc., and (iii) that most of the intracellular ions must be free to move under the influence of an electric field” [34].
By proving the electrical communication of Purkinje cells, Weidmann’s study provided the earliest example of direct cell-cell communication in vertebrates. In addition, and more importantly, it further proved the “healing-over” ability of these cells, a phenomenon first described by Theodor W. Engelmann [35] and later confirmed by Karl E. Rothschuh [36]. This phenomenon, now called cell-cell uncoupling, is mediated by the gating mechanism of gap junction channels.

4. Cell-to-Cell Communication in the Vertebrate Nervous System

Aside from Weidmann’s study on the mammalian heart [34], in the late 1950s, evidence of direct cell-cell communication was only reported in electrically excitable invertebrate cells. Therefore, the 1959′s report on electrical coupling in supra-medullary neurons of a vertebrate made the news [37]. This important discovery proved that electrical coupling is not restricted to excitable invertebrate cells. Soon after, electrical coupling was also demonstrated in two other vertebrate cells: the caliciform synapses of chick’s ciliary ganglion [38,39] and goldfish Mauthner neurons [40].

5. Direct Cell-to-Cell Communication in Unexcitable Cells

All examples of cell-cell coupling mentioned above were in electrically excitable cells, so no one expected that unexcitable cells would communicate directly. This was proven wrong in 1964 when two major discoveries demonstrated that all cells, but circulating cells, are coupled. Two independent studies published evidence of direct cell-cell communication in virtually all cells: one on insect glands [11,12] and the other on leech glia [13]. Interestingly, these discoveries were accidental.
Indeed, Yoshinobu Kanno and Werner R. Loewenstein were studying the electrical properties of the nuclear envelope in gland cells of Drosophila flavorepleta larvae and were unaware that adjacent gland cells might directly communicate with each other. Much to their surprise, with electrical current injection into the nucleus of one cell, the membrane potential changed not only in the injected cell but also in the neighboring cells [11,12]. Stephen W. Kuffler and David D. Potter also accidentally discovered glial electrical coupling, as their study was trying to determine whether neurons interact with glial cells [13]. These two studies opened an exciting new chapter in the history of cell biology because, after that, no one questioned whether cells were independent units of tissue.

Gap Junction Structure

Direct cell-cell communication is mediated by channels grouped at cell contact domains known as gap junctions (Figure 1). Each channel is made of two hemichannels (connexons in vertebrates and innexons in invertebrates) that form a hydrophilic pathway spanning two apposed plasma membranes and a narrow extracellular space (gap). In turn, each hemichannel is formed by the radial interaction of six proteins known as connexins in vertebrates and innexins in invertebrates (Figure 1). Connexins/innexins are intramembrane proteins that cross the membrane thickness four times (trans-membrane chains) and contain two extracellular loops and three cytoplasmic domains: a short NH2-terminus, a cytoplasmic loop, and a COOH-terminus domain of various lengths (Figure 2). For channels functioning as cell-to-cell pathways well insulated from the extracellular medium, their frameworks (innexons/connexons) must be in register with each other and bind to each other to bridge the extracellular gap (Figure 1).

6. Chemical Gating of Gap Junction Channels

Gap junction channels are chemically gated by a rise in cytosolic calcium or hydrogen ion concentrations ([Ca2+]i and [H+]i, respectively). We have reported that cytosolic acidification activates the gating mechanism via a secondary [Ca2+]i increase in the high nanomolar to the low micromolar range [41,42]. Over the years, we have proposed a direct role of Ca-calmodulin (Ca-CaM) via a cork-like pore-plugging mechanism, probably involving conformational changes in connexins as well (Figure 3); rev. in [43,44,45,46].
The model proposes two types of CaM-driven gating: “Ca-CaM-Cork” and “CaM-Cork”. In the first, gating involves Ca2+-induced CaM-activation. In the second, gating takes place without [Ca2+]i rise. The Ca-CaM-Cork gating is only reversed by a return of [Ca2+]i to resting values, while the CaM-Cork gating is reversed by trans-junctional voltage (Vj) positive at the gated side [45,47].

7. Direct Cell-Cell Communication via Membrane Pores

In the spring of 1968, while we were working on the ultrastructure of gap junctions between lateral giant axons of crayfish located at the ganglia of the ventral nerve cord, we were startled to see areas of cell contact between axons and sheath glial cells in which the two plasma membranes appeared to fuse forming clear images of membrane pores (Figure 4, Figure 5 and Figure 6). Several pores were usually grouped in the same region (Figure 4). In these regions, there were often aggregates of endoplasmic reticulum (ER) tubules, mitochondria, and other membrane structures (Figure 5). The pores were generally 15–20 nm in size (Figure 4), but larger pores were also seen (Figure 5 and Figure 6). Pores of this size are expected to allow wide communication between axoplasm and glial cell cytoplasm. It seemed unlikely that such structures are artifacts of the preparative procedure because these vascular perfused samples’ membranes and other cellular structures were always extremely well preserved (Figure 4, Figure 5 and Figure 6). Indeed, pores between axons and sheath glial cells seemed reasonable from a theoretical viewpoint because only passageways of this size could allow the exchange of molecules such as RNA and proteins as heavy as 200,000 molecular weight (MW) [48,49]. The pores may form transiently but are unlikely to be a preparation artifact because the radius of membrane fusion at the pores (Figure 4, inset “b”) is consistent with the minimum curvature radius of biological membranes.
If indeed pores existed, it would have meant that axons and sheath glial cells could exchange larger molecules than those exchanged by gap junctions. In those years, evidence for the transfer of molecules as large as proteins from glial cells to axons had been reported [50,51]. Still, most of the data supported the transfer from glial cells to axons only, and mechanisms different from cell-cell pores were proposed. We felt that the pores were probably a fixation artifact and decided not to publish these data.
However, the findings of the Viancour’s team contacted us a decade later, asking if we had ever seen gap junctions between crayfish axons and their adjacent sheath glial cells. Viancour asked this question because his team had found that when crayfish median giant axons were intracellularly injected with the fluorescent dye Lucifer Yellow CH, the dye rapidly diffused from the axoplasm to the cytoplasm of the adjacent glial cells (Grossfeld, R.M. et al. Soc. Neurosci. Abstr. 6, 385, 1980). The images he sent us were startling as they showed that the nuclei of the glial cells bordering the giant axon were stained by the fluorescent dye (Figure 4, inset “a”). In our answer, we mentioned that while we had reported specialized regions in which axon and glial cell surface membranes were regularly curved and projected into the axoplasm [52], the membranes of these regions of axon-glia apposition were separated by a gap of 130–140 Å, which is much wider than the typical 20 Å gap of gap junctions. Moreover, in freeze-fracture replicas, the structures at the projections did not bridge the extracellular space, so they could not contain cell-cell channels [52], answering the Viancour team’s claim that no gap junction existed between axons and glia.
But, Viancour’s findings prompted us to reconsider our earlier interpretation of the pores and immediately decided to publish the data in Nature [53]. Incidentally, our Nature’s publication, accompanied by our picture on the front page (Figure 7), caused a big uproar. The principal investigator of gap junctions in the nervous system wrote a rebuttal to Nature suggesting the artifactual nature of the pores. However, Nature accepted our answer to his comments and refused to publish the rebuttal in “Matters Arising.”
Soon after our Nature’s publication, Viancour’s team published their data in Nature as well [54]. Indeed, since our original observation of the late 1960s, evidence for the exchange of molecules much larger than gap junction permeants between axons and sheath glial cells of crayfish and squid had surfaced [48,49,55,56,57], indicating that in some tissues, and exceptional circumstances, cell-cell communication could also be mediated by membrane pores. In the four decades that followed our 1981’s publication [53], only one study on crayfish stretch receptors confirmed the presence of membrane pores [58]. Therefore, for several decades the possibility that some cells might be able under special conditions to directly communicate via membrane pores was by and large ignored.
In 2004, however, Rustom and coworkers [59] reported the presence of nanotubular structures between cells. They proposed a novel biological means of cell-to-cell interaction based on membrane continuity and intercellular transfer of organelles. Following this provocative report [59], several publications confirmed that some cells are capable of interacting directly with other cells via tubular membranous structures named “tunneling nanotubes” (TNT), capable of creating large membranous pores by fusing with the plasma membrane of the receiving cell and transferring cytoplasmic organelles and large as mitochondria; rev. in [60,61,62,63,64] among others. Evidence of tunneling nanotubes finally confirmed our data suggesting that some neighboring cells can communicate directly with each other via membrane pores [53].

8. TNT or Cytonemes, a New Concept for a Novel Long-Range

8.1. Cell-to-Cell Communication System

Both TNTs and cytonemes are thin actin-based long processes that communicate two or more cells. Two different kinds of TNTs or cytonemes have been described as open-ended (a long-range pore that communicates the cytoplasm of 2 or more cells) and a synaptic type of TNT/cytoneme connection like neuronal synapses. The work of Dr. Thomas Kornberg in Drosophila [65,66], as well as the early description of cell-to-cell pores [53], opened a new system of communication that had multiple benefits, including, first, a targeted delivery of a ligand into a targeted cell preventing the diffusion of the ligand into the extracellular space. Second, delivering a high concentration of a factor or receptor at a long distance, and third, establishing a syncytium that can extend up to 500 µm in tissues. Third, a complex transfer of organelles and associated second messengers can change the targeted cell’s metabolism and differentiation. Thus, TNTs/cytoneme/pores provide a new dimension to the cell-to-cell communication that can expand for millimeters depending on the extent of the processes and type of connections. Description of these kinds of cell-to-cell communications has been described in Drosophila wing imaginal disc [65]; abdominal histoblastnest [67], gonadal stem cell niche [68], zebrafish neural plate [69], Chick limb bud [70], and human embryonic stem cells [71]. However, as described below also, several pathogens use these developmental pathways to amplify inflammation and infection.
Early studies were limited to electron microscopy analysis. Several new approaches have confirmed these processes’ essential role in the development of several pathological conditions such as bacterial, viral, and cancer types, as well as acute conditions such as stroke and tissue recovery/resolution. In the developmental examples, most of the Hedgehog (Hh), decapentaplegic (Dpp), wingless (Wg/Wnt), and branchless (Bnl) growth factors are dependent on cytonemes formation and contact with the targeted cell. Preventing these long-range specific signaling compromises development [66,72]. In addition, reevaluation of pioneer data in cell-to-cell communication can be further proven and explained. Thus, a new area or dimension of cell-to-cell communication was born.

8.2. Tunneling Nanotubes (TNT)

The development of live-cell imaging and better technologies to fix, identify and characterize the structure and function of these thin processes was essential for obtaining reliable data. TNT have been described relatively recently (early 2000), which may be due to their small diameters and fragility upon fixation. TNTs are long membrane extensions that allow the exchange of several small molecules, vesicles, mitochondria, and pathogenic components, including bacteria, viruses, and pathogenic proteins. TNTs are around 50 to 200 nm in diameter and several µm in length with no contact with the substrate. Works of several laboratories demonstrated that TNT is positive for f-actin, myosin Va, and myosin X, GAP-43, 14-3-3γ, but mostly tubulin negative [60,73,74,75,76]. However, depending on the cell type, differentiation and treatment examined, significant differences in size, diameter, composition (actin and tubulin), transport capacity (transport of molecules), permeability (cut off of the exchanged molecules or components or electrical signals), stability (second, minutes or hours), and regulation has been described [77,78,79].
Several groups have suggested, especially using cell lines, that TNTs are formed from filopodium [59,76,80]. However, our experiments in primary cells indicate that TNT and filopodium are formed in different cellular areas and play different roles (see details below). In conclusion, it is essential to clarify and standardize the terminology or classification of these processes and define a TNT process versus other processes and cell-to-cell pores.
Experiments in several cell lines indicate that TNT has heterogeneous morphology, composition, lengths, and thickness and could form by different mechanisms. TNTs are transient structures with variable lifetimes ranging from a few minutes to hours (see a compressive review [81]. Depending on the TNT analyzed, TNT transfer organelles, ER-related vesicles, endosomal/lysosomal vesicles, mitochondria, and electric and small second messengers, including small proteins, microRNAs, calcium, IP3, and several other molecules. The exact formation and transfer mechanism are unknown, but the recent developments in cell-to-cell communication and inter-organelle interactions further developed these research areas [61,82,83,84].
Overall, TNT differs from filopodia in several ways. First, TNT structures establish physical contact between connected cells, while filopodia or long processes do not. Second, the length of the TNT differs from filopodium but can be similar to the non-connecting process that looks like TNT. Filopodia are 2.0 to 6 µm long, while TNT can reach 150 µm. In contrast, both have diameters between 50 to 200 nm. However, this is upon debate, as will be discussed below. These differences have been described using light and confocal microscopy, but better resolution shows multiple processes per TNT. Third, TNT communication requires exchanging cellular or pathogenic components between communicated cells, including second messengers, electrical signals, cytoplasm, vesicles, mitochondria, and pathogenic agents. Fourth, these processes are formed from a different cellular area than filopodium in primary cells. Filopodium and other TNT-like structures require interaction with substrates; TNT does not. Fifth, identifying at least two different kinds of TNTs, open-ended and synaptic, opens the possibility of having pores at long range with the subsequent consequences of sharing organelles and large molecules.

8.3. Major Differences between Gap Junctions, Pores, and TNT

The main differences between these communication systems are the distance between connected cells and the size of the molecules exchanged between connected cells. In the case of gap junction channels and cellular pores, the main limitation of these communication systems is the dilution factor of the cytoplasmic molecules exchanged between communicated cells. Most molecules exchanged by gap junctions/pores are small second messengers up to 1.5 kDa, including cAMP, cGMP, IP3, ATP, calcium, glutamate, aspartate, and small peptides and RNA, but no organelles or big protein complexes can be transferred (see [85]). Despite that, diffusion of these second messengers is a major limitation; auto regenerative long-range calcium waves going through gap junctions have been described [86,87]. An example of this long-distance communication system mediated by gap junctions has been observed in the liver but depends on cell-to-cell contact. Hepatocytes expressing vasopressin or glucagon receptors are only localized in the pericentral area of the acini. However, most hepatocytes that respond to vasopressin and glucagon stimulation by degrading glycogen are in the periportal area, at least 100 µm behind the blood flow. Despite this odd localization, upon binding vasopressin or glucagon to their receptors in the pericentral area, retrograde calcium waves are generated and transmitted through gap junctions by an intracellular pathway against the blood flow to reach specialized hepatocytes in the periportal area to release glucose [86,87]. Thus, even though diffusion is a limitation between gap junctions in communicated cells, several systems evolved to amplify these signals to other communicated cells. In the cell-to-cell pores, the possibilities are multiple, including the transfer of cytoplasmic soluble components, organelles, complexes of organelles, and even genetic material due to the large pore size observed by electron microscopy, 130–140 Å [53,58] (Figure 4, Figure 5 and Figure 6).
TNTs, due to a targeted transmission, diffusion of second messengers or pathogens is minimal. Also, to exchange second messengers, TNT allows the exchange of cellular materials, including cellular cargos and second messengers such as vesicles, MCH class I molecules, mitochondria, IP3 receptors, and endoplasmic reticulum (ER), calcium and electrical signals [60,88,89,90,91]. Most of the transport through TNT is actin-dependent because blocking actin reduces the transfer of these cellular components [59,92].
Even though TNT is a novel discovery, several TNT-like structures have been previously described in neuronal cells, migratory cells, pathfinding processes, and invadopodia in the case of tumor cells. Also, re-examinations of older reports show TNT-like structures in several inflammatory conditions. For example, in in vitro pathological conditions, the formation of TNT-like structures has been observed after infection with Listeria monocytogenes and Mycobacterium bovis [93,94,95]; in astrocytes treated with H2O2 [96], microglia activated with PMA and calcium ionophore [97], monocyte/macrophages treated with LPS plus IFN-γ [98], mouse neuronal CAD cells infected with exogenous PrP [99] and more recently in lymphocytes infected with HIV [91] and in human macrophages infected with HIV (Figure 8) [79]. In vivo, TNT-like structures have been observed in Drosophila [100,101], between immune cells in lymph nodes (see review by [89,102]), in the dendritic cells (DC) of the gut [103,104], and the MHC class II+ cells in the mouse cornea [105]. The clinical relevance of TNT has already been demonstrated in several pathological conditions (see below). As an example, Mesenchymal Stromal Cells (MSC) have been shown to improve the clearance of bacteria in models of acute respiratory distress syndrome (ARDS) by mitochondrial transfer from MSC to macrophages via tunneling nanotubes [63]. However, all new and old reports require the classification of these processes.

8.4. TNT-like Structures In Vivo

Currently, most of the data available about TNT have been obtained in vitro. However, whether these processes are present in vivo is still debated, mainly due to the unclear definition and lack of makers to identify TNTs. However, a consensus point for several laboratories is that TNT requires communication with another cell and exchange signaling, cargo, or organelles components. So far, in vivo, these two components have not been proven.
Currently, several suggestive publications show TNT-like structures in MHC class II dendritic cells in the corneal stroma [105], non-neural ectoderm cells in the midbrain [106], trophectoderm cells in the neural crest [107], epiblast cells in the blastula [108] and the ectoderm [109]. Also, several groups have suggested that neuronal processes may be similar to TNT. However, we will exclude from this review this possibility because dendrites are a clear formation, transport, and stability and lack many components of TNT.
One of the best examples of TNT in vivo has been described in flies. Only recently, the role of TNT or cytoneme has been well described for several groups, indicating the critical role of these processes in the signaling and delivery of developmental proteins such as Dpp, fibroblast growth factor (FGF), Hedgehog, Wingless (in flies), sonic Hedgehold (chick limbs), and wnt proteins (in developing zebrafish) directly into sites of cell to cell contact without dilution of these factors into the extracellular space [67,68,110,111,112,113]. TNT is a changing paradigm in the area because it was assumed that most of these proteins were released into the extracellular space. Roy et al. [114] describe that Dpp and FGF signaling is required for signal-producing wing disc cells, and several of the genes activated by these factors are required for TNT (cytonemes) [114,115] or synapses formation [114,115,116]. Most of the evidence seems to indicate that other factors important in the development of flies are TNT-mediated, such as fibroblast growth factor branchless [117], Dpp [113,114], EGF, Hh [67], SHh [70], and Notch [114,115,116,118]. Most of these factors are also present in humans, but the role of TNTs is unknown. Thus, it may be possible to extrapolate some of these data into human development.
In chicken embryos, the filopodia-like protrusion (or TNT-like processes) span the sub-ectodermal space to establish contact with the ectoderm. These actin and tubulin positive processes require Rac1 to form and participate in the retrograde trafficking of the transmembrane Wnt-receptor Frizzled-7. Another great example of TNT-like processes has been described in zebrafish embryos; several manuscripts describe their key role in controlling the spread of Wnt morphogens such as wnt8a that activates Wnt/β-catenin signaling pathways upon TNT contact. The formation and transfer of these molecules were cdc42/N-wasp dependent [119]. However, experiments in chicks suggest that shh-positive transfer and spread are dependent on long TNT-like processes (hundreds of micrometers) that are cdc42 negative [70]. Thus, in these experiments, using actin blockers such as latrunculin B, cytochalasin D, or overexpression of IRSp53 reduces filopodium formation, suggesting that actin assembling and f-actin-bundling are necessary for filopodium formation [69]. In these experiments, it has been suggested that the formation of filopodia (no-TNT) can be initiated in response to morphogenic factors such as FGF by the mDia3C mechanism [120]. Kondo’s laboratory (Osaka University, Japan) is a pioneer in identifying TNT-like structures in controlling zebrafish pigmentation [121,122]. This transfer of information between xanthophores extends processes (or dendrites) to melanophores, and contact with these processes induces the melanophores to migrate away, changing the pigmentation pattern [123].
In heart development, TNT-like processes participate in the cardiac differentiation of progenitor cells that give rise to the heart tube. In this case, filopodial extensions from the dorsal pericardial wall are directed into the endoderm and adjacent mesenchymal cells to coordinate signaling events related to the hedgehog, Notch, BMT, and non-canonical wnt pathways and to maintain cardiac progenitor status during heart tube extension [124]. Genetic deletion of tbx (22q11.2 deletion syndrome gene) and aPKCζ activator resulted in the loss of filopodia and decreased FGF signaling, reduced proliferation, and ectopic differentiation [124,125].

8.5. TNT Expression under Pathological Conditions

The data discussed above clearly indicate that TNT-like processes are expressed to deliver and spread in a specific manner wnt and notch-related morphogenic molecules during development. Thus, are pathogens that induce TNT for spread and inflammation using these mechanisms?
Interestingly, viruses, such as African swine fever, Ebola, Herpes Simplex, Marburg filoviruses, SARS-CoV-2, and Poxvirus Vaccinia encode viral factors or alter cell activation to induce the formation of filopodia structures to allow viral trafficking between the extracellular matrix or environment into cells [126,127,128,129,130,131,132], suggesting that viruses are adapted to use filopodia and TNT-like structures to improve viral spread.
Cytonemes (also called TNT, membrane tubulovesicular extensions, or TNT-like structures) have been described in neutrophils directly contacting yeast or bacteria. The role of TNT is to provide a directed delivery of anti-pathogen molecules at the site of infection. This group using human neutrophils describes that the mechanism of formation of these processes is GTPase dynamin or actin-dependent [133]. However, most of these reports lack an accurate characterization of these processes. These data indicate that pores and/or TNT can be present in activated immune cells.

8.6. Molecular Mechanism of TNT Formation

Despite the multiple questions in the TNT area, several groups using cell lines of different origins have described increased TNT formation in several stress conditions. Still, how they are formed and the signal(s) involved in directing formation and stability is unknown. Several reviews and primary communications indicate that M-sec, part of sec6 family proteins, and p53 activation are the key master regulator of TNT formation in several cell lines [134,135]. The mechanisms by which M-sec activates Ras-like small GTPase ral-A promote the binding to filamin. This protein cross-links actin filaments and promotes filopodia formation, as well as cdc42 [134,135]. Also, H2O2 treatment or serum starvation in neuronal cell lines is dependent on p53 and subsequent activation of epidermal growth factor (EGF) receptor, Akt1, PI3K, and mTOR [136]. Despite the beauty of this signaling system in cell lines, our data in primary human cells are confusing about the role of these proteins in TNT formation. Clearly, there is a missing link between primary cells and immortalized cell lines, where most of them have affected p53 expression and signaling.
Our data, using human primary macrophages or T cells, indicate that blocking M-sec expression using siRNA or treatment of primary cells with p53 activators such as CGK733, p53-stabilizing agents such as CP31398, reactivators of P53 mutant such as NSC319726, or MIRO-1, a proapoptotic p53 related protein, does not alter the formation of TNT in basal or in HIV-infected conditions or the form of TNT including fused open-ended TNT or synaptic kind of TNTs. Interestingly, several TNTs behave like pores (described above—direct cell to cell communication), though some were also sensitive to gap junction blockers suggesting that the synaptic kind of TNTs contain gap junctions (GJ). In addition, in different systems, functional GJ channels increase the invasion and dissemination of Shigella in epithelial cells [137] and increase toxicity between connected HIV-infected cells [138,139,140]. Suggesting the possibility that particular infectious agents, such as Shigella and HIV, can use GJ channels to sensitize uninfected cells and spread infection/toxicity to healthy cells, but whether TNT or TNT-like structures play a role in these infectious diseases is still under investigation. Alternatively, GJ benefits the host immune system by mediating a cross-antigen presentation phenomenon. This enables coupled cells to share viral peptides (antigens) and trigger a response in CTL cells, even when some cells were never directly exposed to the pathogen [141]. GJ-mediated immune coupling suggests the possibility that GJ expressed by monocytes/macrophages in inflammatory conditions cross-present antigens to lymphocytes and other inflammatory cells to maintain an immune memory in cells never exposed directly to specific antigen [141]. In the agreement, Cx43 is recruited to the immunological synapses during T cell priming, suggesting that GJ and hemichannels also participate in antigen presentation [142]. Thus, some pathogens such as HIV may potentially use GJ and maybe TNT (see below) to spread toxicity and its normal role in enhancing the immune response. However, most viral and bacterial infections down-regulate expression and GJ function, probably to reduce damage. Thus, pathogens require hijacking this communication system to spread toxicity and damage. We propose at least three kinds of direct cell to cell communication: GJ, pore mediated (close interactions), and TNT mediated (pore and synaptic type, long-range combination of both communication systems).
Several pioneer studies using T cells described the potential role of TNT in the propagation of Fas ligand-related cell death signals between communicated T lymphocytes [143,144]. Also, TNT has been proposed to provide an exchange pathway between injured cardiomyoblasts or endothelial cells by mesenchymal stem cells (MSC) through transferred mitochondria [145,146]. The direct transfer of mitochondria between connected cells, pores, or TNT provides metabolic advantages for metabolic distress cells, such as during stroke [147]. However, tumor cells utilize mitochondrial transfer in several types of cancers to adapt the tissue to the metabolic tumor environment [61,148,149,150,151,152]. However, whether these cells also show direct pore communication is unknown.

8.7. Cell-Cell Mitochondrial Transfer via Tunneling Nanotubes

Overall, TNTs are minimally expressed under physiological conditions [84,153]. However, they proliferate under stress and in response to several pathogens such as HIV, as well as pathogenic conditions such as cancer, Alzheimer’s, Parkinson’s, and other neurodegenerative diseases. The proposed function of TNT in pathogenic conditions is to amplify toxicity, apoptosis, and pathogenesis between connected cells. TNTs are generated in the “sick” cells into healthy surrounding cells to maximize efficiency in these conditions. In contrast, TNTs are also induced by stress conditions, including stroke, spinal cord, and traumatic brain injury. In these cases, TNTs are generated from compromised cells (cells in the penumbra area) into healthy areas. In these cases, it has been proposed that compromised cells “request” healthy factors and organelles to survive the damage in these conditions. These two mechanisms of TNT-induced formation, pathogenic and recovery, could be therapeutically targeted to regulate TNT formation and associated transport. In addition, several groups described exosomes containing mitochondria elements [154,155]. However, we will not discuss this type of mitochondrial element transfer for two reasons; first, released exosomes diffuse into the extracellular space losing the advantage provided by GJ, pores, and TNTs by maintaining a close mitochondrial transfer. This is essential to transfer enough organelles or their DNA to change the metabolism of the targeted cell. Second, there is a lack of proper cell-to-cell selectivity. This despite that exosomes had potential receptors to provide selectivity. Even though the exosomal mediated mitochondrial transfer or some of its elements can be exciting, whether the exosomal transfer can effectively change the phenotype of the targeted cell is still unclear.
Several groups suggested that TNT-transmitted organelles such as lysosomes and mitochondria could alter the fate of the recipient cells. Mitochondria are interesting TNT-transmitted organelles. In addition to their traditional role as cell powerhouses, mitochondria also play a key role in calcium buffering, ER interactions, ROS generation, apoptosis, ER stress response activation, and a broad variety of mitochondrial dysfunction. Furthermore, mitochondrial genetic alterations have been associated with metabolic diseases, including Alzheimer’s disease (AD) and Parkinson’s disease (PD), muscular dystrophy, and the process of normal aging [156]. Thus, TNT exchange of compromised mitochondria could result in accelerated disease.
The mitochondrial TNT-mediated exchange has been proposed as a general principle to rescue damaged cells and genetic diseases [157,158,159,160]. Mitochondrial transfer can rescue aerobic respiration in stem cells [161], mitochondrial function even if mtDNA is damaged [162], and epithelial function if mitochondria are transferred from stem cells [163]. However, few studies demonstrated that TNT could be the mechanism of mitochondrial transfer and, more importantly, the transferred mitochondria could change the TNT-targeted cell’s metabolism or function. For example, microinjection of intact mitochondria into oocytes prevented apoptosis [164]. These data indicate that these communication systems can change the metabolism of the targeted cell, probably due to the highly concentrated organelle transfer in a cell-to-cell specific manner.
Our data demonstrated that TNT generated in response to heterogeneous interaction between tumor cells and stromal cells promotes the transfer of a unique type of mitochondria from tumor cells into surrounding astrocytes resulting in their metabolic and hypoxic adaptation [61]. Currently, it is unknown whether the TNT-transferred mitochondria work independently in the targeted astrocytes, fused with the existing ones, or replace the mitochondrial system of non-cancer cells. However, blocking TNT mitochondrial transfer prevents healthy astrocytes’ adaptation to tumor-related conditions such as ischemia. More concerning is that radiation alone could induce TNT formation and transfer, suggesting that anti-cancer treatment may contribute to the tumor’s adaptation to subsequent treatment or tumor survival. In contrast, UV treatment induces the formation of TNT to enable the transfer of healthy mitochondria from healthy cells to rescue cell apoptosis with compromised DNA [165]. Both conditions indicate that DNA damage or compromise activates TNT formation and associated mitochondrial transfer.
Our data indicate that mitochondrial transfer enables “healthy cells,” such as primary astrocytes, to become tumor-like cells from a metabolic point of view. Furthermore, we identified a critical “fingerprint” of the TNT-transferred mitochondria in glioblastoma. Mitochondria expressed few genes and depended on nuclear transcription for the synthesis of all proteins for the organelle; however, we identified critical mutations in the mitochondrial DNA that favor the use of glutamine/glutamate instead of glucose and lipids to produce energy, a critical phenotype on aggressive tumors [166,167]. Glutamine use is a marker of aggressive glioblastoma [168]. Glutamine is a nitrogen source to synthesize nucleotides, amino acids, and ATP and is a major anaplerotic precursor for the TCA cycle. Glutamine dependency in different cancers promotes invasion and tumor aggressiveness [169]. Also, glutamine dependency can downregulate glycolysis reducing glucose uptake and lactate production, contributing to the Warburg effects observed in ischemic tumors. We identified that TNT formation also protects astrocytes from hypoxic conditions. Tumor persistence and growth relay in the survival of cancer stem-like cells promoted by a hypoxic microenvironment [170,171,172]. It is believed that low oxygen levels prevent ROS formation and DNA damage, nuclear and mitochondrial. Our data support both ideas that mitochondrial transfer changes the metabolism and contributes to adaptation to hypoxic conditions—both essential components to promote tumor growth.
Interestingly, TNTs are the only communication system that enables targeted and selected transfer of pathogenic material, including mitochondria. Thus, blocking TNT formation and associated communication could provide additional treatment to reduce tumor adaptation and prevent chemotherapeutic resistance [157,173,174,175,176,177]. Also, because TNTs are not expressed in healthy adult tissues, the expected toxic effects may be reduced.
We recently demonstrated TNT communication between heterogeneous glioblastoma tumor cells based on their resistance to TMZ and radiation therapy. First, surprisingly we identify that anti-tumor treatment promotes TNT formation and transport. Second, TNT concentrates the anti-apoptotic enzyme MGMT and distributes it among cells with insufficient MGMT expression to avoid apoptosis in response to TMZ and radiation treatment. Blocking TNTs prevented MGMT diffusion and survival of cells with minimal MGMT expression. Third, we identify that tumor TNTs are selective because MGMT protein, but not its mRNA, was transferred in vitro and in vivo in glioblastoma and breast cancer.
In agreement with the data in glioblastoma, in HIV-infected conditions, the mitochondrial transfer is essential to spread infection and associated inflammation. In immune cells, it has been demonstrated that TNT can propagate cell death induced by Fas ligand among T lymphocytes [143,144]; however, how amplification of disease and their protective mechanism remain unsolved. Our data on HIV indicate that viral spread is associated with mitochondrial function, not only to provide a formidable amount of ATP to induce the TNT formation and associated transfer but also because some of these organelles nurse some of the infectious HIV material transferred by TNTs. However, the contribution of these organelles is still unknown.
Several manuscripts identified specific molecules involved in mitochondrial transport, such as the protein Miro. Miro 1 and 2 are mitochondrial outer membrane-associated GTPases that regulate mitochondrial trafficking and distribution by coordinating actin- and microtubule-dependent mitochondrial movement due to interactions with TRAK and KIF5 [178]. It was reported that Miro1 knockout significantly inhibited microtubule-dependent mitochondrial motion, whereas Miro2 deficiency showed no effect on microtubule-dependent mitochondrial movement, suggesting that Miro1 is important for microtubule-dependent mitochondrial trafficking [179].
Several stimuli of TNT formation, such as the Fas-ligand receptor in the immune system or M-sec, a protein associated with the component Sec6 of the exocyst complex required for the docking of exocytic vesicles on the plasma membrane [143], have been discovered. Involved molecules in the TNT formation machinery are Rho and Ras small GTPases families such as Cdc42, Ral or the exocyst effector, myosin V/X, and the transmembrane MHC class III protein LST1. Also, Dephosphorylated β-Ca2+-Calmodulin-dependent protein kinase II (βCaMKII) has been shown to stabilize TNT formation [180]. βCaMKII not only stabilizes F-actin and binds to G-actin, preventing its nucleation, and in TNT, the protein is localized at the base of the TNT. These findings suggest that the TNT formation might be like filopodia and lamellipodia regulation mechanisms.
First, the interaction of M-Sec with RalA is necessary for the formation of TNT. Further, TNT formation has been associated with M-Sec expression. Also, RalA has been shown to interact with filamin to promote filopodia formation; however, whether these interactions play a role in TNT formation, stability, and associated transport is unknown.
Overall, identifying TNT as a new mechanism of long-range communication during development and pathological conditions opens several potential innovative therapeutic possibilities. First, TNTs are minimally expressed in healthy individuals; Second, TNTs proliferate upon pathogenesis; Third, pathogens such as viruses and bacteria use them to amplify inflammation and infection; and Fourth, traumatic conditions use TNT to rescue compromised cells. Thus, identifying potential blockers or inducers of TNT and the cargo associated with its formation can provide novel medicine and therapeutic approaches for life-threatening conditions.

8.8. Clinical Relevance of Tunneling Nanotubes

Currently, there are few examples of TNT in vivo, including in HIV animal models [77] and glioblastoma tumors [61,82,181]. However, observation of TNT processes in clinical samples has been proposed but not examined [182,183], though several laboratories are working on similar microtentacle (well-defined tubulin content) processes. Microtentacles are microtubule-based protrusions with high actin and vimentin content, as originally observed in detached breast cancer cells [184]. The main proposal is that detached cancer cells may influence the metastatic and chemoresistance profile of metastatic cells. Both these are also critical functions of TNT. However, a critical difference with TNT is that they do not communicate with cells until they establish contact with endothelial cells or tissues to invade. Thus, these microtentacles could establish TNT-mediated communication with other cells at one point. The growth of the microtentacles is regulated by kinesins and microtubule-associated proteins, including tau [185]. Also, cdc42 acts as molecular switches by alternating their active GTP-bound form and inactive GDP-bound form in cells. Aberrant cdc42 activity is implicated in tumor metastasis by promoting transendothelial migration [185,186]. Currently, several clinical trials block and/or promote microtentacle extension or attachment [185,187,188,189]; however, whether these drugs alter TNT formation or associated communication is unknown.
Interestingly, tau induces the formation of microtentacles and reduces the effects or binding of the anti-cancer drug paclitaxel [190]. However, these results had significant implications for cancer, Alzheimer’s disease, and other tau-related pathologies [191,192,193,194].
The identification of the mechanism of TNT formation (as well as other types of long communication processes) and associated function in cancer and other pathologies is an essential step to move forward in the TNT field from a mostly in vitro system into in vivo and the use of potential drugs to block or promote TNT formation. Block to prevent the spread of pathogenesis (cancer, Alzheimer’s disease, and other related diseases). In this case, TNT formation spread toxic organelles and second messengers into neighboring healthy cells. However, it also promotes TNT formation to rescue damaged cells from ischemic events such as stroke and traumatic events. In this case, healthy cells send healthy mitochondria and second messengers to compromised cells. These mechanisms denote a high impact of TNT formation and associated transport on human diseases that have not been exploited until now. In addition, there is no formation of TNTs in healthy individuals, increasing the possibility of not having side effects. Thus, the clinical and translational possibilities are highly significant for several incurable diseases and provide an alternative treatment for reducing brain/spinal cord physical damage or preventing/recovering ischemic events such as heart attacks and stroke.
A recent commentary [64] updated the present trends in mitochondrial research to three areas: (1) the causal role of mitochondrial dysfunction in several neurodegenerative ailments, such as Parkinson’s and Alzheimer’s disease; (2) the control of Ca2+ milIeu in selected intracellular micro-domains, changing intracellular responses; and (3) microvesicle (mV) or TNT-mediated transfer of mitochondria within different cells and tissues, of healthy or of degenerated mitochondria, as a mechanism of functional renewal in diseased cells, or fostering propagation of cell distress or pathology, such as in metastatic cancer evolution.
Until recently, mitochondria were considered to remain housed intracellularly for the life duration of each cell. The current review builds on the evidence that the intercellular exchange of mitochondria (and other intracellular organelles) continues during normal tissue growth and evolution. This exchange between cells appears robust during normal development. It continues in various pathological conditions, as demonstrated separately, especially in the lungs, kidneys, and the myocardium, especially in the ischemic brain’s repair. The earliest paper [195] demonstrated that bone marrow-derived mesenchymal stem cells (MSC) reduce acute lung injury in animals challenged by bacterial infection. They also suggested that MSC has a crucial role in preventing VILI (volume-induced lung injury) in healthy rats subjected to high volume ventilation. A potential therapeutic tool for patients emerged from this early paper, suggesting several clinical trials. In a later publication, Chen et al. [196] summarized in a table the several clinical application of mitochondrial transplantation by detailing a register of ongoing human clinical trials. This paper also discusses how mitochondrial transfer to damaged cells can help revive cells energetics in recipient cells.
Jackson et al. [63] demonstrated that mesenchymal stromal cell MSCs exert their antimicrobial effect via macrophage phagocytic activity, which can be mediated through mitochondrial transfer. In this model, fluorescent imaging and flow cytometry demonstrated extensive mitochondrial transfer from MSC to macrophages, which occurred at least partially through TNT structures. Finally, in a different opinion paper and a more recent review [197,198], we can appreciate the effect of mitochondrial transfer as a therapeutic strategy against ischemic stroke. These comments discuss how healthy mitochondria can be transferred to stroke cells during ischemic stroke.
Lippert and Borlongan [199] extend these concepts and elucidate the prophylactic use of hyperbaric oxygen treatment. This novel paper describes the mechanism of action of preventive treatments by hyperbaric oxygen exposure. For the first time, it demonstrates how a treatment preceding the exposure to a noxa could transfer the resistance mechanism to a tissue. Since oxygen levels inside a tissue are fleeting and can only last for a short time, the authors visualized directly mitochondrial transfer between cells induced by hyperbaric oxygen pretreatment. The process results in the transfer of healthy mitochondria and induces increased resistance of the receiving cells to the inflammatory response typical of ischemia and traumatic brain injury. These results are robust, with preconditioning doubling the salvage rate of the cells subjected to the experimental trauma, and explain the long-term effect of this technique which is clinically used in several surgical and medical patients.
A comprehensive review of mesenchymal stem cells and their ability to provide mitochondrial transfer has been organized by Cheng et al., 2019 [200]. In this review, the authors underline the improvement in clinical outcomes obtained in animal models of alveolar cells and pulmonary infection in improving the function of cardiomyocytes and the transfer of mitochondria via astrocyte into ischemic brain cells. However, it also demonstrates that cancer cells may enhance their ability to produce metastases by shifting damaged mitochondria and inducing chemoresistance. Because of this, the title of this review contains the definition: of a double-edged sword. The highlight of his article is a summary of different mechanisms of mitochondrial transfer via TNT and cell fusion or through GJ. The transfer mechanism is reviewed in this paper. It illustrates the formation of TNT between the recipient cell and the donor cell, or the organization of GJ, via the help of the connexin-43 or direct transfer of mitochondrial DNA from MSC cells via extracellular vesicles (eV), and also the possibility of selective loss of donor nuclei after complete cell fusion. They mediate the transfer of chemoresistance in a tumor and can be utilized as a clinical tool to provide pharmaceutical blockage sites and restoration of healthy tissue.
A recent review by Crew et al. [201] underlines how inter-organ transport of Mitochondria can be generated from the abundant supply provided by adipocytes. They can readily donate eV-encapsulated mitochondria from oxidatively damaged adipocytes. These are injected into the circulation and can be uptaken in cardiac tissue. This up-regulation of cardiac tissue results in resistance to cardiac ischemia, a paradoxical observation in obese patients, repeatedly reported.
Finally, in a recent review article Liu et al. [202] provide specific tables summarizing intercellular mitochondrial transfer in physiological conditions between cells and between different tissues and an extensive list of dozens of papers providing transfer in animal models and the various tissues studied so far. This comprehensive review also discusses all the mechanisms of transfer and especially the stimuli necessary to evoke the beginning of the intercellular transfer. They learn the means of organization and cell connection to propagate and initiate intercellular communication and transfer of organelles between cells. The evolution of this novel field will provide clinical novelty for maintaining organs suffering from aging and ischemic cells. And hopefully offer a new mechanism to support chemotherapy and limit cancer expansion.
Therefore, mitochondrial transfer is a frequent phenomenon associated with various bodily physiological and pathological activities. It may result in the restoration of normal physiological functions, and may help in the recovery from disease in animal models, as it already has in a few patient cases. Meanwhile, the mechanism of inhibiting transfer from stromal cells to cancer may develop the potential therapeutic target of a new anti-tumoral agent.

Author Contributions

All authors contributed to performing the experiments, analyzing the data, and writing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by grants from the National Institute of Mental Health, MH128082, the National Institute of Neurological Disorders and Stroke, NS105584, the UTMB Sealy Institute for Vaccine Sciences, and the UTMB Institute for Human Infection & Immunity (to EAE).

Institutional Review Board Statement

No applicable.

Informed Consent Statement

No applicable.

Data Availability Statement

No applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schleiden, M.J. Beiträge zur Phytogenesis. Arch. Anat. Physiol. Wiss. Med. 1839, 13, 137–176. [Google Scholar]
  2. Schwann, T. Mikroskopische Untersuchungen über die Uebereinstimmung in der Struktur und dem Wachsthum der Thiere und Pflanzen; Sander’schen Buchhandlung: Berlin, Germany, 1839. [Google Scholar]
  3. Turner, W. The Cell Theory, Past and Present. J. Anat. Physiol. 1890, 24, 253–287. [Google Scholar] [PubMed]
  4. Hodgkin, A.L.; Huxley, A.F. Propagation of electrical signals along giant nerve fibers. Proc. R. Soc. Lond. B Biol. Sci. 1952, 140, 177–183. [Google Scholar] [PubMed]
  5. Hodgkin, A.L.; Huxley, A.F. Currents carried by sodium and potassium ions through the membrane of the giant axon of Loligo. J. Physiol. 1952, 116, 449–472. [Google Scholar] [CrossRef]
  6. Hodgkin, A.L.; Huxley, A.F.; Katz, B. Measurement of current-voltage relations in the membrane of the giant axon of Loligo. J. Physiol. 1952, 116, 424–448. [Google Scholar] [CrossRef]
  7. Hodgkin, A.L.; Huxley, A.F.; Katz, B. Ionic currents underlying activiti in the giant axon of the squid. Arch. Sci. Physiol. 1949, 3, 129–150. [Google Scholar]
  8. Hodgkin, A.L.; Katz, B. The effect of sodium ions on the electrical activity of giant axon of the squid. J. Physiol. 1949, 108, 37–77. [Google Scholar] [CrossRef]
  9. Hille, B. Ion Channels of Excitable Membranes, 2nd ed.; Sinauer Associates, Inc.: Sunderland, MA, USA, 1992. [Google Scholar]
  10. Peracchia, C. (Ed.) Handbook of Membrane Channels. In Molecular and Cellular Physiology; Academic Press Inc.: San Diego, CA, USA, 1994. [Google Scholar]
  11. Kanno, Y.; Loewenstein, W.R. Low-resistance coupling between gland cells. Some observations on intercellular contact membranes and intercellular space. Nature 1964, 201, 194–195. [Google Scholar] [CrossRef]
  12. Loewenstein, W.R.; Kanno, Y. Studies on an epithelial (gland) cell junction. I. Modifications of surface membrane permeability. J. Cell Biol. 1964, 22, 565–586. [Google Scholar] [CrossRef] [Green Version]
  13. Kuffler, S.W.; Potter, D.D. Glia in the leech central nervous system: Physiological properties and neuron-glia relationship. J. Neurophysiol. 1964, 27, 290–320. [Google Scholar] [CrossRef]
  14. Stough, H.B. Giant nerve fibers of the earthworn. J. Comp. Neurol. 1926, 40, 409–463. [Google Scholar] [CrossRef]
  15. Stough, H.B. Polarization of the giant nerve fibers of the earthworm. J. Comp. Neurol. 1930, 50, 217–229. [Google Scholar] [CrossRef]
  16. Williams, L.W. Anatomy of the Common Squid; American Museum of Natural History: New York, NY, USA, 1909. [Google Scholar]
  17. Leydig, F. Zum Nervensystem und den Sinnesorganen der Würmer und Gleiderfüssler. In Tafeln zur Vergleichen Anatomie; H. Laupp: Tubingen, Germany, 1864. [Google Scholar]
  18. Young, J.Z. The functioning of the giant nerve fibres of the squid. J. Exp. Biol. 1938, 15, 170–185. [Google Scholar] [CrossRef]
  19. Johnson, G.E. Giant nerve fibers in crustaceans, with special reference to Cambarus and Palaemonetes. J. Comp. Neurol. 1924, 36, 323–375. [Google Scholar] [CrossRef]
  20. Eccles, J.C.; Granit, R.; Young, J.Z. Impulses in the giant nerve fibers of earthworm. J. Physiol. 1932, 77, 23P–25P. [Google Scholar]
  21. Bullock, T.H. Functional organization of the giant fiber system of Lumbricus. J. Neurophysiol. 1945, 8, 55–71. [Google Scholar] [CrossRef]
  22. Rushton, W.A.H. Action potentials from the isolated nerve cord of the earthworm. Proc. Roy. Soc. B 1945, 132, 423–437. [Google Scholar]
  23. Kao, C.Y.; Grundfest, H. Postsynaptic electrogenesis in septate giant axons. I. Earthworm median giant axon. J. Neurophysiol. 1957, 20, 553–573. [Google Scholar] [CrossRef]
  24. Wilson, D.M. The connections between the lateral giant fibers of earthworms. Comp. Biochem. Physiol. 1961, 3, 274–284. [Google Scholar] [CrossRef]
  25. Hama, K. Some observations on the fine structure of the giant nerve fibers of the earthworm, Eisenia foetida. J. Biophys. Biochem. Cytol. 1959, 6, 61–66. [Google Scholar] [CrossRef]
  26. Wiersma, C.A.G. Giant nerve fiber system of the crayfish. A contribution to comparative physiology of synapse. J. Neurophysiol. 1947, 10, 23–38. [Google Scholar] [CrossRef] [PubMed]
  27. Watanabe, A. The interaction of electrical activity among neurons of lobster cardiac ganglion. Jap. J. Physiol. 1958, 8, 305–318. [Google Scholar] [CrossRef] [PubMed]
  28. Hagiwara, S.; Watanabe, A.; Saito, N. Potential changes in syncytial neurons of lobster cardiac ganglion. J. Neurophysiol. 1959, 22, 554–572. [Google Scholar] [CrossRef] [PubMed]
  29. Watanabe, A.; Bullock, T.H. Modulation of activity of one neuron by subthreshold slow potentials in another in lobster cardiac ganglion. J. Gen. Physiol. 1960, 43, 1031–1045. [Google Scholar] [CrossRef] [Green Version]
  30. Watanabe, A.; Grundfest, H. Impulse propagation at the septal and commissural junctions of crayfish lateral giant axons. J. Gen. Physiol. 1961, 45, 267–308. [Google Scholar] [CrossRef] [Green Version]
  31. Reuben, J.P. Electrotonic connections between lobster muscle fibers. Biol. Bull. Woods Hole 1960, 49, 334. [Google Scholar]
  32. Hagiwara, S.; Morita, H. Electrotonic transmission between two nerve cells in leech ganglion. J. Neurophysiol. 1962, 25, 721–731. [Google Scholar] [CrossRef]
  33. Eckert, R. Electrical Interaction of Paired Ganglion Cells in the Leech. J. Gen. Physiol. 1963, 46, 573–587. [Google Scholar] [CrossRef] [Green Version]
  34. Weidmann, S. The electrical constants of Purkinje fibres. J. Physiol. 1952, 118, 348–360. [Google Scholar] [CrossRef]
  35. Engelmann, T.W. Vergleichende Untersuchungen zur Lehre von der Muskel und Nervenelektricität. Pflügers Arch. 1877, 15, 116–148. [Google Scholar] [CrossRef]
  36. Rothschuh, K.E. Über den funktionellen aufbaudes herzens aus elektrophysiologischen Elementen und üiber den mechanismus der erregungsleitung im herzen. Pflüg. Arch. 1951, 253, 238–251. [Google Scholar] [CrossRef]
  37. Bennett, M.V.; Crain, S.M.; Grundfest, H. Electrophysiology of supramedullary neurons in Spheroides maculatus. III. Organization of the supramedullary neurons. J. Gen. Physiol. 1959, 43, 221–250. [Google Scholar] [CrossRef] [Green Version]
  38. Martin, A.R.; Pilar, G. Dual mode of synaptic transmission in the avian ciliary ganglion. J. Physiol. 1963, 168, 443–463. [Google Scholar] [CrossRef]
  39. Martin, A.R.; Pilar, G. Transmission through the ciliary ganglion of the chick. J. Physiol. 1963, 168, 464–475. [Google Scholar] [CrossRef] [Green Version]
  40. Furukawa, T.; Furshpan, E.J. Two inhibitory mechanisms in the Mauthner neurons of goldfish. J. Neurophysiol. 1963, 26, 140–176. [Google Scholar] [CrossRef]
  41. Peracchia, C. Increase in gap junction resistance with acidification in crayfish septate axons is closely related to changes in intracellular calcium but not hydrogen ion concentration. J. Membr. Biol. 1990, 113, 75–92. [Google Scholar] [CrossRef]
  42. Lazrak, A.; Peracchia, C. Gap junction gating sensitivity to physiological internal calcium regardless of pH in Novikoff hepatoma cells. Biophys. J. 1993, 65, 2002–2012. [Google Scholar] [CrossRef] [Green Version]
  43. Peracchia, C.; Sotkis, A.; Wang, X.G.; Peracchia, L.L.; Persechini, A. Calmodulin directly gates gap junction channels. J. Biol. Chem. 2000, 275, 26220–26224. [Google Scholar] [CrossRef] [Green Version]
  44. Sotkis, A.; Wang, X.G.; Yasumura, T.; Peracchia, L.L.; Persechini, A.; Rash, J.E.; Peracchia, C. Calmodulin colocalizes with connexins and plays a direct role in gap junction channel gating. Cell Commun. Adhes. 2001, 8, 277–281. [Google Scholar] [CrossRef]
  45. Peracchia, C. Calmodulin-cork model of gap junction channel gating-one molecule, two mechanisms. Int. J. Mol. Sci. 2020, 21, 4938. [Google Scholar] [CrossRef]
  46. Peracchia, C. Gap junction stucture and chemical regulation. In Direct Calmodulin Role in Cell-To-Cell Channel Gating; Academic Press: London, UK, 2019. [Google Scholar]
  47. Peracchia, C.; Leverone Peracchia, L.M. Calmodulin-connexin partnership in gap junction channel regulation-Calmodulin-cork gating model. Int. J. Mol. Sci. 2021, 22, 13055. [Google Scholar] [CrossRef]
  48. Lasek, R.J.; Gainer, H.; Przybylski, R.J. Transfer of newly synthesized proteins from Schwann cells to the squid giant axon. Proc. Natl. Acad. Sci. USA 1974, 71, 1188–1192. [Google Scholar] [CrossRef] [Green Version]
  49. Lasek, R.J.; Gainer, H.; Barker, J.L. Cell-to-cell transfer of glial proteins to the squid giant axon. The glia-neuron protein trnasfer hypothesis. J. Cell Biol. 1977, 74, 501–523. [Google Scholar] [CrossRef]
  50. Singer, M.; Green, M.R. Autoradiographic studies of uridine incorporation in peripheral nerve of the newt, Triturus. J. Morphol. 1968, 124, 321–344. [Google Scholar] [CrossRef]
  51. Singer, M.; Salpeter, M.M. The transport of 3H-l-histidine through the Schwann and myelin sheath into the axon, including a reevaluation of myelin function. J. Morphol. 1966, 120, 281–315. [Google Scholar] [CrossRef]
  52. Peracchia, C. Excitable membrane ultrastructure. I. Freeze fracture of crayfish axons. J. Cell Biol. 1974, 61, 107–122. [Google Scholar] [CrossRef]
  53. Peracchia, C. Direct communication between axons and sheath glial cells in crayfish. Nature 1981, 290, 597–598. [Google Scholar] [CrossRef]
  54. Viancour, T.A.; Bittner, G.D.; Ballinger, M.L. Selective transfer of Lucifer yellow CH from axoplasm to adaxonal glia. Nature 1981, 293, 65–67. [Google Scholar] [CrossRef]
  55. Andersson, E.; Edstrom, A.; Jarlstedt, J. Properties of RNA from giant axons of the crayfish. Acta Physiol. Scand. 1970, 78, 491–502. [Google Scholar] [CrossRef]
  56. Gainer, H.; Tasaki, I.; Lasek, R.J. Evidence for the glia-neuron protein transfer hypothesis from intracellular perfusion studies of squid giant axons. J. Cell Biol. 1977, 74, 524–530. [Google Scholar] [CrossRef]
  57. Meyer, M.R.; Bittner, G.D. Histological studies of trophic dependencies in crayfish giant axons. Brain Res. 1978, 143, 195–211. [Google Scholar] [CrossRef]
  58. Fedorenko, G.; Neginskaya, M.; Fedorenko, A.; Uzdensky, A. The paired neuroglial and interglial membranes in the crayfish stretch receptor and their local disorganization. J. Neurosci. Res. 2015, 93, 707–713. [Google Scholar] [CrossRef] [PubMed]
  59. Rustom, A.; Saffrich, R.; Markovic, I.; Walther, P.; Gerdes, H.H. Nanotubular highways for intercellular organelle transport. Science 2004, 303, 1007–1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Abounit, S.; Zurzolo, C. Wiring through tunneling nanotubes—From electrical signals to organelle transfer. J. Cell Sci. 2012, 125, 1089–1098. [Google Scholar] [CrossRef] [Green Version]
  61. Valdebenito, S.; Malik, S.; Luu, R.; Loudig, O.; Mitchell, M.; Okafo, G.; Bhat, K.; Prideaux, B.; Eugenin, E.A. Tunneling nanotubes, TNT, communicate glioblastoma with surrounding non-tumor astrocytes to adapt them to hypoxic and metabolic tumor conditions. Sci. Rep. 2021, 11, 14556. [Google Scholar] [CrossRef]
  62. Eugenin, E.A.; Gaskill, P.J.; Berman, J.W. Tunneling nanotubes (TNT): A potential mechanism for intercellular HIV trafficking. Commun. Integr. Biol. 2009, 2, 243–244. [Google Scholar] [CrossRef]
  63. Jackson, M.V.; Morrison, T.J.; Doherty, D.F.; McAuley, D.F.; Matthay, M.A.; Kissenpfennig, A.; O’Kane, C.M.; Krasnodembskaya, A.D. Mitochondrial Transfer via Tunneling Nanotubes is an Important Mechanism by Which Mesenchymal Stem Cells Enhance Macrophage Phagocytosis in the in Vitro and in Vivo Models of ARDS. Stem. Cells 2016, 34, 2210–2223. [Google Scholar] [CrossRef] [Green Version]
  64. Pizzo, P.; Pozzan, T. Mitochondrialand: What will come next? Function 2021, 3, 1–2. [Google Scholar] [CrossRef]
  65. Chen, W.; Huang, H.; Hatori, R.; Kornberg, T.B. Essential basal cytonemes take up Hedgehog in the Drosophila wing imaginal disc. Development 2017, 144, 3134–3144. [Google Scholar] [CrossRef] [Green Version]
  66. Kornberg, T.B. Distributing signaling proteins in space and time: The province of cytonemes. Curr. Opin. Genet. Dev. 2017, 45, 22–27. [Google Scholar] [CrossRef]
  67. Bischoff, M.; Gradilla, A.C.; Seijo, I.; Andres, G.; Rodriguez-Navas, C.; Gonzalez-Mendez, L.; Guerrero, I. Cytonemes are required for the establishment of a normal Hedgehog morphogen gradient in Drosophila epithelia. Nat. Cell Biol. 2013, 15, 1269–1281. [Google Scholar] [CrossRef] [Green Version]
  68. Rojas-Rios, P.; Guerrero, I.; Gonzalez-Reyes, A. Cytoneme-mediated delivery of hedgehog regulates the expression of bone morphogenetic proteins to maintain germline stem cells in Drosophila. PLoS Biol. 2012, 10, e1001298. [Google Scholar] [CrossRef] [Green Version]
  69. Stanganello, E.; Hagemann, A.I.; Mattes, B.; Sinner, C.; Meyen, D.; Weber, S.; Schug, A.; Raz, E.; Scholpp, S. Filopodia-based Wnt transport during vertebrate tissue patterning. Nat. Commun. 2015, 6, 5846. [Google Scholar] [CrossRef] [Green Version]
  70. Sanders, T.A.; Llagostera, E.; Barna, M. Specialized filopodia direct long-range transport of SHH during vertebrate tissue patterning. Nature 2013, 497, 628–632. [Google Scholar] [CrossRef] [Green Version]
  71. Junyent, S.; Garcin, C.L.; Szczerkowski, J.L.A.; Trieu, T.J.; Reeves, J.; Habib, S.J. Specialized cytonemes induce self-organization of stem cells. Proc. Natl. Acad. Sci. USA 2020, 117, 7236–7244. [Google Scholar] [CrossRef] [Green Version]
  72. Wood, B.M.; Baena, V.; Huang, H.; Jorgens, D.M.; Terasaki, M.; Kornberg, T.B. Cytonemes with complex geometries and composition extend into invaginations of target cells. J. Cell Biol. 2021, 220, e202101116. [Google Scholar] [CrossRef]
  73. Las, G.; Shirihai, O.S. Miro1: New wheels for transferring mitochondria. Embo. J. 2014, 33, 939–941. [Google Scholar] [CrossRef] [Green Version]
  74. Austefjord, M.W.; Gerdes, H.H.; Wang, X. Tunneling nanotubes: Diversity in morphology and structure. Commun. Integr. Biol. 2014, 7, e27934. [Google Scholar] [CrossRef]
  75. Schiller, C.; Diakopoulos, K.N.; Rohwedder, I.; Kremmer, E.; von Toerne, C.; Ueffing, M.; Weidle, U.H.; Ohno, H.; Weiss, E.H. LST1 promotes the assembly of a molecular machinery responsible for tunneling nanotube formation. J. Cell Sci. 2013, 126, 767–777. [Google Scholar] [CrossRef] [Green Version]
  76. Gerdes, H.H.; Rustom, A.; Wang, X. Tunneling nanotubes, an emerging intercellular communication route in development. Mech. Dev. 2013, 130, 381–387. [Google Scholar] [CrossRef]
  77. Okafo, G.; Prevedel, L.; Eugenin, E. Tunneling nanotubes (TNT) mediate long-range gap junctional communication: Implications for HIV cell to cell spread. Sci. Rep. 2017, 7, 16660. [Google Scholar] [CrossRef]
  78. Malik, S.; Eugenin, E.A. Mechanisms of HIV Neuropathogenesis: Role of Cellular Communication Systems. Curr. HIV Res. 2016, 14, 400–411. [Google Scholar] [CrossRef] [Green Version]
  79. Eugenin, E.A.; Gaskill, P.J.; Berman, J.W. Tunneling nanotubes (TNT) are induced by HIV-infection of macrophages: A potential mechanism for intercellular HIV trafficking. Cell Immunol. 2009, 254, 142–148. [Google Scholar] [CrossRef] [Green Version]
  80. Wang, X.; Gerdes, H.H. Long-distance electrical coupling via tunneling nanotubes. Biochim. Biophys. Acta 2012, 1818, 2082–2086. [Google Scholar] [CrossRef] [Green Version]
  81. Sisakhtnezhad, S.; Khosravi, L. Emerging physiological and pathological implications of tunneling nanotubes formation between cells. Eur. J. Cell Biol. 2015, 94, 429–443. [Google Scholar] [CrossRef]
  82. Valdebenito, S.; Audia, A.; Bhat, K.P.L.; Okafo, G.; Eugenin, E.A. Tunneling Nanotubes Mediate Adaptation of Glioblastoma Cells to Temozolomide and Ionizing Radiation Treatment. iScience 2020, 23, 101450. [Google Scholar] [CrossRef]
  83. Eugenin, E.A. Role of cell-to-cell communication in cancer: New features, insights, and directions. Cancer Rep. (Hoboken) 2019, 2, e1228. [Google Scholar] [CrossRef] [Green Version]
  84. Valdebenito, S.; Lou, E.; Baldoni, J.; Okafo, G.; Eugenin, E. The Novel Roles of Connexin Channels and Tunneling Nanotubes in Cancer Pathogenesis. Int. J. Mol. Sci. 2018, 19, 1270. [Google Scholar] [CrossRef] [Green Version]
  85. Harris, A.L. Connexin specificity of second messenger permeation: Real numbers at last. J. Gen. Physiol. 2008, 131, 287–292. [Google Scholar] [CrossRef] [Green Version]
  86. Eugenin, E.A.; Gonzalez, H.; Saez, C.G.; Saez, J.C. Gap junctional communication coordinates vasopressin-induced glycogenolysis in rat hepatocytes. Am. J. Physiol. 1998, 274, G1109–G1116. [Google Scholar] [CrossRef]
  87. Nathanson, M.H.; Burgstahler, A.D.; Mennone, A.; Fallon, M.B.; Gonzalez, C.B.; Saez, J.C. Ca2+ waves are organized among hepatocytes in the intact organ. Am. J. Physiol. 1995, 269, G167–G171. [Google Scholar] [CrossRef] [PubMed]
  88. Onfelt, B.; Davis, D.M. Can membrane nanotubes facilitate communication between immune cells? Biochem. Soc. Trans. 2004, 32, 676–678. [Google Scholar] [CrossRef] [PubMed]
  89. Onfelt, B.; Nedvetzki, S.; Yanagi, K.; Davis, D.M. Cutting edge: Membrane nanotubes connect immune cells. J. Immunol. 2004, 173, 1511–1513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Gerdes, H.H.; Carvalho, R.N. Intercellular transfer mediated by tunneling nanotubes. Curr. Opin. Cell Biol. 2008, 20, 470–475. [Google Scholar] [CrossRef] [PubMed]
  91. Sowinski, S.; Jolly, C.; Berninghausen, O.; Purbhoo, M.A.; Chauveau, A.; Kohler, K.; Oddos, S.; Eissmann, P.; Brodsky, F.M.; Hopkins, C.; et al. Membrane nanotubes physically connect T cells over long distances presenting a novel route for HIV-1 transmission. Nat. Cell Biol. 2008, 10, 211–219. [Google Scholar] [CrossRef] [PubMed]
  92. Gurke, S.; Barroso, J.F.; Gerdes, H.H. The art of cellular communication: Tunneling nanotubes bridge the divide. Histochem. Cell Biol. 2008, 129, 539–550. [Google Scholar] [CrossRef] [Green Version]
  93. Dramsi, S.; Cossart, P. Intracellular pathogens and the actin cytoskeleton. Annu. Rev. Cell Dev. Biol. 1998, 14, 137–166. [Google Scholar] [CrossRef]
  94. Onfelt, B.; Nedvetzki, S.; Benninger, R.K.; Purbhoo, M.A.; Sowinski, S.; Hume, A.N.; Seabra, M.C.; Neil, M.A.; French, P.M.; Davis, D.M. Structurally distinct membrane nanotubes between human macrophages support long-distance vesicular traffic or surfing of bacteria. J. Immunol. 2006, 177, 8476–8483. [Google Scholar] [CrossRef] [Green Version]
  95. Wehland, J.; Carl, U.D. The sophisticated survival strategies of the pathogen Listeria monocytogenes. Int. Microbiol. 1998, 1, 11–18. [Google Scholar]
  96. Zhu, D.; Tan, K.S.; Zhang, X.; Sun, A.Y.; Sun, G.Y.; Lee, J.C. Hydrogen peroxide alters membrane and cytoskeleton properties and increases intercellular connections in astrocytes. J. Cell Sci. 2005, 118, 3695–3703. [Google Scholar] [CrossRef] [Green Version]
  97. Martinez, A.D.; Eugenin, E.A.; Branes, M.C.; Bennett, M.V.; Saez, J.C. Identification of second messengers that induce expression of functional gap junctions in microglia cultured from newborn rats. Brain Res. 2002, 943, 191–201. [Google Scholar] [CrossRef]
  98. Eugenin, E.A.; Branes, M.C.; Berman, J.W.; Saez, J.C. TNF-alpha plus IFN-gamma induce connexin43 expression and formation of gap junctions between human monocytes/macrophages that enhance physiological responses. J. Immunol. 2003, 170, 1320–1328. [Google Scholar] [CrossRef] [Green Version]
  99. Gousset, K.; Schiff, E.; Langevin, C.; Marijanovic, Z.; Caputo, A.; Browman, D.T.; Chenouard, N.; de Chaumont, F.; Martino, A.; Enninga, J.; et al. Prions hijack tunnelling nanotubes for intercellular spread. Nat. Cell Biol. 2009, 11, 328–336. [Google Scholar] [CrossRef]
  100. Hsiung, F.; Ramirez-Weber, F.A.; Iwaki, D.D.; Kornberg, T.B. Dependence of Drosophila wing imaginal disc cytonemes on Decapentaplegic. Nature 2005, 437, 560–563. [Google Scholar] [CrossRef]
  101. Kornberg, T. Pictures in cell biology. Cytonemes. Trends Cell Biol. 1999, 9, 434. [Google Scholar] [CrossRef]
  102. Gerdes, H.H.; Bukoreshtliev, N.V.; Barroso, J.F. Tunneling nanotubes: A new route for the exchange of components between animal cells. FEBS Lett. 2007, 581, 2194–2201. [Google Scholar] [CrossRef] [Green Version]
  103. Niess, J.H.; Brand, S.; Gu, X.; Landsman, L.; Jung, S.; McCormick, B.A.; Vyas, J.M.; Boes, M.; Ploegh, H.L.; Fox, J.G.; et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 2005, 307, 254–258. [Google Scholar] [CrossRef] [Green Version]
  104. Rescigno, M.; Urbano, M.; Valzasina, B.; Francolini, M.; Rotta, G.; Bonasio, R.; Granucci, F.; Kraehenbuhl, J.P.; Ricciardi-Castagnoli, P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat. Immunol. 2001, 2, 361–367. [Google Scholar] [CrossRef]
  105. Chinnery, H.R.; Pearlman, E.; McMenamin, P.G. Cutting edge: Membrane nanotubes in vivo: A feature of MHC class II+ cells in the mouse cornea. J. Immunol. 2008, 180, 5779–5783. [Google Scholar] [CrossRef] [Green Version]
  106. Pyrgaki, C.; Trainor, P.; Hadjantonakis, A.K.; Niswander, L. Dynamic imaging of mammalian neural tube closure. Dev. Biol. 2010, 344, 941–947. [Google Scholar] [CrossRef] [Green Version]
  107. Teddy, J.M.; Kulesa, P.M. In vivo evidence for short- and long-range cell communication in cranial neural crest cells. Development 2004, 131, 6141–6151. [Google Scholar] [CrossRef] [Green Version]
  108. Caneparo, L.; Pantazis, P.; Dempsey, W.; Fraser, S.E. Intercellular bridges in vertebrate gastrulation. PLoS ONE 2011, 6, e20230. [Google Scholar] [CrossRef] [Green Version]
  109. Miller, J.; Fraser, S.E.; McClay, D. Dynamics of thin filopodia during sea urchin gastrulation. Development 1995, 121, 2501–2511. [Google Scholar] [CrossRef]
  110. Bilioni, A.; Sanchez-Hernandez, D.; Callejo, A.; Gradilla, A.C.; Ibanez, C.; Mollica, E.; Carmen Rodriguez-Navas, M.; Simon, E.; Guerrero, I. Balancing Hedgehog, a retention and release equilibrium given by Dally, Ihog, Boi and shifted/DmWif. Dev. Biol. 2013, 376, 198–212. [Google Scholar] [CrossRef]
  111. Gradilla, A.C.; Gonzalez, E.; Seijo, I.; Andres, G.; Bischoff, M.; Gonzalez-Mendez, L.; Sanchez, V.; Callejo, A.; Ibanez, C.; Guerra, M.; et al. Exosomes as Hedgehog carriers in cytoneme-mediated transport and secretion. Nat. Commun. 2014, 5, 5649. [Google Scholar] [CrossRef] [Green Version]
  112. Kornberg, T.B. The contrasting roles of primary cilia and cytonemes in Hh signaling. Dev. Biol. 2014, 394, 1–5. [Google Scholar] [CrossRef] [Green Version]
  113. Roy, S.; Hsiung, F.; Kornberg, T.B. Specificity of Drosophila cytonemes for distinct signaling pathways. Science 2011, 332, 354–358. [Google Scholar] [CrossRef] [Green Version]
  114. Roy, S.; Huang, H.; Liu, S.; Kornberg, T.B. Cytoneme-mediated contact-dependent transport of the Drosophila decapentaplegic signaling protein. Science 2014, 343, 1244624. [Google Scholar] [CrossRef] [Green Version]
  115. Roy, S.; Kornberg, T.B. Paracrine signaling mediated at cell-cell contacts. Bioessays 2015, 37, 25–33. [Google Scholar] [CrossRef] [Green Version]
  116. Kornberg, T.B.; Roy, S. Cytonemes as specialized signaling filopodia. Development 2014, 141, 729–736. [Google Scholar] [CrossRef] [Green Version]
  117. Sato, M.; Kornberg, T.B. FGF is an essential mitogen and chemoattractant for the air sacs of the drosophila tracheal system. Dev. Cell. 2002, 3, 195–207. [Google Scholar] [CrossRef]
  118. Callejo, A.; Bilioni, A.; Mollica, E.; Gorfinkiel, N.; Andres, G.; Ibanez, C.; Torroja, C.; Doglio, L.; Sierra, J.; Guerrero, I. Dispatched mediates Hedgehog basolateral release to form the long-range morphogenetic gradient in the Drosophila wing disk epithelium. Proc. Natl. Acad. Sci. USA 2011, 108, 12591–12598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Kesavan, G.; Lieven, O.; Mamidi, A.; Ohlin, Z.L.; Johansson, J.K.; Li, W.C.; Lommel, S.; Greiner, T.U.; Semb, H. Cdc42/N-WASP signaling links actin dynamics to pancreatic beta cell delamination and differentiation. Development 2014, 141, 685–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Koizumi, K.; Takano, K.; Kaneyasu, A.; Watanabe-Takano, H.; Tokuda, E.; Abe, T.; Watanabe, N.; Takenawa, T.; Endo, T. RhoD activated by fibroblast growth factor induces cytoneme-like cellular protrusions through mDia3C. Mol. Biol. Cell. 2012, 23, 4647–4661. [Google Scholar] [CrossRef]
  121. Watanabe, M.; Kondo, S. Fish pigmentation. Comment on “Local reorganization of xanthophores fine-tunes and colors the striped pattern of zebrafish”. Science 2015, 348, 297. [Google Scholar] [CrossRef] [Green Version]
  122. Watanabe, M.; Kondo, S. Is pigment patterning in fish skin determined by the Turing mechanism? Trends Genet 2015, 31, 88–96. [Google Scholar] [CrossRef]
  123. Inaba, M.; Yamanaka, H.; Kondo, S. Pigment pattern formation by contact-dependent depolarization. Science 2012, 335, 677. [Google Scholar] [CrossRef] [Green Version]
  124. Cohen, M.; Georgiou, M.; Stevenson, N.L.; Miodownik, M.; Baum, B. Dynamic filopodia transmit intermittent Delta-Notch signaling to drive pattern refinement during lateral inhibition. Dev. Cell. 2010, 19, 78–89. [Google Scholar] [CrossRef] [Green Version]
  125. Vitelli, F.; Taddei, I.; Morishima, M.; Meyers, E.N.; Lindsay, E.A.; Baldini, A. A genetic link between Tbx1 and fibroblast growth factor signaling. Development 2002, 129, 4605–4611. [Google Scholar] [CrossRef]
  126. Cudmore, S.; Cossart, P.; Griffiths, G.; Way, M. Actin-based motility of vaccinia virus. Nature 1995, 378, 636–638. [Google Scholar] [CrossRef]
  127. Favoreel, H.W.; Van Minnebruggen, G.; Adriaensen, D.; Nauwynck, H.J. Cytoskeletal rearrangements and cell extensions induced by the US3 kinase of an alphaherpesvirus are associated with enhanced spread. Proc. Natl. Acad. Sci. USA 2005, 102, 8990–8995. [Google Scholar] [CrossRef] [Green Version]
  128. Gill, M.B.; Edgar, R.; May, J.S.; Stevenson, P.G. A gamma-herpesvirus glycoprotein complex manipulates actin to promote viral spread. PLoS ONE 2008, 3, e1808. [Google Scholar] [CrossRef]
  129. Hartlieb, B.; Weissenhorn, W. Filovirus assembly and budding. Virology 2006, 344, 64–70. [Google Scholar] [CrossRef] [Green Version]
  130. Jouvenet, N.; Windsor, M.; Rietdorf, J.; Hawes, P.; Monaghan, P.; Way, M.; Wileman, T. African swine fever virus induces filopodia-like projections at the plasma membrane. Cell Microbiol. 2006, 8, 1803–1811. [Google Scholar] [CrossRef]
  131. Kolesnikova, L.; Bohil, A.B.; Cheney, R.E.; Becker, S. Budding of Marburgvirus is associated with filopodia. Cell Microbiol. 2007, 9, 939–951. [Google Scholar] [CrossRef]
  132. Noda, T.; Ebihara, H.; Muramoto, Y.; Fujii, K.; Takada, A.; Sagara, H.; Kim, J.H.; Kida, H.; Feldmann, H.; Kawaoka, Y. Assembly and budding of Ebolavirus. PLoS Pathog. 2006, 2, e99. [Google Scholar] [CrossRef] [Green Version]
  133. Galkina, S.I.; Fedorova, N.V.; Serebryakova, M.V.; Arifulin, E.A.; Stadnichuk, V.I.; Gaponova, T.V.; Baratova, L.A.; Sud’ina, G.F. Inhibition of the GTPase dynamin or actin depolymerisation initiates outward plasma membrane tubulation/vesiculation (cytoneme formation) in neutrophils. Biol. Cell. 2015, 107, 144–158. [Google Scholar] [CrossRef]
  134. Ohno, H.; Hase, K.; Kimura, S. M-Sec: Emerging secrets of tunneling nanotube formation. Commun. Integr. Biol. 2010, 3, 231–233. [Google Scholar] [CrossRef] [Green Version]
  135. Kimura, S.; Yamashita, M.; Yamakami-Kimura, M.; Sato, Y.; Yamagata, A.; Kobashigawa, Y.; Inagaki, F.; Amada, T.; Hase, K.; Iwanaga, T.; et al. Distinct Roles for the N- and C-terminal Regions of M-Sec in Plasma Membrane Deformation during Tunneling Nanotube Formation. Sci. Rep. 2016, 6, 33548. [Google Scholar] [CrossRef] [Green Version]
  136. Wang, Y.; Cui, J.; Sun, X.; Zhang, Y. Tunneling-nanotube development in astrocytes depends on p53 activation. Cell Death Differ. 2011, 18, 732–742. [Google Scholar] [CrossRef] [Green Version]
  137. Tran Van Nhieu, G.; Clair, C.; Bruzzone, R.; Mesnil, M.; Sansonetti, P.; Combettes, L. Connexin-dependent inter-cellular communication increases invasion and dissemination of Shigella in epithelial cells. Nat. Cell Biol. 2003, 5, 720–726. [Google Scholar] [CrossRef]
  138. Eugenin, E.A.; Berman, J.W. Gap junctions mediate human immunodeficiency virus-bystander killing in astrocytes. J. Neurosci. Off. J. Soc. Neurosci. 2007, 27, 12844–12850. [Google Scholar] [CrossRef] [Green Version]
  139. Eugenin, E.A.; Berman, J.W. Cytochrome C dysregulation induced by HIV infection of astrocytes results in bystander apoptosis of uninfected astrocytes by an IP and Calcium dependent mechanism. J. Neurochem. 2013, 127, 644–651. [Google Scholar] [CrossRef] [Green Version]
  140. Eugenin, E.A.; Clements, J.E.; Zink, M.C.; Berman, J.W. Human immunodeficiency virus infection of human astrocytes disrupts blood-brain barrier integrity by a gap junction-dependent mechanism. J. Neurosci. 2011, 31, 9456–9465. [Google Scholar] [CrossRef] [Green Version]
  141. Neijssen, J.; Herberts, C.; Drijfhout, J.W.; Reits, E.; Janssen, L.; Neefjes, J. Cross-presentation by intercellular peptide transfer through gap junctions. Nature 2005, 434, 83–88. [Google Scholar] [CrossRef]
  142. Mendoza-Naranjo, A.; Bouma, G.; Pereda, C.; Ramirez, M.; Webb, K.F.; Tittarelli, A.; Lopez, M.N.; Kalergis, A.M.; Thrasher, A.J.; Becker, D.L.; et al. Functional Gap Junctions Accumulate at the Immunological Synapse and Contribute to T Cell Activation. J. Immunol. 2011, 187, 3121–3132. [Google Scholar] [CrossRef]
  143. Arkwright, P.D.; Luchetti, F.; Tour, J.; Roberts, C.; Ayub, R.; Morales, A.P.; Rodriguez, J.J.; Gilmore, A.; Canonico, B.; Papa, S.; et al. Fas stimulation of T lymphocytes promotes rapid intercellular exchange of death signals via membrane nanotubes. Cell Res. 2010, 20, 72–88. [Google Scholar] [CrossRef] [Green Version]
  144. Luchetti, F.; Canonico, B.; Arcangeletti, M.; Guescini, M.; Cesarini, E.; Stocchi, V.; Degli Esposti, M.; Papa, S. Fas signalling promotes intercellular communication in T cells. PLoS ONE 2012, 7, e35766. [Google Scholar] [CrossRef] [Green Version]
  145. Liu, K.; Ji, K.; Guo, L.; Wu, W.; Lu, H.; Shan, P.; Yan, C. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc. Res. 2014, 92, 10–18. [Google Scholar] [CrossRef]
  146. Cselenyak, A.; Pankotai, E.; Horvath, E.M.; Kiss, L.; Lacza, Z. Mesenchymal stem cells rescue cardiomyoblasts from cell death in an in vitro ischemia model via direct cell-to-cell connections. BMC Cell Biol. 2010, 11, 29. [Google Scholar] [CrossRef] [Green Version]
  147. Yip, H.K.; Dubey, N.K.; Lin, K.C.; Sung, P.H.; Chiang, J.Y.; Chu, Y.C.; Huang, C.R.; Chen, Y.L.; Deng, Y.H.; Cheng, H.C.; et al. Melatonin rescues cerebral ischemic events through upregulated tunneling nanotube-mediated mitochondrial transfer and downregulated mitochondrial oxidative stress in rat brain. Biomed. Pharm. 2021, 139, 111593. [Google Scholar] [CrossRef] [PubMed]
  148. Allegra, A.; Di Gioacchino, M.; Cancemi, G.; Casciaro, M.; Petrarca, C.; Musolino, C.; Gangemi, S. Specialized Intercellular Communications via Tunnelling Nanotubes in Acute and Chronic Leukemia. Cancers 2022, 14, 659. [Google Scholar] [CrossRef] [PubMed]
  149. Chinnery, H.R.; Keller, K.E. Tunneling Nanotubes and the Eye: Intercellular Communication and Implications for Ocular Health and Disease. Biomed. Res. Int. 2020, 2020, 7246785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Moore, J.A.; Mistry, J.J.; Rushworth, S.A. Mitochondria and the Tumour Microenvironment in Blood Cancer. Adv. Exp. Med. Biol. 2021, 1329, 181–203. [Google Scholar] [CrossRef] [PubMed]
  151. Saenz-de-Santa-Maria, I.; Bernardo-Castineira, C.; Enciso, E.; Garcia-Moreno, I.; Chiara, J.L.; Suarez, C.; Chiara, M.D. Control of long-distance cell-to-cell communication and autophagosome transfer in squamous cell carcinoma via tunneling nanotubes. Oncotarget 2017, 8, 20939–20960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Thayanithy, V.; Dickson, E.L.; Steer, C.; Subramanian, S.; Lou, E. Tumor-stromal cross talk: Direct cell-to-cell transfer of oncogenic microRNAs via tunneling nanotubes. Transl. Res. 2014, 164, 359–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Roehlecke, C.; Schmidt, M.H.H. Tunneling Nanotubes and Tumor Microtubes in Cancer. Cancers 2020, 12, 857. [Google Scholar] [CrossRef] [Green Version]
  154. Picca, A.; Guerra, F.; Calvani, R.; Bucci, C.; Lo Monaco, M.R.; Bentivoglio, A.R.; Coelho-Junior, H.J.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial Dysfunction and Aging: Insights from the Analysis of Extracellular Vesicles. Int. J. Mol. Sci. 2019, 20, 805. [Google Scholar] [CrossRef] [Green Version]
  155. Soto-Heredero, G.; Baixauli, F.; Mittelbrunn, M. Interorganelle Communication between Mitochondria and the Endolysosomal System. Front. Cell Dev. Biol. 2017, 5, 95. [Google Scholar] [CrossRef] [Green Version]
  156. Diaz-Vegas, A.; Sanchez-Aguilera, P.; Krycer, J.R.; Morales, P.E.; Monsalves-Alvarez, M.; Cifuentes, M.; Rothermel, B.A.; Lavandero, S. Is Mitochondrial Dysfunction a Common Root of Noncommunicable Chronic Diseases? Endocr. Rev. 2020, 41, bnaa005. [Google Scholar] [CrossRef]
  157. Singh, A.K.; Cancelas, J.A. Gap Junctions in the Bone Marrow Lympho-Hematopoietic Stem Cell Niche, Leukemia Progression, and Chemoresistance. Int. J. Mol. Sci. 2020, 21, 796. [Google Scholar] [CrossRef] [Green Version]
  158. Kolba, M.D.; Dudka, W.; Zareba-Koziol, M.; Kominek, A.; Ronchi, P.; Turos, L.; Chroscicki, P.; Wlodarczyk, J.; Schwab, Y.; Klejman, A.; et al. Tunneling nanotube-mediated intercellular vesicle and protein transfer in the stroma-provided imatinib resistance in chronic myeloid leukemia cells. Cell Death Dis. 2019, 10, 817. [Google Scholar] [CrossRef] [Green Version]
  159. Li, C.J.; Chen, P.K.; Sun, L.Y.; Pang, C.Y. Enhancement of Mitochondrial Transfer by Antioxidants in Human Mesenchymal Stem Cells. Oxidative Med. Cell. Longev. 2017, 2017, 8510805. [Google Scholar] [CrossRef]
  160. Astanina, K.; Koch, M.; Jungst, C.; Zumbusch, A.; Kiemer, A.K. Lipid droplets as a novel cargo of tunnelling nanotubes in endothelial cells. Sci. Rep. 2015, 5, 11453. [Google Scholar] [CrossRef] [Green Version]
  161. Spees, J.L.; Olson, S.D.; Whitney, M.J.; Prockop, D.J. Mitochondrial transfer between cells can rescue aerobic respiration. Proc. Natl. Acad. Sci. USA 2006, 103, 1283–1288. [Google Scholar] [CrossRef] [Green Version]
  162. Cho, Y.M.; Kim, J.H.; Kim, M.; Park, S.J.; Koh, S.H.; Ahn, H.S.; Kang, G.H.; Lee, J.B.; Park, K.S.; Lee, H.K. Mesenchymal stem cells transfer mitochondria to the cells with virtually no mitochondrial function but not with pathogenic mtDNA mutations. PLoS ONE 2012, 7, e32778. [Google Scholar] [CrossRef] [Green Version]
  163. Islam, M.N.; Das, S.R.; Emin, M.T.; Wei, M.; Sun, L.; Westphalen, K.; Rowlands, D.J.; Quadri, S.K.; Bhattacharya, S.; Bhattacharya, J. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med. 2012, 18, 759–765. [Google Scholar] [CrossRef] [Green Version]
  164. Perez, G.I.; Acton, B.M.; Jurisicova, A.; Perkins, G.A.; White, A.; Brown, J.; Trbovich, A.M.; Kim, M.R.; Fissore, R.; Xu, J.; et al. Genetic variance modifies apoptosis susceptibility in mature oocytes via alterations in DNA repair capacity and mitochondrial ultrastructure. Cell Death Differ. 2007, 14, 524–533. [Google Scholar] [CrossRef]
  165. Wang, X.; Gerdes, H.H. Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ. 2015, 22, 1181–1191. [Google Scholar] [CrossRef] [Green Version]
  166. Obara-Michlewska, M.; Szeliga, M. Targeting Glutamine Addiction in Gliomas. Cancers 2020, 12, 310. [Google Scholar] [CrossRef] [Green Version]
  167. Oizel, K.; Yang, C.; Renoult, O.; Gautier, F.; Do, Q.N.; Joalland, N.; Gao, X.; Ko, B.; Vallette, F.; Ge, W.P.; et al. Glutamine uptake and utilization of human mesenchymal glioblastoma in orthotopic mouse model. Cancer Metab. 2020, 8, 9. [Google Scholar] [CrossRef]
  168. Quinones, A.; Le, A. The Multifaceted Metabolism of Glioblastoma. Adv. Exp. Med. Biol. 2018, 1063, 59–72. [Google Scholar] [CrossRef]
  169. Simoes, R.V.; Serganova, I.S.; Kruchevsky, N.; Leftin, A.; Shestov, A.A.; Thaler, H.T.; Sukenick, G.; Locasale, J.W.; Blasberg, R.G.; Koutcher, J.A.; et al. Metabolic plasticity of metastatic breast cancer cells: Adaptation to changes in the microenvironment. Neoplasia 2015, 17, 671–684. [Google Scholar] [CrossRef] [Green Version]
  170. Da Ros, M.; De Gregorio, V.; Iorio, A.L.; Giunti, L.; Guidi, M.; de Martino, M.; Genitori, L.; Sardi, I. Glioblastoma Chemoresistance: The Double Play by Microenvironment and Blood-Brain Barrier. Int. J. Mol. Sci. 2018, 19, 2879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Qin, J.; Liu, Y.; Lu, Y.; Liu, M.; Li, M.; Li, J.; Wu, L. Hypoxia-inducible factor 1 alpha promotes cancer stem cells-like properties in human ovarian cancer cells by upregulating SIRT1 expression. Sci. Rep. 2017, 7, 10592. [Google Scholar] [CrossRef] [PubMed]
  172. Velaei, K.; Samadi, N.; Barazvan, B.; Soleimani Rad, J. Tumor microenvironment-mediated chemoresistance in breast cancer. Breast 2016, 30, 92–100. [Google Scholar] [CrossRef] [PubMed]
  173. Soundara Rajan, T.; Gugliandolo, A.; Bramanti, P.; Mazzon, E. Tunneling Nanotubes-Mediated Protection of Mesenchymal Stem Cells: An Update from Preclinical Studies. Int. J. Mol. Sci. 2020, 21, 3481. [Google Scholar] [CrossRef] [PubMed]
  174. Wang, J.; Liu, X.; Qiu, Y.; Shi, Y.; Cai, J.; Wang, B.; Wei, X.; Ke, Q.; Sui, X.; Wang, Y.; et al. Cell adhesion-mediated mitochondria transfer contributes to mesenchymal stem cell-induced chemoresistance on T cell acute lymphoblastic leukemia cells. J. Hematol. Oncol. 2018, 11, 11. [Google Scholar] [CrossRef]
  175. Omsland, M.; Bruserud, O.; Gjertsen, B.T.; Andresen, V. Tunneling nanotube (TNT) formation is downregulated by cytarabine and NF-kappaB inhibition in acute myeloid leukemia (AML). Oncotarget 2017, 8, 7946–7963. [Google Scholar] [CrossRef] [Green Version]
  176. Desir, S.; Dickson, E.L.; Vogel, R.I.; Thayanithy, V.; Wong, P.; Teoh, D.; Geller, M.A.; Steer, C.J.; Subramanian, S.; Lou, E. Tunneling nanotube formation is stimulated by hypoxia in ovarian cancer cells. Oncotarget 2016, 7, 43150–43161. [Google Scholar] [CrossRef] [Green Version]
  177. Pasquier, J.; Guerrouahen, B.S.; Al Thawadi, H.; Ghiabi, P.; Maleki, M.; Abu-Kaoud, N.; Jacob, A.; Mirshahi, M.; Galas, L.; Rafii, S.; et al. Preferential transfer of mitochondria from endothelial to cancer cells through tunneling nanotubes modulates chemoresistance. J. Transl. Med. 2013, 11, 94. [Google Scholar] [CrossRef] [Green Version]
  178. Macaskill, A.F.; Rinholm, J.E.; Twelvetrees, A.E.; Arancibia-Carcamo, I.L.; Muir, J.; Fransson, A.; Aspenstrom, P.; Attwell, D.; Kittler, J.T. Miro1 is a calcium sensor for glutamate receptor-dependent localization of mitochondria at synapses. Neuron 2009, 61, 541–555. [Google Scholar] [CrossRef] [Green Version]
  179. Lopez-Domenech, G.; Covill-Cooke, C.; Ivankovic, D.; Halff, E.F.; Sheehan, D.F.; Norkett, R.; Birsa, N.; Kittler, J.T. Miro proteins coordinate microtubule- and actin-dependent mitochondrial transport and distribution. EMBO J. 2018, 37, 321–336. [Google Scholar] [CrossRef]
  180. Ljubojevic, N.; Henderson, J.M.; Zurzolo, C. The Ways of Actin: Why Tunneling Nanotubes Are Unique Cell Protrusions. Trends Cell Biol. 2021, 31, 130–142. [Google Scholar] [CrossRef]
  181. Osswald, M.; Jung, E.; Sahm, F.; Solecki, G.; Venkataramani, V.; Blaes, J.; Weil, S.; Horstmann, H.; Wiestler, B.; Syed, M.; et al. Brain tumour cells interconnect to a functional and resistant network. Nature 2015, 528, 93–98. [Google Scholar] [CrossRef]
  182. Zurzolo, C. Tunneling nanotubes: Reshaping connectivity. Curr. Opin. Cell Biol. 2021, 71, 139–147. [Google Scholar] [CrossRef]
  183. McMillen, P.; Oudin, M.J.; Levin, M.; Payne, S.L. Beyond Neurons: Long Distance Communication in Development and Cancer. Front. Cell Dev. Biol. 2021, 9, 739024. [Google Scholar] [CrossRef]
  184. Whipple, R.A.; Cheung, A.M.; Martin, S.S. Detyrosinated microtubule protrusions in suspended mammary epithelial cells promote reattachment. Exp. Cell Res. 2007, 313, 1326–1336. [Google Scholar] [CrossRef] [Green Version]
  185. Matrone, M.A.; Whipple, R.A.; Thompson, K.; Cho, E.H.; Vitolo, M.I.; Balzer, E.M.; Yoon, J.R.; Ioffe, O.B.; Tuttle, K.C.; Tan, M.; et al. Metastatic breast tumors express increased tau, which promotes microtentacle formation and the reattachment of detached breast tumor cells. Oncogene 2010, 29, 3217–3227. [Google Scholar] [CrossRef] [Green Version]
  186. Maldonado, M.D.M.; Medina, J.I.; Velazquez, L.; Dharmawardhane, S. Targeting Rac and Cdc42 GEFs in Metastatic Cancer. Front. Cell Dev. Biol. 2020, 8, 201. [Google Scholar] [CrossRef]
  187. Whipple, R.A.; Balzer, E.M.; Cho, E.H.; Matrone, M.A.; Yoon, J.R.; Martin, S.S. Vimentin filaments support extension of tubulin-based microtentacles in detached breast tumor cells. Cancer Res. 2008, 68, 5678–5688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Shah, P.; Gau, Y.; Sabnis, G. Histone deacetylase inhibitor entinostat reverses epithelial to mesenchymal transition of breast cancer cells by reversing the repression of E-cadherin. Breast Cancer Res. Treat. 2014, 143, 99–111. [Google Scholar] [CrossRef] [PubMed]
  189. Perry, N.A.; Vitolo, M.I.; Martin, S.S.; Kontrogianni-Konstantopoulos, A. Loss of the obscurin-RhoGEF downregulates RhoA signaling and increases microtentacle formation and attachment of breast epithelial cells. Oncotarget 2014, 5, 8558–8568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Ross, J.L.; Santangelo, C.D.; Makrides, V.; Fygenson, D.K. Tau induces cooperative Taxol binding to microtubules. Proc. Natl. Acad. Sci. USA 2004, 101, 12910–12915. [Google Scholar] [CrossRef] [Green Version]
  191. Valentino, R.R.; Koga, S.; Walton, R.L.; Soto-Beasley, A.I.; Kouri, N.; DeTure, M.A.; Murray, M.E.; Johnson, P.W.; Petersen, R.C.; Boeve, B.F.; et al. MAPT subhaplotypes in corticobasal degeneration: Assessing associations with disease risk, severity of tau pathology, and clinical features. Acta Neuropathol. Commun. 2020, 8, 218. [Google Scholar] [CrossRef]
  192. Savioz, A.; Kovari, E.; Anastasiu, R.; Rossier, C.; Saini, K.; Bouras, C.; Leuba, G. Search for a mutation in the tau gene in a Swiss family with frontotemporal dementia. Exp. Neurol. 2000, 161, 330–335. [Google Scholar] [CrossRef]
  193. Rouzier, R.; Rajan, R.; Wagner, P.; Hess, K.R.; Gold, D.L.; Stec, J.; Ayers, M.; Ross, J.S.; Zhang, P.; Buchholz, T.A.; et al. Microtubule-associated protein tau: A marker of paclitaxel sensitivity in breast cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 8315–8320. [Google Scholar] [CrossRef] [Green Version]
  194. Rossi, G.; Bastone, A.; Piccoli, E.; Mazzoleni, G.; Morbin, M.; Uggetti, A.; Giaccone, G.; Sperber, S.; Beeg, M.; Salmona, M.; et al. New mutations in MAPT gene causing frontotemporal lobar degeneration: Biochemical and structural characterization. Neurobiol. Aging 2012, 33, 834. [Google Scholar] [CrossRef]
  195. Chimenti, L.; Luque, T.; Bonsignore, M.R.; Ramirez, J.; Navajas, D.; Farre, R. Pre-treatment with mesenchymal stem cells reduces ventilator-induced lung injury. Eur. Respir. J. 2012, 40, 939–948. [Google Scholar] [CrossRef] [Green Version]
  196. Chen, W.; Huang, J.; Hu, Y.; Khoshnam, S.E.; Sarkaki, A. Mitochondrial Transfer as a Therapeutic Strategy Against Ischemic Stroke. Transl. Stroke Res. 2020, 11, 1214–1228. [Google Scholar] [CrossRef]
  197. Russo, E.; Napoli, E.; Borlongan, C.V. Healthy mitochondria for stroke cells. Brain Circ. 2018, 4, 95–98. [Google Scholar] [CrossRef]
  198. Lu, M.; Guo, J.; Wu, B.; Zhou, Y.; Wu, M.; Farzaneh, M.; Khoshnam, S.E. Mesenchymal Stem Cell-Mediated Mitochondrial Transfer: A Therapeutic Approach for Ischemic Stroke. Transl. Stroke Res. 2021, 12, 212–229. [Google Scholar] [CrossRef]
  199. Lippert, T.; Borlongan, C.V. Prophylactic treatment of hyperbaric oxygen treatment mitigates inflammatory response via mitochondria transfer. CNS Neurosci. 2019, 25, 815–823. [Google Scholar] [CrossRef]
  200. Cheng, L.; Marco, K.H.C.; Shuo, H.; Zhao, Z.; Ling, C.; Junhui, C.; Hui, X.; Jianxiang, Q. Mesenchymal stem cells and their mitochondrial transfer: A double-edged sword. Biosci. Rep. 2019, 39, BSR20182417. [Google Scholar] [CrossRef] [Green Version]
  201. Crewe, C.; Funcke, J.B.; Li, S.; Joffin, N.; Gliniak, C.M.; Ghaben, A.L.; An, Y.A.; Sadek, H.A.; Gordillo, R.; Akgul, Y.; et al. Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. Cell Metab. 2021, 33, 1853–1868. [Google Scholar] [CrossRef]
  202. Liu, D.; Gao, Y.; Liu, Y.; Huang, J.; Yin, J.; Feng, Y.; Shi, L.; Meloni, B.P.; Zhang, C.; Zheng, M.; et al. Intercellular mitochondrial transfer as a means of tissue revitalization. Sig. Transduct. Target Ther. 2021, 6, 65. [Google Scholar] [CrossRef]
Figure 1. Gap junction model. Each channel comprises two hemichannels that form a hydrophilic pathway spanning two apposed plasma membranes and a narrow extracellular space (gap). Each hemichannel is formed by the radial interaction of six proteins (connexins/innexins).
Figure 1. Gap junction model. Each channel comprises two hemichannels that form a hydrophilic pathway spanning two apposed plasma membranes and a narrow extracellular space (gap). Each hemichannel is formed by the radial interaction of six proteins (connexins/innexins).
Ijms 23 06133 g001
Figure 2. Model of connexin 32 (Cx32) topology. The connexin spans the bilayer four times (M1–M4) and has both NH2- and COOH-termini at the cytoplasmic side of the membrane, forming two extracellular loops (E1, E2) and one cytoplasmic loop (CL).
Figure 2. Model of connexin 32 (Cx32) topology. The connexin spans the bilayer four times (M1–M4) and has both NH2- and COOH-termini at the cytoplasmic side of the membrane, forming two extracellular loops (E1, E2) and one cytoplasmic loop (CL).
Ijms 23 06133 g002
Figure 3. Cork-gating model. Both the positively charged channel’s mouth and the negatively charged CaM lobes are ~35 Å in diameter (A). Therefore, a CaM lobe could fit well in the channel’s mouth (vestibule) and electrostatically interact with it. In (B), the channel is split along its length to display the pore diameter (light blue area) along the channel’s length. Both CaM and connexon images (B) were provided by Dr. Francesco Zonta (Venetian Institute of Molecular Medicine, VIMM, University of Padua, Italy).
Figure 3. Cork-gating model. Both the positively charged channel’s mouth and the negatively charged CaM lobes are ~35 Å in diameter (A). Therefore, a CaM lobe could fit well in the channel’s mouth (vestibule) and electrostatically interact with it. In (B), the channel is split along its length to display the pore diameter (light blue area) along the channel’s length. Both CaM and connexon images (B) were provided by Dr. Francesco Zonta (Venetian Institute of Molecular Medicine, VIMM, University of Padua, Italy).
Ijms 23 06133 g003
Figure 4. Thin section of an axon of a crayfish abdominal ganglion. Axonal and glial cell plasma membranes fuse, forming two pores (P). Inset “a” shows a fluorescence micrograph of a median giant axon intracellularly injected with Lucifer Yellow CH; note that the nuclei of the adaxonal (sheath) glial cells are stained by the fluorescent dye (Inset “a,” red arrows). Inset “b” is an enlargement of the pore region. A, axon; G, glial cell; C, Adjacent glial connective tissue sheath, non-cellular. The fluorescence micrograph of inset “a” (provided by Dr. Terry A. Viancour) represents a crayfish median giant axon. The electron micrograph is an original, unpublished image taken by one of the authors (C. Peracchia).
Figure 4. Thin section of an axon of a crayfish abdominal ganglion. Axonal and glial cell plasma membranes fuse, forming two pores (P). Inset “a” shows a fluorescence micrograph of a median giant axon intracellularly injected with Lucifer Yellow CH; note that the nuclei of the adaxonal (sheath) glial cells are stained by the fluorescent dye (Inset “a,” red arrows). Inset “b” is an enlargement of the pore region. A, axon; G, glial cell; C, Adjacent glial connective tissue sheath, non-cellular. The fluorescence micrograph of inset “a” (provided by Dr. Terry A. Viancour) represents a crayfish median giant axon. The electron micrograph is an original, unpublished image taken by one of the authors (C. Peracchia).
Ijms 23 06133 g004
Figure 5. Thin section of an axon of a crayfish abdominal ganglion. The arrows point to a large pore area. Note the accumulation of cytoplasmic organelles near the pore region. A, axon; G, glial satellite cell; C, Adjacent Glial connective tissue sheath, non-cellular; M, mitochondrion; TL, tubular lattice (Peracchia and Robertson, J. Cell Biol. 51, 223–239, 1971). The electron micrograph is an original, unpublished image taken by one of the authors (C. Peracchia).
Figure 5. Thin section of an axon of a crayfish abdominal ganglion. The arrows point to a large pore area. Note the accumulation of cytoplasmic organelles near the pore region. A, axon; G, glial satellite cell; C, Adjacent Glial connective tissue sheath, non-cellular; M, mitochondrion; TL, tubular lattice (Peracchia and Robertson, J. Cell Biol. 51, 223–239, 1971). The electron micrograph is an original, unpublished image taken by one of the authors (C. Peracchia).
Ijms 23 06133 g005
Figure 6. Thin section of an axon of a crayfish abdominal ganglion. The arrows point to a large pore area. A, axon; G, glial cell; C, Adjacent Glial connective tissue sheath, non-cellular; M, mitochondrion. The electron micrograph is an original unpublish image taken by one of the authors (C. Peracchia).
Figure 6. Thin section of an axon of a crayfish abdominal ganglion. The arrows point to a large pore area. A, axon; G, glial cell; C, Adjacent Glial connective tissue sheath, non-cellular; M, mitochondrion. The electron micrograph is an original unpublish image taken by one of the authors (C. Peracchia).
Ijms 23 06133 g006
Figure 7. Nature’s front page shows one of our pore images.
Figure 7. Nature’s front page shows one of our pore images.
Ijms 23 06133 g007
Figure 8. Morphological characteristic of TNT in human macrophages. Representative image after 3 days post HIV infection analyzed by SEM. (A) Representative image of HIV-infected cultures after 3 days post-infection. (B) High magnification of the end of the TNT process in HIV-infected conditions. Arrows denote TNT. n  =  5.
Figure 8. Morphological characteristic of TNT in human macrophages. Representative image after 3 days post HIV infection analyzed by SEM. (A) Representative image of HIV-infected cultures after 3 days post-infection. (B) High magnification of the end of the TNT process in HIV-infected conditions. Arrows denote TNT. n  =  5.
Ijms 23 06133 g008
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Eugenin, E.; Camporesi, E.; Peracchia, C. Direct Cell-Cell Communication via Membrane Pores, Gap Junction Channels, and Tunneling Nanotubes: Medical Relevance of Mitochondrial Exchange. Int. J. Mol. Sci. 2022, 23, 6133. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23116133

AMA Style

Eugenin E, Camporesi E, Peracchia C. Direct Cell-Cell Communication via Membrane Pores, Gap Junction Channels, and Tunneling Nanotubes: Medical Relevance of Mitochondrial Exchange. International Journal of Molecular Sciences. 2022; 23(11):6133. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23116133

Chicago/Turabian Style

Eugenin, Eliseo, Enrico Camporesi, and Camillo Peracchia. 2022. "Direct Cell-Cell Communication via Membrane Pores, Gap Junction Channels, and Tunneling Nanotubes: Medical Relevance of Mitochondrial Exchange" International Journal of Molecular Sciences 23, no. 11: 6133. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23116133

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop