Next Article in Journal
Comparing Protection of Remote Limb with Resveratrol Preconditioning following Rodent Subarachnoid Hemorrhage
Next Article in Special Issue
Nuclear and Cytoplasmatic Players in Mitochondria-Related CNS Disorders: Chromatin Modifications and Subcellular Trafficking
Previous Article in Journal
Deciphering Cardiac Biology and Disease by Single-Cell Transcriptomic Profiling
Previous Article in Special Issue
Molecular Mechanisms behind Inherited Neurodegeneration of the Optic Nerve
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Interplay of Microtubules with Mitochondria–ER Contact Sites (MERCs) in Glioblastoma

by
Francesca Grespi
1,†,
Caterina Vianello
1,†,
Stefano Cagnin
1,2,3,
Marta Giacomello
1,4,* and
Agnese De Mario
4,*
1
Department of Biology, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
2
CRIBI Biotechnology Center, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
3
CIR-Myo Myology Center, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
4
Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58b, 35100 Padua, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2022, 12(4), 567; https://doi.org/10.3390/biom12040567
Submission received: 24 February 2022 / Revised: 1 April 2022 / Accepted: 6 April 2022 / Published: 12 April 2022
(This article belongs to the Special Issue Mitochondria and Central Nervous System Disorders)

Abstract

:
Gliomas are heterogeneous neoplasms, classified into grade I to IV according to their malignancy and the presence of specific histological/molecular hallmarks. The higher grade of glioma is known as glioblastoma (GB). Although progress has been made in surgical and radiation treatments, its clinical outcome is still unfavorable. The invasive properties of GB cells and glioma aggressiveness are linked to the reshaping of the cytoskeleton. Recent works suggest that the different susceptibility of GB cells to antitumor immune response is also associated with the extent and function of mitochondria–ER contact sites (MERCs). The presence of MERCs alterations could also explain the mitochondrial defects observed in GB models, including abnormalities of energy metabolism and disruption of apoptotic and calcium signaling. Based on this evidence, the question arises as to whether a MERCs–cytoskeleton crosstalk exists, and whether GB progression is linked to an altered cytoskeleton–MERCs interaction. To address this possibility, in this review we performed a meta-analysis to compare grade I and grade IV GB patients. From this preliminary analysis, we found that GB samples (grade IV) are characterized by altered expression of cytoskeletal and MERCs related genes. Among them, the cytoskeleton-associated protein 4 (CKAP4 or CLIMP-63) appears particularly interesting as it encodes a MERCs protein controlling the ER anchoring to microtubules (MTs). Although further in-depth analyses remain necessary, this perspective review may provide new hints to better understand GB molecular etiopathogenesis, by suggesting that cytoskeletal and MERCs alterations cooperate to exacerbate the cellular phenotype of high-grade GB and that MERCs players can be exploited as novel biomarkers/targets to enhance the current therapy for GB.

1. Introduction

Glioblastoma (GB), also known as glioblastoma multiforme, is the most aggressive type of astrocytoma. GB derives both from glial and glioma stem cells and affects glia and astrocytes [1]. Although the current therapeutic strategy accounts for complementary approaches, as surgical resection, radiotherapy, chemotherapy and antiangiogenesis agents, the prognosis of GB patients is still poor [2].
Based on well-defined genome-wide gene expression changes, somatic mutations and copy number changes, GB is commonly divided into four subtypes: classical, proneural, mesenchymal, and neural [3,4]. The classical subtype (97% of tumors) is characterized by an altered expression of the epidermal growth factor receptor (EGFR) while the proneural subtype by alterations in the genes coding for the tumor protein p53 (TP53), platelet-derived growth factor receptor (PDGFRA), isocitrate dehydrogenase-1 (IDH1) [5]. The mesenchymal subtype is due mostly to mutations in the neurofibromin 1 (NF1) gene, while a low percentage of the cases are linked to EGFR gene changes. The mesenchymal subtype is also enriched in the expression of astrocytes and microglia markers. The neural subtype is enriched in astrocytic and oligodendrocytic markers such as Oligodendrocyte transcription factor 1 (OLIG1), 3-O-methyl-[3H]-glucose (OMG), proteolipid protein 1 (PLP1), Tenascin-R (TNR), Glial fibrillary acidic protein (GFAP), Excitatory amino acid transporter 1 (SLC1A3), astrocyte-specific glutamate transporter (GLAST), Megalencephalic leukoencephalopathy with subcortical cysts protein-1 (MLC1), SRY-Box Transcription Factor (SOX) 4, SOX11, and Doublecortin (DCX) [6,7].
Most of the GB cases are idiopathic: only a minority of patients (~5%) harbor critical germline alterations, while 20% have a strong family history of cancer [8]. GBs arise mainly in the supratentorial cerebral hemispheres, with variable clinical presentations. GB symptoms include persistent weakness, numbness, loss of vision or alteration of language; those located in the frontal lobe, temporal lobe, or corpus callosum are associated with, executive dysfunctions, mood disorders, fatigue and mild memory disorders [9].
Gliomas are classified by the WHO (World Health Organization Histological Classification of Tumors) into different grades based on the severity of the disease—grade IV being the most aggressive—with a mean survival of 15 months from the diagnosis [10]. The effectiveness of therapeutic treatments is hampered by many factors like heterogeneous molecular and genetic backgrounds of the different tumors and of the various cells within the same tumor [2,10,11]. Despite many studies aimed to investigate the molecular mechanisms underlying GB, its cellular origin remains enigmatic. It has been hypothesized that GB raises from neuroectodermal stem cells: endowed of high proliferative potential, the latter display strong migratory abilities and can undergo differentiation into GB cells [12]. However, the cells that populate GB are not homogeneous: the tumor mass contains endothelial cells, stem cells, astrocytes, progenitor cells and immune cells.
During GB progression the blood brain barrier (BBB) is compromised, allowing entry of immune cells and neuroinflammation due to chemoattraction and activation of glial cells. Microglial cells produce high levels of proinflammatory molecules, such as nitric oxide (NO) and tumor necrosis factor alpha (TNF-α), further enhancing BBB breakdown by disruption of the astrocyte–BBB junctions (as comprehensively described in [13,14,15]. The heterogeneous nature of GB cells also underlies their distinct susceptibility to cytotoxic T lymphocytes, which are crucial for immune defense. Accordingly, Bassoy and co-authors found that glioma stem cells were more readily attacked and killed in vitro and in vivo by cytotoxic T lymphocytes than their differentiated counterpart [16]. Here the authors show that the interaction among the Endoplasmic Reticulum (ER) and mitochondria drives the expression of surface glycans in glioma stem-like cells and thereby dictates their response to killer lymphocytes [17].
GB rarely metastasize outside the central nervous system (CNS). However, it can be highly invasive within the brain parenchyma, a feature that severely limits the efficacy of surgery and radiotherapy. Invasion of GB cells into the brain reflects the dynamic interplay between cell-to-cell adhesion, remodeling of the extracellular matrix and cell motility. Specifically, during epithelial-mesenchymal transition (EMT), cancer cells enhance migratory and invasive capabilities through the reorganization of the cytoskeleton and the formation of membrane protrusions [18]. Treatments targeting Rho GTPases, that modulate cell migration through downstream rearrangement of the actin cytoskeleton, have been proposed as a novel approach for GB treatment [19,20].
Molecularly, GB have been associated with alterations in cell cycle regulatory genes such as p16, cyclin-dependent kinase (cdk) 4, cdk6, cyclin D1 and retinoblastoma protein, as well as of molecules belonging to tumor activation pathways like receptor tyrosine kinases responsible for activation of the Phosphoinositide 3-kinases (PI3K), Protein kinase B, (Akt), mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) axes [21]. As in other solid tumors, dysfunction in mitochondria physiology has been reported and is considered a key driver of GB progression. Abnormalities of the cell bioenergetics, changes in mitochondria membrane potential regulation, disruption of apoptotic signaling pathways, mitochondrial swelling with partial or total breakdown of mitochondrial cristae (cristolysis) and deranged fusion and fission processes have been observed [22,23]. These ultrastructural changes are consistent with the alterations of mitochondrial bioenergetics, pointing to a critical derangement of oxidative phosphorylation (OXPHOS) in GB cells [23].
These pathways are controlled both by the interaction among ER and mitochondria at the mitochondria–Endoplasmic Reticulum contact sites (MERCs [24]) and by cytoskeletal remodeling, thus suggesting that these two factors cooperate towards the development of GB. In this review we discuss this possibility, that is: the role of microtubule rearrangement and its interplay with MERCs in the progression of GB.

2. Cytoskeleton and Glial Cells: A Focus on Microtubule and Microfilaments

The cytoskeleton is formed by three different types of dynamic filaments that determine the cell structural organization: microtubules (heterodimers of α- and β-tubulins), intermediate filaments (neurofilaments or glial filaments) and microfilaments (two strands of actin subunits). In this review we will focus mainly on microtubules (MTs). Accounting for 20% of total protein in the brain, microtubules are involved in important cell functions: mitotic spindle formation, ensuring proper chromosome segregation and cell division, maintenance of the cellular architecture and structure, intracellular organelles transport and signal propagation. MTs interact with diverse organelles, including the endoplasmic reticulum (ER), Golgi apparatus, lysosomes and mitochondria [25,26,27]. MTs assemble from dimers of α- and β-tubulin: the latter align head-to-tail to form protofilaments, which in turn associate laterally and form tubes. The extremity where α-tubulin is exposed (termed the minus end) grows slowly in vitro, whereas the opposite end (termed the plus end) grows rapidly [28]. Assembly (polymerization) and disassembly (depolymerization) of microtubules is driven by hydrolysis of guanosine triphosphate (GTP) on the β-tubulin monomer. Polymerization is typically initiated from a pool of GTP-loaded tubulin subunits [29]. GTP hydrolysis occurs shortly after incorporation of tubulin subunits and induces a conformational change of protofilaments: from a slightly curved tubulin-GTP bound rod to a more profoundly curved tubulin-guanosine diphosphate (GDP) structure [29]. Growing microtubule sheets maintain a “cap” of tubulin-GTP subunits whose loss results in rapid depolymerization. The microtubule network is controlled by many proteins, such as microtubule associated proteins (MAP), the stabilizing proteins Microtubule-associated protein RP/EB family member 1 and Cytoplasmic linker protein CLIP-170, as well as by microtubule-destabilizing or depolymerizing proteins, like spastin and katanin, the depolymerizing motor protein kinesin and the αβ-tubulin dimer-binding protein stathmin [30]. MTs interact also with regulators of the cell cycle and apoptosis, including the tumor suppressor protein p53, Bcl-2 and survivin and with the intracellular transport proteins kinesins and dyneins [31]. Dyneins transport various cell cargo by crawling along cytoskeletal MTs towards their minus-end. Kinesin family members (KIF) are instead a group of proteins playing a role in cytoskeleton organization: KIF move along microtubule filaments after hydrolysis of ATP and are responsible for their assembly into parallel arrays [32]. Post-translational modifications (tyrosination, glutamylation, and acetylation) also control their polymerization, the functions of the microtubule network (e.g., chromosome segregation, spindle formation, transport of organelles, including mitochondria [33,34] and the affinity for Microtubule-Associated Proteins (MAPs) [35]. MAPs are key regulator of MTs dynamics, as they can (de) stabilize protofilaments, guide their growth towards specific cellular domains, mediate their interaction with other proteins. Tubulin is also subjected to palmytoilation (that is, protein fatty acid acylation) [36]. This covalent attachment of lipids allows anchoring of organelles membranes to the tubulin filaments and their positioning along the MTs.
The organization of microtubules differs among cell types: in neurons, the MTs network determines cell polarity to support the growth and structure of axon and dendrites [37]. The glial environment constantly influences the axonal cytoskeleton, providing an example of the dynamic nature of the neuronal cytoskeleton and of the interplay among different cell types in the brain [38]. Indeed, radial cells contribute to the overall structural and functional organization of the central nervous system via microtubule dependent processes. This is demonstrated by the fact that lissencephaly (a clinical condition in which deranged cortical development affects the surface of the brain, due to the failure of neuronal migration) is associated with altered biogenesis of radial glia microtubules [39]. The microtubule cytoskeleton controls the morphology and polarization of the glial cells. It also provides a dynamic platform for the development and extension of membrane processes at the basis of inter-cellular communication and migration. While on one hand these evidences point to the central role of MTs for glial cells function, they also support the possibility that altered microtubule dynamics contribute to the development of human disorders characterized by altered cell motility [40]. It is important to briefly mention the role of actin filaments in regulating brain cells physiology. In general, actin filaments are crucial for various processes such as proliferation, migration, cell morphogenesis and apoptosis (for a more detailed and comprehensive overview of actin dynamics and function we refer the reader to [41,42,43,44]. In the case of microglia, actin plays a fundamental role by shaping its physiology and motility [41,45,46,47] -actin monomers assemble into actin filaments (F-actin) that form lamellipodia and filopodia which are responsible of cell movement.
Cytoskeleton dynamics are also shaped by other intermediate filaments proteins, which are expressed in a tissue-specific manner, in contrast to actin microfilaments and microtubules. Among these, vimentin is particularly important, being fundamentally expressed in radial glia and immature astrocytes during development. Vimentin filaments are characterized by higher elasticity than actin and microtubules, a property which is typical also of desmin, the major intermediate filament protein of muscle [48]. Vimentin importance is highlighted by the fact that vimentin intermediate filaments and actin cooperate to redefine the cell cortex. Further, Vimentin is a key component of migrasomes, recently discovered vesicles located within retraction fibers that control the cell migration speed [49]. Migration does not depend on the function of a single protein class. Rather, it depends on the interplay among different types of intermediate filaments, as in the case of epithelial cells whose migration requires the interaction between vimentin and keratin intermediate filaments [50]. Finally, intermediate filaments proteins include lamins, which play an important role in maintaining the shape of the nuclei of all cells, as well as in the organization of the cytoskeleton, mechanotransduction, and cell motility [51].

3. Cytoskeletal Proteins: Key Players of Glioblastoma Cell Invasive Properties

Recent reports indicate a role for MTs in GB progression and invasion. As migration and infiltration of GB cells is governed by reshaping of the cytoskeleton, it is not surprising that the composition and organization of the cytoskeleton in GB cells differs from that of healthy brain cells. Besides the altered expression of tubulin isoforms [52,53], many findings link GB to altered microtubule dynamics, especially those mediated by microtubule associated proteins (MAPs). Microarray analyses and immunohistochemical studies have found that GB tissues express an astrocytoma-specific splice variant (MAP2e) of the neuronal MAP2, particularly in the cells at the invasive front of the tumor [54]. Another MAP overexpressed in GB cells is the receptor for hyaluronic acid mediated motility (RHAMM)/intracellular hyaluronic acid binding protein (IHABP) that exhibits several splice variants, some of which are specific to GB [55].
Invading Cells are also enriched in dynein, in dynein-associated and actin-associated proteins, substantially contributing to the subcellular processes underlying their proliferative and migration abilities [56]. Notably, advillin (AVIL), a member of the villin/gelsolin family that regulates actin filament reorganization, also contributes to GB tumorigenesis [57]. Xie and colleagues found that AVIL is overexpressed in GB cells (including GB stem cells), and that its overexpression promotes cell movement, GB proliferation and migration. The latter are fostered in GB also by overexpression of formin (FMN1), that controls polymerization of actin cables, e.g., linear cytoskeletal structures that assemble at nascent adherens junctions and drive intercellular adhesion [58,59].
Another cytoskeletal protein linked to GB is Nestin, a class IV intermediate filament protein that is considered as a putative marker of neural progenitor stem cells in the central nervous system. Not only Nestin expression is significantly overexpressed in GB, compared with other types of glioma, but its deletion in tumor cells from GB patients derived xenografts caused cell cycle blockage, finally bringing to tumor cells death. This effect is due to the physical association of Nestin with β II-tubulin: indeed, deletion of either Nestin or βII-tubulin disrupted spindle morphology in GB cells [60].
Higher expression levels of lamin A/C have been also linked with a reduced overall survival of patients affected by glioblastoma multiforme, as well as with the enrichment of cancer-related pathways related to cell migration and adhesion [61]. Recent analyses also highlight that some keratin forms (KRT33B, KRT75) can be used as prognostic biomarkers for early detection of low-grade gliomas, and others (KRT75) can be used to reveal transformation tumors with higher probability to switch from low-grade gliomas to GB [62].
Overall, these findings suggest the existence of a relationship between the invasive properties of GB cells and the derangement of cytoskeleton network and functions.

4. Mitochondria Dynamics and Glial Cells

Proper MTs organization is essential also for the movement of mitochondria, whose dysfunctions have been also associated with the infiltrative and proliferative capacity of glioma cells [18,63]. Anterograde and retrograde transport of mitochondria along MTs are mediated by plus end-directed kinesin-1/KIF5 and minus-end-directed dynein motor proteins respectively [64]. The interaction between mitochondria and kinesin/dynein depends on the formation of a complex between the outer mitochondria membrane (OMM), the mitochondrial rho GTPase Miro 1 (Miro1) and TRAK1/2 (also known as Milton) [65]. Miro-1 binds the cytoplasmic adaptor protein Milton and the kinesin heavy chain through its cytoplasmic domain, thereby connecting mitochondria to microtubules [66]. The movement of the organelles is tightly coordinated with changes in their morphology [26]. Their shape and assembly into a network result from a net balance of fusion–fission processes of the mitochondria membranes. These processes are regulated by a family of “mitochondria-shaping proteins”: e.g., in mammals, fission is controlled by the concerted action of the cytosolic Dynamin-1-like protein (DRP-1) [67] and of proteins that recruit Drp1 at the surface of mitochondria like human Mitochondrial fission 1 protein (hFis1), Mitochondrial elongation factor 2 (Mid49), Mitochondrial fission factors (MFFs). Among the protein that control fusion, the most studied are Optic Atrophy 1 (Opa1) and Mitofusins (Mfn; for a comprehensive view of mitochondrial dynamics, see [68]). As mentioned before, several defects in mitochondrial morphology, positioning and functionality have been described in brain cancer cells [69] and in other cancer cells [70].
The subcellular distribution of mitochondria has been correlated with the migration and invasion capabilities of GB cells [71]. It has been shown that PI3K antagonists trigger the transport of energetically active, elongated mitochondria to the cortical cytoskeleton of tumor cells. The repositioned mitochondria apparently speed up lamellipodia dynamics, induce faster turnover of focal adhesion complexes, increase speed and width of cell migration, ultimately leading to higher rates of GB cell invasion [52,72,73]. Mitochondria support GB invasiveness not only though ATP supply, but also by controlling the expression of surface glycans in glioma stem-like cells [16]. Indeed, although the exact mechanism has not been completely clarified, Bassoy and colleagues showed that ER–mitochondria interactions, rather than mitochondria per se, determine GSC sialylated glycan surface expression. The crosstalk of mitochondria and ER fosters the expression of the sialylated glycans, that are subsequently exposed to the cell surface and contribute to cell adhesion [74].
Mitochondria–ER contact sites (MERCs), the sites of contacts among ER and mitochondria, participate to different cell functions and pathologies [24,75,76]. The environment generated by the MERCs represents a privileged site for non-vesicular lipid trafficking between these organelles and facilitates transport of lipids via carrier proteins [77]. Several key enzymes, located in MAMs, mediate phospholipid synthesis, such as phosphatidylserine synthase-1/2 (PSS1/2) and phosphatidylethanolamine N-methyltransferase 2 (PEMT2), [78], while oxysterol-binding protein (OSBP)-related protein 5 and 8 (ORP5/ORP8) have been recently reported to locate at MERCs where they mediate the transport of phospholipids [79]. In addition, key regulators of triacylglycerol synthesis and steroidogenesis, such as acyl-CoA/diacylglycerol acyltransferase 2 (DGAT2) [80], the steroidogenic acute regulatory protein (StAR) [81], the long-chain-fatty-acid-CoA ligase 4 (FACL4) [82] are enriched at MERCs. The cholesterol acyltransferase-1 (ACAT1/SOAT1), an enzyme that catalyzes the generation of cholesterol esters, is also enriched in MERCs. The contribution of MERCs to lipid homeostasis appears fundamental also for the etiology of cancer: the amount and proportion of the various types of lipids available within cells dictate their membrane composition, fluidity and stiffness, ultimately controlling their adhesion ability. Although a detailed overview is beyond the scope of this review (please refer to [24,76] for a more comprehensive description), we want to highlight here another MERCs function that is a decision-making node in the balance between cell survival and death: its contribution to the regulation of Ca2+ homeostasis [83,84,85]. Thanks to the tight tether between the ER membranes and mitochondria, Ca2+ can be rapidly transferred through the formation of MERCs-resident microdomains that overcome the low Ca2+ affinity of the mitochondrial Ca2+ uniporter (MCU) [83,86]. The IP3R is physically linked to the OMM voltage-dependent anion channel 1 (VDAC1) by means of the molecular chaperone glucose-regulated protein 75 (Grp75) [87]. Moreover, the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) pump localized to the ER membrane is regulated by several proteins residing at MERCs, thereby modulating Ca2+ kinetics [88].
As reported above, mammalian cells proteins involved in the anterograde and retrograde movement of organelles are controlled by local Ca2+ rises. For example, the binding of Ca2+ to the EF hands domains of the OMM Rho-GTPase protein Miro1 drives its conformational change and decoupling from microtubule motors (specifically, kinesin and dynein), thereby controlling mitochondrial motility [89]. Not only do cytosolic Ca2+ levels control mitochondrial transport, but they also control changes in the concentration of mitochondrial matrix Ca2+: of note, it has been demonstrated that Miro1 regulates intramitochondrial Ca2+ levels. Although Miro1 has been shown to localize at MERCs, where it contributes to lipids exchange and biosynthesis [90,91], and although it has been reported to interact/be regulated with/by MERCs-associated proteins (e.g., Mitofusin, Parkin, Pink1 [92,93]), how it functions within MERCs relates with its extra-MERCs role, which has not been completely clarified.
Finally, while regulated Ca2+ transport at MAMs favors bioenergetics, it is well established that massive and prolonged mitochondrial Ca2+ load promotes increased reactive oxygen species production and may trigger the opening of the mitochondrial permeability transition pore, eventually leading to apoptosis [94]. As a result of the consequent OMM permeabilization, caspase-activating and proapoptotic factors, including cytochrome c, are released into the cytoplasm, exacerbating Ca2+ release from the IP3R and avoiding the Ca2+-dependent inhibition of the receptor itself [95].
All these findings suggest a specialized role of MERCs in glial cells by influencing the mitochondria and Ca2+-dependent processes that support their normal physiology.

5. MERCs in Glioblastoma

Considering the role of MERCs in the maintenance of cell homeostasis, it is not surprising that some experimental reports highlighted their contribution to the etiology of different types of tumors, including GB.
A first link between MAMs and cancer has been shown in case of the protein promyelocytic leukemia (PML), a nuclear tumor suppressor protein, which has been located at MERCs. Here, PML modulates the IP3R-Akt axis trough the regulation of Ca2+ transfer from the ER to the mitochondria and ultimately induces pro-apoptotic signaling [96]. Notably, the Akt pathway has been also connected with the increased tumorigenicity, stem cell-ness, and invasiveness of GB cells [97]. This could strengthen the MERCs-dependent effect on glycan expression that has been associated with enhanced migration abilities and diverse susceptibility to cytotoxic lymphocyte-mediated killing of GB cells [16]. Interestingly, studies on MERCs ultrastructure in surgical specimens obtained from human astrocytic neoplasms revealed a striking difference among well-differentiated glioma cells and poorly differentiated glioma like-stem cells. The latter appeared almost deprived of MERCs [98]. Although this finding is interesting, it remains unclear whether this is a typical feature of stem cells and of stem cell-like cells, which could be MERCs contribution to glioma stem cells physiology. Notably, glioma stem cells appear less glycolytic than their differentiated counterparts and rely mainly on oxidative phosphorylation [99]. At variance, differentiated GB cells were reported as “MERCs-enriched”, with MERCs driving mitochondria functionality and sustaining their metabolic demands [100].
Another protein important in the context of tumors is the cytoskeleton-binding protein trichoplein/mitostatin (TpM), which binds the major intermediate filaments of epithelial cells. TpM acts as a negative modulator of ER–mitochondria juxtaposition, inhibiting apoptosis induced by Ca2+-dependent stimuli through interaction with MFN2 [94]. Interestingly, TpM is involved in the formation of the primary cilium, a dynamic protrusion present in virtually every type of cell that senses extracellular signals and coordinates the subcellular responses with the progression of cell cycle, proliferation, and differentiation. This is particularly interesting in the context of GB, since aberrant ciliogenesis has been proposed as an early event of GB etiology [101,102]. The potential contribution of TpM to carcinogenesis is corroborated by findings that regards other types of solid tumors. As an example, it has been demonstrated that TpM inhibits cell migration, invasion, and tumorigenicity of prostate cancer cells, and that it is down-regulated in advanced stages of human prostate cancers [95]. All these data suggest that TpM can act as a tumor suppressor: however, it has not been clarified whether these findings are due to altered TpM function at MERCs or to variations of its extra-MERCs physiological roles.

6. MERCs and Microtubules Crosstalk in Glioblastoma

In the previous paragraphs, we summarized the published findings on the potential contribution of MERCs and cytoskeletal dynamics to GB etiology. Considering the role of the latter in the maintenance of the cell bioenergetics, signaling, and migration abilities, the question rises as to whether and how MERCs and MTs contribute and coordinate their activities during GB growth and diffusion into different brain regions. The finding that the interaction between tubulin and the MERCs–resident protein VDAC1 actively controls the cell metabolism in muscle and in cell models of neuroblastoma and breast cancer hints at this possibility [103]. Data from different cell models support the idea that tubulin can control mitochondrial respiration by diminishing the VDAC-mediated permeability of the OMM to ATP, ADP, and other metabolic substrates [104,105]. It is tempting to speculate that this could be a mechanism to shape mitochondrial respiration and support the metabolic changes that characterize GB [106]. Interestingly, VDAC1 binding to tubulin dimers and the consequent blockage of its activity is controlled by phosphorylation either by glycogen synthase kinase-3β or by cyclic AMP-dependent protein kinase (PKA), whose expression differs in glioma cells with respect to normal tissue [107,108,109]. Theoretically, altered levels of these kinases could influence the levels of phosphorylated VDAC1 and in turn influence its tubulin binding, a possibility that has not yet been demonstrated in the context of GB.
In addition, several recent studies have utilized high-throughput genomic, epigenomic, and transcriptomic approaches for detailed molecular characterization of GB. A recent proteomic study found that cells derived from GB patients upregulate the expression not only of cytoskeletal (vimentin, α- and β-tubulin, β-actin, and glial fibrillary acidic protein), but also of mitochondrial (aldehyde dehydrogenase and manganese superoxide dismutase) and MERCs/ER-resident chaperone proteins (GRP-75, GRP-57, and HSP27) [109].
To provide details on the crosstalk among MERCs and the cytoskeleton in glial cells tumors, we took advantage of published genome-wide gene expression analyses to evaluate alterations occurring during GB progression. We identified differentially expressed genes (DEGs) in GB considering two GEO datasets that are unique because they were obtained upon analysis of human GB biopsies (GSE147352 and GSE79878) [110]. These datasets include 93 glioblastoma, 18 glioma, and 15 normal samples studied though two different techniques: microarray and RNA sequencing.
DESeq normalized read counts were used to identify differentially expressed genes using the AltAnalyze software [111]. Genes were considered as differentially expressed whenever their fold change would be > 2 and p-value < 0.05.
In case of microarray gene expression analysis, we excluded lincRNAs from the analysis before normalizing data because they are lower expressed than coding RNAs, as we did in [112]. Raw data with gene expression information for coding genes were quantile normalized using R, and the expression of each gene was calculated as the average of multiple probes for the same gene with an intensity fluorescence significantly above the background (using the filter gIsPosAndSignif in the Agilent microarray quantification files [113]). DEGs were identified using TMev_4.8.1 software (Quantitative Biomedical Research Center, Harvard T.H. Chan School of Public Health, Boston, USA) [114] (www.mev.tm4.org, accessed on 5 April 2022). Genes were considered altered when fold change was > 1.5 and p-value < 0.05. DEGs (Supplementary Table S1) were categorized according to Gene Ontology (GO) using the Panther 17.0 database [115] (www.pantherdb.org, accessed on 17 January 2022) (Supplementary Table S2) and GO terms were reduced using the REVIGO tool with default parameters [116]. Downregulated genes in GB were involved in neuronal functions as the formation of neuron projection or in the excitatory of GABAergic signal (Supplementary Figure S1 and Table S2). The list of upregulated genes in GB was also enriched of molecules controlling cytoskeletal and mitochondrial functions (Figure 1). By analyzing RNAseq data (GSE147352) (microarray analyses are based on the comparison between gliomas grade I and GB grade IV), we highlighted that genes associated with ER are also expressed (Figure 1A).
We then evaluated alterations in genes involved in the synthesis of MERCs proteins (Supplementary Table S3). Using normal samples as control, we found that 37% (62 of 169) of genes coding for MERCs proteins were altered in GB (Supplementary Table S3), and 22% (38 of 169) were altered in glioma tumors (Supplementary Table S3 and Figure 2). Interestingly, most of the genes coding for proteins controlling MERCs formation were also differentially expressed in glioblastoma samples. Genes most downregulated in glioblastoma are also highly expressed in glioma (JPH1, SNCA, ITPR1, and FRAS1). On the other hand, those highly expressed in glioblastoma (CAV1, TP53, TMX1, MTTP, RAB32, ITGB1, PEMT, SOAT1, BAX, PKMYT1) are not altered in glioma (Figure 2 and Supplementary Table S3). Genes involved in MAMs formation whose expression was altered only in glioma samples were FUNDC1, FACL4, VDAC1, ATP6AP2, CISD2, ERO1A, TP53I11, MAVS, and CDK5RAP3, while those altered only in GB and not in gliomas were TDRKH, SOAT1, LMAN1, TRIM4, CAV1, BAX, ITGB1, SEC61B, PDIA6, CFLAR, RAB32, MTTP, TMPO, ELOVL1, PKMYT1, p66shc/SHC1, PEMT, LRRC59, BET1, TBL2, ALG9, TOMM5, SERAC1, TCHP, VMA21, RPS7, PTRH2, ATG5, CHCHD2, SCD5, SERP1, PCCA, and CKAP4 (Supplementary Table S3).
Among the identified DEGs, CKAP4, Cytoskeleton-associated protein 4, appears particularly interesting. The corresponding protein is also known as Cytoskeleton-Linking Membrane Protein 63 (CLIMP-63) or p63, because it is a reversibly palmitoylated protein located in ER which stabilizes the ER structure and mediates the interaction of this organelle with MTs [117]. Notably, CKAP4 plays a role in maintaining mitochondrial functions through binding to VDAC2 at ER–mitochondria contact sites [118]. It also forms a complex with PI3K upon the binding of Dickkopf WNT Signaling Pathway Inhibitor 1 (DKK1), leading to the activation of Akt, which has been implied in glial cell tumors, as specified above. Both DKK1 and CKAP4 are frequently expressed in pancreatic and lung tumors, and their simultaneous expression is inversely correlated with cancer prognosis [119].
Another important interactor protein of CKAP4 is VIMP, Valosin-containing protein-interacting membrane protein (VIMP): together, they link the ER with microtubules [120]. Indeed, VIMP contributes to the organization of the ER acting as a scaffold for CLIMP-63 and polymerized MTs. Interestingly, we identified VIMP among the cytoskeletal genes upregulated in GB, suggesting the possibility that VIMP and CKAP4 could integrate the mitochondria–ER and cytoskeleton dynamics and functions, potentially contributing to the progression of GB.

7. Conclusions

GB is the most malignant glial tumor in the brain, with a devastating prognosis because of its complex biological behaviors and limited therapeutical strategies. While abnormalities in the expression/function of cytoskeletal proteins and in microtubules associated proteins in GB have been already reported [54,121], the potential contribution of MERCs in GB has been suggested only recently [65]. Recent advances in the knowledge of mitochondrial biology, with consequent implications in cancer, highlight the existence of interactions among cytoskeletal and OMM proteins implied in the control of mitochondrial functions and consequently of tissue-specific metabolic outcomes [65,121].
Here we propose for the first time the existence of a crosstalk among MTs and MERCs in the context of glial cells cancer. By identifying genes which are differentially expressed in normal samples with respect to GB, or in different GB grades (grade I versus grade IV), we found that about 37% of the known genes coding for MERCs proteins are abnormally expressed. This finding goes along with the altered expression of genes coding for cytoskeletal components or regulators. Both MERCs and microtubule dynamics are strong determinants of mitochondrial function: theoretically, their defects could also account for the metabolic hallmarks that characterize GB. Of course, future studies are needed to analyze in detail the MERCs–microtubules axis in GB, as our preliminary data rely on gene expression levels. Connecting them with changes in the function of MERCs and microtubules proteins requires further experimental analyses, guaranteeing new knowledge on the etiology of this severe brain tumor.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom12040567/s1, Figure S1. Tree Map. Results describes a summarization of GO terms of downregulated genes in GB. Analysis was performed using REVIGO application with default parameters. Each rectangle is a single cluster representative that are joined into ‘superclusters’ of related terms (same color). Table S1. Differentially expressed genes in GB and Glioma. Table S2. Differentially expressed genes categorized according to Gene Ontology. Table S3. MERCs genes altered in GB and in Glioma.

Author Contributions

F.G., C.V., S.C., M.G. and A.D.M. wrote and reviewed the manuscript, S.C., M.G. and A.D.M. analysed datasets; S.C., A.D.M. and M.G. conceived and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

We have used the following GEO datasets: GSE147352 and GSE79878.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pisapia, D.J. The Updated World Health Organization Glioma Classification: Cellular and Molecular Origins of Adult Infiltrating Gliomas. Arch. Pathol. Lab. Med. 2017, 141, 1633–1645. [Google Scholar] [CrossRef] [Green Version]
  2. Ostrom, Q.T.; Bauchet, L.; Davis, F.G.; Deltour, I.; Fisher, J.L.; Langer, C.E.; Pekmezci, M.; Schwartzbaum, J.A.; Turner, M.C.; Walsh, K.M.; et al. The Epidemiology of Glioma in Adults: A State of the Science Review. Neuro-Oncology 2014, 16, 896–913. [Google Scholar] [CrossRef] [Green Version]
  3. Perry, A.; Wesseling, P. Histologic Classification of Gliomas. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2016; Volume 134, pp. 71–95. ISBN 9780128029978. [Google Scholar]
  4. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A Summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [Green Version]
  5. Hayden, E.C. Genomics Boosts Brain-Cancer Work. Nature 2010, 463, 278. [Google Scholar] [CrossRef]
  6. Kuehn, B.M. Genomics Illuminates a Deadly Brain Cancer. JAMA-J. Am. Med. Assoc. 2010, 303, 925–927. [Google Scholar] [CrossRef]
  7. Neftel, C.; Laffy, J.; Filbin, M.G.; Hara, T.; Shore, M.E.; Rahme, G.J.; Richman, A.R.; Silverbush, D.; Shaw, M.L.; Hebert, C.M.; et al. An Integrative Model of Cellular States, Plasticity, and Genetics for Glioblastoma. Cell 2019, 178, 835–849. [Google Scholar] [CrossRef]
  8. Fisher, J.L.; Schwartzbaum, J.A.; Wrensch, M.; Wiemels, J.L. Epidemiology of Brain Tumors. Neurol. Clin. 2007, 25, 867–890. [Google Scholar] [CrossRef] [Green Version]
  9. Davis, M.E. Glioblastoma: Overview of Disease and Treatment. Clin. J. Oncol. Nurs. 2016, 20, S2–S8. [Google Scholar] [CrossRef] [Green Version]
  10. Katz, A.M.; Amankulor, N.M.; Pitter, K.; Helmy, K.; Squatrito, M.; Holland, E.C. Astrocyte-Specific Expression Patterns Associated with the PDGF-Induced Glioma Microenvironment. PLoS ONE 2012, 7, e32453. [Google Scholar] [CrossRef] [Green Version]
  11. Chakravarti, A.; Delaney, M.A.; Loeffler, J.S.; Muzikansky, A.; Dyson, N.J.; Noll, E.; Black, P.M.L. Prognostic and Pathologic Significance of Quantitative Protein Expression Profiling in Human Gliomas. Clin. Cancer Res. 2001, 7, 2387–2395. [Google Scholar]
  12. Bachoo, R.M.; Maher, E.A.; Ligon, K.L.; Sharpless, N.E.; Chan, S.S.; You, M.J.; Tang, Y.; DeFrances, J.; Stover, E.; Weissleder, R.; et al. Epidermal Growth Factor Receptor and Ink4a/Arf: Governing Terminal Differentiation and Transformation Stem Cell to Astrocyte Axis. Cancer Cell 2002, 1, 269–277. [Google Scholar] [CrossRef] [Green Version]
  13. Guan, X.; Hasan, M.N.; Maniar, S.; Jia, W.; Sun, D. Reactive Astrocytes in Glioblastoma Multiforme. Mol. Neurobiol. 2018, 55, 6927–6938. [Google Scholar] [CrossRef]
  14. Ishizaki, T.; Morishima, Y.; Okamoto, M.; Furuyashiki, T.; Kato, T.; Narumiya, S. Coordination of Microtubules and the Actin Cytoskeleton by the Rho Effector MDia1. Nat. Cell Biol. 2001, 3, 8–14. [Google Scholar] [CrossRef]
  15. Bunse, L.; Pusch, S.; Bunse, T.; Sahm, F.; Sanghvi, K.; Friedrich, M.; Alansary, D.; Sonner, J.K.; Green, E.; Deumelandt, K.; et al. Suppression of Antitumor T Cell Immunity by the Oncometabolite (R)-2-Hydroxyglutarate. Nat. Med. 2018, 24, 1192–1203. [Google Scholar] [CrossRef]
  16. Bassoy, E.Y.; Kasahara, A.; Chiusolo, V.; Jacquemin, G.; Boydell, E.; Zamorano, S.; Riccadonna, C.; Pellegatta, S.; Hulo, N.; Dutoit, V.; et al. ER–Mitochondria Contacts Control Surface Glycan Expression and Sensitivity to Killer Lymphocytes in Glioma Stem-like Cells. EMBO J. 2017, 36, 1493–1512. [Google Scholar] [CrossRef]
  17. Lee-Chang, C.; Rashidi, A.; Miska, J.; Zhang, P.; Pituch, K.C.; Hou, D.; Xiao, T.; Fischietti, M.; Kang, S.J.; Appin, C.L.; et al. Myeloid-Derived Suppressive Cells Promote B Cell-Mediated Immunosuppression via Transfer of PD-L1 in Glioblastoma. Cancer Immunol. Res. 2019, 7, 1928–1943. [Google Scholar] [CrossRef]
  18. Yilmaz, M.; Christofori, G. EMT, the Cytoskeleton, and Cancer Cell Invasion. Cancer Metastasis Rev. 2009, 28, 15–33. [Google Scholar] [CrossRef] [Green Version]
  19. Yeung, C.Y.C.; Taylor, S.H.; Garva, R.; Holmes, D.F.; Zeef, L.A.; Soininen, R.; Boot-Handford, R.P.; Kadler, K.E. Arhgap28 Is a RhoGAP That Inactivates RhoA and Downregulates Stress Fibers. PLoS ONE 2014, 9, e107036. [Google Scholar] [CrossRef] [Green Version]
  20. Fortin Ensign, S.P.; Mathews, I.T.; Symons, M.H.; Berens, M.E.; Tran, N.L. Implications of Rho GTPase Signaling in Glioma Cell Invasion and Tumor Progression. Front. Oncol. 2013, 3, 241. [Google Scholar] [CrossRef] [Green Version]
  21. McLendon, R.; Friedman, A.; Bigner, D.; van Meir, E.G.; Brat, D.J.; Mastrogianakis, G.M.; Olson, J.J.; Mikkelsen, T.; Lehman, N.; Aldape, K.; et al. Comprehensive Genomic Characterization Defines Human Glioblastoma Genes and Core Pathways. Nature 2008, 455, 1061–1068. [Google Scholar] [CrossRef]
  22. Liang, B.C.; Grootveld, M. The Importance of Mitochondria in the Tumourigenic Phenotype: Gliomas as the Paradigm (Review). Int. J. Mol. Med. 2011, 27, 159–171. [Google Scholar] [CrossRef] [Green Version]
  23. Arismendi-Morillo, G. Electron Microscopy Morphology of the Mitochondrial Network in Gliomas and Their Vascular Microenvironment. Biochim. Biophys. Acta-Bioenerg. 2011, 1807, 602–608. [Google Scholar] [CrossRef] [Green Version]
  24. Giacomello, M.; Pellegrini, L. The Coming of Age of the Mitochondria-ER Contact: A Matter of Thickness. Cell Death Differ. 2016, 23, 1417–1427. [Google Scholar] [CrossRef] [Green Version]
  25. Gurel, P.S.; Hatch, A.L.; Higgs, H.N. Connecting the Cytoskeleton to the Endoplasmic Reticulum and Golgi. Curr. Biol. 2014, 24, 660–672. [Google Scholar] [CrossRef] [Green Version]
  26. Friedman, J.R.; Webster, B.M.; Mastronarde, D.N.; Verhey, K.J.; Voeltz, G.K. ER Sliding Dynamics and ER-Mitochondrial Contacts Occur on Acetylated Microtubules. J. Cell Biol. 2010, 190, 363–375. [Google Scholar] [CrossRef] [Green Version]
  27. Almeida, C.G.; Yamada, A.; Tenza, D.; Louvard, D.; Raposo, G.; Coudrier, E. Myosin 1b Promotes the Formation of Post-Golgi Carriers by Regulating Actin Assembly and Membrane Remodelling at the Trans-Golgi Network. Nat. Cell Biol. 2011, 13, 779–789. [Google Scholar] [CrossRef]
  28. Nogales, E.; Wang, H.W. Structural Intermediates in Microtubule Assembly and Disassembly: How and Why? Curr. Opin. Cell Biol. 2006, 18, 179–184. [Google Scholar] [CrossRef]
  29. Akhmanova, A.; Steinmetz, M.O. Tracking the Ends: A Dynamic Protein Network Controls the Fate of Microtubule Tips. Nat. Rev. Mol. Cell Biol. 2008, 9, 309–322. [Google Scholar] [CrossRef]
  30. Desai, A.; Mitchison, T.J. Microtubule Polymerization Dynamics. Annu. Rev. Cell Dev. Biol. 1997, 13, 83–117. [Google Scholar] [CrossRef] [Green Version]
  31. Dumontet, C.; Jordan, M.A. Microtubule-Binding Agents: A Dynamic Field of Cancer Therapeutics. Nat. Rev. Drug Discov. 2010, 9, 790–803. [Google Scholar] [CrossRef] [Green Version]
  32. Liu, M.; Nadar, V.C.; Kozielski, F.; Kozlowska, M.; Yu, W.; Baas, P.W. Kinesin-12, a Mitotic Microtubule-Associated Motor Protein, Impacts Axonal Growth, Navigation, and Branching. J. Neurosci. 2010, 30, 14896–14906. [Google Scholar] [CrossRef] [Green Version]
  33. Chen, Y.; Sheng, Z.H. Kinesin-1-Syntaphilin Coupling Mediates Activity-Dependent Regulation of Axonal Mitochondrial Transport. J. Cell Biol. 2013, 202, 351–364. [Google Scholar] [CrossRef]
  34. Brickley, K.; Stephenson, F.A. Trafficking Kinesin Protein (TRAK)-Mediated Transport of Mitochondria in Axons of Hippocampal Neurons. J. Biol. Chem. 2011, 286, 18079–18092. [Google Scholar] [CrossRef] [Green Version]
  35. Wloga, D.; Joachimiak, E.; Louka, P.; Gaertig, J. Posttranslational Modifications of Tubulin and Cilia. Cold Spring Harb. Perspect. Biol. 2017, 9, a028159. [Google Scholar] [CrossRef]
  36. Caron, J.M. Posttranslational Modification of Tubulin by Palmitoylation: I. Mol. Biol. Cell. 1997, 8, 637–645. [Google Scholar] [CrossRef] [Green Version]
  37. Cardanho-Ramos, C.; Faria-Pereira, A.; Morais, V.A. Orchestrating Mitochondria in Neurons: Cytoskeleton as the Conductor. Cytoskeleton 2020, 77, 65–75. [Google Scholar] [CrossRef] [Green Version]
  38. Kapitein, L.C.; Hoogenraad, C.C. Building the Neuronal Microtubule Cytoskeleton. Neuron 2015, 87, 492–506. [Google Scholar] [CrossRef] [Green Version]
  39. Nulty, J.; Alsaffar, M.; Barry, D. Radial Glial Cells Organize the Central Nervous System via Microtubule Dependant Processes. Brain Res. 2015, 1625, 171–179. [Google Scholar] [CrossRef]
  40. Bossing, T.; Barros, C.S.; Fischer, B.; Russell, S.; Shepherd, D. Disruption of Microtubule Integrity Initiates Mitosis during CNS Repair. Dev. Cell 2012, 23, 433–440. [Google Scholar] [CrossRef] [Green Version]
  41. Ren, W.; Zhao, W.; Cao, L.; Huang, J. Involvement of the Actin Machinery in Programmed Cell Death. Front. Cell Dev. Biol. 2021, 8, 1889. [Google Scholar] [CrossRef]
  42. Gentile, J.E.; Carrizales, M.G.; Koleske, A.J. Control of Synapse Structure and Function by Actin and Its Regulators. Cells 2022, 11, 603. [Google Scholar] [CrossRef]
  43. Illescas, M.; Peñas, A.; Arenas, J.; Martín, M.A.; Ugalde, C. Regulation of Mitochondrial Function by the Actin Cytoskeleton. Front. Cell Dev. Biol. 2021, 9, 795838. [Google Scholar] [CrossRef]
  44. Tokuraku, K.; Kuragano, M.; Uyeda, T.Q.P. Long-Range and Directional Allostery of Actin Filaments Plays Important Roles in Various Cellular Activities. Int. J. Mol. Sci. 2020, 21, 3209. [Google Scholar] [CrossRef]
  45. Yang, T.D.; Park, K.; Park, J.S.; Lee, J.H.; Choi, E.; Lee, J.; Choi, W.; Choi, Y.; Lee, K.J. Two Distinct Actin Waves Correlated with Turns-and-Runs of Crawling Microglia. PLoS ONE 2019, 14, e0220810. [Google Scholar] [CrossRef]
  46. Das, R.; Chinnathambi, S. Actin-Mediated Microglial Chemotaxis via G-Protein Coupled Purinergic Receptor in Alzheimer’s Disease. Neuroscience 2020, 448, 325–336. [Google Scholar] [CrossRef]
  47. Uhlemann, R.; Gertz, K.; Boehmerle, W.; Schwarz, T.; Nolte, C.; Freyer, D.; Kettenmann, H.; Endres, M.; Kronenberg, G. Actin Dynamics Shape Microglia Effector Functions. Brain Struct. Funct. 2016, 221, 2717–2734. [Google Scholar] [CrossRef]
  48. Kreplak, L.; Bär, H.; Leterrier, J.F.; Herrmann, H.; Aebi, U. Exploring the Mechanical Behavior of Single Intermediate Filaments. J. Mol. Biol. 2005, 354, 569–577. [Google Scholar] [CrossRef]
  49. Fan, C.; Shi, X.; Zhao, K.; Wang, L.; Shi, K.; Liu, Y.-J.; Li, H.; Ji, B.; Jiu, Y. Cell Migration Orchestrates Migrasome Formation by Shaping Retraction Fibers. J. Cell Biol. 2022, 221, e202109168. [Google Scholar] [CrossRef]
  50. Velez-Delvalle, C.; Marsch-Moreno, M.; Castro-Muñozledo, F.; Galván-Mendoza, I.J.; Kuri-Harcuch, W. Epithelial Cell Migration Requires the Interaction between the Vimentin and Keratin Intermediate Filaments. Sci. Rep. 2016, 6, 24389. [Google Scholar] [CrossRef] [Green Version]
  51. Maurer, M.; Lammerding, J. The Driving Force: Nuclear Mechanotransduction in Cellular Function, Fate, and Disease. Annu. Rev. Biomed. Eng. 2019, 21, 443–468. [Google Scholar] [CrossRef]
  52. Katsetos, C.D.; Anni, H.; Dráber, P. Mitochondrial Dysfunction in Gliomas. Semin. Pediatric Neurol. 2013, 20, 216–227. [Google Scholar] [CrossRef]
  53. Dráberová, E.; Lukáš, Z.; Ivanyi, D.; Viklický, V.; Dráber, P. Expression Class III β-Tubulin in Normal and Neoplastic Human Tissues. Histochem. Cell Biol. 1998, 109, 231–239. [Google Scholar] [CrossRef]
  54. Suzuki, S.O.; Kitai, R.; Llena, J.; Lee, S.C.; Goldman, J.E.; Shafit-Zagardo, B. MAP-2e, a Novel MAP-2 Isoform, Is Expressed in Gliomas and Delineates Tumor Architecture and Patterns of Infiltration. J. Neuropathol. Exp. Neurol. 2002, 61, 403–412. [Google Scholar] [CrossRef] [Green Version]
  55. Lim, E.J.; Suh, Y.; Yoo, K.C.; Lee, J.H.; Kim, I.G.; Kim, M.J.; Chang, J.H.; Kang, S.G.; Lee, S.J. Tumor-Associated Mesenchymal Stem-like Cells Provide Extracellular Signaling Cue for Invasiveness of Glioblastoma Cells. Oncotarget 2017, 8, 1438–1448. [Google Scholar] [CrossRef] [Green Version]
  56. Suzuki, S.O.; McKenney, R.J.; Mawatari, S.Y.; Mizuguchi, M.; Mikami, A.; Iwaki, T.; Goldman, J.E.; Canoll, P.; Vallee, R.B. Expression Patterns of LIS1, Dynein and Their Interaction Partners Dynactin, NudE, NudEL and NudC in Human Gliomas Suggest Roles in Invasion and Proliferation. Acta Neuropathol. 2007, 113, 591–599. [Google Scholar] [CrossRef]
  57. Xie, Z.; Janczyk, P.; Zhang, Y.; Liu, A.; Shi, X.; Singh, S.; Facemire, L.; Kubow, K.; Li, Z.; Jia, Y.; et al. A Cytoskeleton Regulator AVIL Drives Tumorigenesis in Glioblastoma. Nat. Commun. 2020, 11, 3457. [Google Scholar] [CrossRef]
  58. Kobielak, A.; Pasolli, H.A.; Fuchs, E. Mammalian Formin-1 Participates in Adherens Junctions and Polymerization of Linear Actin Cables. Nat. Cell Biol. 2004, 6, 21–30. [Google Scholar] [CrossRef]
  59. Monzo, P.; Crestani, M.; Chong, Y.K.; Ghisleni, A.; Hennig, K.; Li, Q.; Kakogiannos, N.; Giannotta, M.; Richichi, C.; Dini, T.; et al. Adaptive Mechanoproperties Mediated by the Formin FMN1 Characterize Glioblastoma Fitness for Invasion. Dev. Cell 2021, 56, 2841–2855.e8. [Google Scholar] [CrossRef]
  60. Wang, Q.; Wu, H.; Hu, J.; Fu, H.; Qu, Y.; Yang, Y.; Cai, Q.; Efimov, A.; Wu, M.; Yen, T.; et al. Nestin Is Required for Spindle Assembly and Cell Cycle Progression in Glioblastoma Cells. Mol. Cancer Res. 2021, 19, 1651–1665. [Google Scholar] [CrossRef]
  61. Gatti, G.; Vilardo, L.; Musa, C.; di Pietro, C.; Bonaventura, F.; Scavizzi, F.; Torcinaro, A.; Bucci, B.; Saporito, R.; Arisi, I.; et al. Role of Lamin A/C as Candidate Biomarker of Aggressiveness and Tumorigenicity in Glioblastoma Multiforme. Biomedicines 2021, 9, 1343. [Google Scholar] [CrossRef]
  62. Mottaghitalab, F.; Lanjanian, H.; Masoudi-Nejad, A. Revealing Transcriptional and Post-Transcriptional Regulatory Mechanisms of γ-Glutamyl Transferase and Keratin Isoforms as Novel Cooperative Biomarkers in Low-Grade Glioma and Glioblastoma Multiforme. Genomics 2021, 113, 2623–2633. [Google Scholar] [CrossRef] [PubMed]
  63. Evans, S.M.; Judy, K.D.; Dunphy, I.; Jenkins, W.T.; Hwang, W.-T.; Nelson, P.T.; Lustig, R.A.; Jenkins, K.; Magarelli, D.P.; Hahn, S.M.; et al. Hypoxia Is Important in the Biology and Aggression of Human Glial Brain Tumors. Clin. Cancer Res. 2004, 10, 8177–8184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Theos, A.C.; Martina, A.; Hurbain, I.; Peden, A.A.; Sviderskaya, E.V.; Stewart, A.; Robinson, M.S.; Bennett, D.C.; Cutler, D.F.; Bonifacino, J.S.; et al. Functions of Adaptor Protein (AP)-3 and AP-1 in Tyrosinase Sorting from Endosomes to Melanosomes. Mol. Biol. Cell 2005, 16, 5356–5372. [Google Scholar] [CrossRef] [PubMed]
  65. van Spronsen, M.; Mikhaylova, M.; Lipka, J.; Schlager, M.A.; van den Heuvel, D.J.; Kuijpers, M.; Wulf, P.S.; Keijzer, N.; Demmers, J.; Kapitein, L.C.; et al. TRAK/Milton Motor-Adaptor Proteins Steer Mitochondrial Trafficking to Axons and Dendrites. Neuron 2013, 77, 485–502. [Google Scholar] [CrossRef] [Green Version]
  66. Glater, E.E.; Megeath, L.J.; Stowers, R.S.; Schwarz, T.L. Axonal Transport of Mitochondria Requires Milton to Recruit Kinesin Heavy Chain and Is Light Chain Independent. J. Cell Biol. 2006, 173, 545–557. [Google Scholar] [CrossRef] [Green Version]
  67. Smirnova, E.; Griparic, L.; Shurland, D.-L.; van der Bliek, A.M. Drp1 Is Required for Mitochondrial Division in Mammalian Cells. Mol. Biol. Cell 2001, 12, 2245–2256. [Google Scholar] [CrossRef] [Green Version]
  68. Giacomello, M.; Pyakurel, A.; Glytsou, C.; Scorrano, L. The Cell Biology of Mitochondrial Membrane Dynamics. Nat. Rev. Mol. Cell Biol. 2020, 21, 204–224. [Google Scholar] [CrossRef]
  69. Xie, Q.; Wu, Q.; Horbinski, C.M.; Flavahan, W.A.; Yang, K.; Zhou, W.; Dombrowski, S.M.; Huang, Z.; Fang, X.; Shi, Y.; et al. Mitochondrial Control by DRP1 in Brain Tumor Initiating Cells. Nat. Neurosci. 2015, 18, 501–510. [Google Scholar] [CrossRef] [Green Version]
  70. De Mario, A.; Tosatto, A.; Hill, J.M.; Kriston-Vizi, J.; Ketteler, R.; Vecellio Reane, D.; Cortopassi, G.; Szabadkai, G.; Rizzuto, R.; Mammucari, C. Identification and Functional Validation of FDA-Approved Positive and Negative Modulators of the Mitochondrial Calcium Uniporter. Cell Rep. 2021, 35, 109275. [Google Scholar] [CrossRef]
  71. Iranmanesh, Y.; Jiang, B.; Favour, O.C.; Dou, Z.; Wu, J.; Li, J.; Sun, C. Mitochondria’s Role in the Maintenance of Cancer Stem Cells in Glioblastoma. Front. Oncol. 2021, 11, 101. [Google Scholar] [CrossRef]
  72. Caino, M.C.; Ghosh, J.C.; Chae, Y.C.; Vaira, V.; Rivadeneira, D.B.; Faversani, A.; Rampini, P.; Kossenkov, A.V.; Aird, K.M.; Zhang, R.; et al. PI3K Therapy Reprograms Mitochondrial Trafficking to Fuel Tumor Cell Invasion. Proc. Natl. Acad. Sci. USA 2015, 112, 8638–8643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Altieri, D.C. Mitochondrial Dynamics and Metastasis. Cell. Mol. Life Sci. 2019, 76, 827–835. [Google Scholar] [CrossRef] [PubMed]
  74. Templeton, K.; Ramos, M.; Rose, J.; Le, B.; Zhou, Q.; Cressman, A.; Ferreyra, S.; Lebrilla, C.B.; Fierro, F.A. Mesenchymal Stromal Cells Regulate Sialylations of N-Glycans, Affecting Cell Migration and Survival. Int. J. Mol. Sci. 2021, 22, 6868. [Google Scholar] [CrossRef] [PubMed]
  75. De Mario, A.; Scarlatti, C.; Costiniti, V.; Primerano, S.; Lopreiato, R.; Calì, T.; Brini, M.; Giacomello, M.; Carafoli, E. Calcium Handling by Endoplasmic Reticulum and Mitochondria in a Cell Model of Huntington’s Disease. PLoS Curr. 2016, 8. [Google Scholar] [CrossRef]
  76. De Mario, A.; Quintana-Cabrera, R.; Martinvalet, D.; Giacomello, M. (Neuro)Degenerated Mitochondria-ER Contacts. Biochem. Biophys. Res. Commun. 2017, 483, 1096–1109. [Google Scholar] [CrossRef]
  77. Vance, J.E. Phospholipid Synthesis and Transport in Mammalian Cells. Traffic 2015, 16, 1–18. [Google Scholar] [CrossRef]
  78. Cui, Z.; Vance, J.E.; Chen, M.H.; Voelker, D.R.; Vance, D.E. Cloning and Expression of a Novel Phosphatidylethanolamine N-Methyltransferase. A Specific Biochemical and Cytological Marker for a Unique Membrane Fraction in Rat Liver. J. Biol. Chem. 1993, 268, 16655–16663. [Google Scholar] [CrossRef]
  79. Galmes, R.; Houcine, A.; Vliet, A.R.; Agostinis, P.; Jackson, C.L.; Giordano, F. ORP5/ORP8 Localize to Endoplasmic Reticulum–Mitochondria Contacts and Are Involved in Mitochondrial Function. EMBO Rep. 2016, 17, 800–810. [Google Scholar] [CrossRef] [Green Version]
  80. Stone, S.J.; Levin, M.C.; Zhou, P.; Han, J.; Walther, T.C.; Farese, R.V. The Endoplasmic Reticulum Enzyme DGAT2 Is Found in Mitochondria-Associated Membranes and Has a Mitochondrial Targeting Signal That Promotes Its Association with Mitochondria. J. Biol. Chem. 2009, 284, 5352–5361. [Google Scholar] [CrossRef] [Green Version]
  81. Prasad, M.; Kaur, J.; Pawlak, K.J.; Bose, M.; Whittal, R.M.; Bose, H.S. Mitochondria-Associated Endoplasmic Reticulum Membrane (MAM) Regulates Steroidogenic Activity via Steroidogenic Acute Regulatory Protein (StAR)-Voltage-Dependent Anion Channel 2 (VDAC2) Interaction. J. Biol. Chem. 2015, 290, 2604–2616. [Google Scholar] [CrossRef] [Green Version]
  82. Lewin, T.M.; Kim, J.H.; Granger, D.A.; Vance, J.E.; Coleman, R.A. Acyl-CoA Synthetase Isoforms 1, 4, and 5 Are Present in Different Subcellular Membranes in Rat Liver and Can Be Inhibited Independently. J. Biol. Chem. 2001, 276, 24674–24679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Giacomello, M.; Drago, I.; Bortolozzi, M.; Scorzeto, M.; Gianelle, A.; Pizzo, P.; Pozzan, T. Ca2+ Hot Spots on the Mitochondrial Surface Are Generated by Ca2+ Mobilization from Stores, but Not by Activation of Store-Operated Ca2+ Channels. Mol. Cell 2010, 38, 280–290. [Google Scholar] [CrossRef] [PubMed]
  84. Rebelo, A.P.; Dal Bello, F.; Knedlik, T.; Volpin, F.; Shin, S.H.; Giacomello, M. Chemical modulation of Mitochondria-Endoplasmic Reticulum Contact Sites. Cells 2020, 9, 1637. [Google Scholar] [CrossRef] [PubMed]
  85. De Mario, A.; Peggion, C.; Massimino, M.L.; Viviani, F.; Castellani, A.; Giacomello, M.; Lim, D.; Bertoli, A.; Sorgato, M.C. The Prion Protein Regulates Glutamate-Mediated Ca2+ Entry and Mitochondrial Ca2+ Accumulation in Neurons. J. Cell Sci. 2017, 130, 2736–2746. [Google Scholar] [CrossRef] [Green Version]
  86. Csordás, G.; Várnai, P.; Golenár, T.; Roy, S.; Purkins, G.; Schneider, T.G.; Balla, T.; Hajnóczky, G. Imaging Interorganelle Contacts and Local Calcium Dynamics at the ER-Mitochondrial Interface. Mol. Cell 2010, 39, 121–132. [Google Scholar] [CrossRef]
  87. Szabadkai, G.; Bianchi, K.; Várnai, P.; de Stefani, D.; Wieckowski, M.R.; Cavagna, D.; Nagy, A.I.; Balla, T.; Rizzuto, R. Chaperone-Mediated Coupling of Endoplasmic Reticulum and Mitochondrial Ca2+ Channels. J. Cell Biol. 2006, 175, 901–911. [Google Scholar] [CrossRef] [Green Version]
  88. Vandecaetsbeek, I.; Vangheluwe, P.; Raeymaekers, L.; Wuytack, F.; Vanoevelen, J. The Ca2+ Pumps of the Endoplasmic Reticulum and Golgi Apparatus. Cold Spring Harb. Perspect. Biol. 2011, 3, a004184. [Google Scholar] [CrossRef] [Green Version]
  89. Das, S.; Fransson, Å.; Aspenstrom, P. Bidirectional Ca2+-Dependent Control of Mitochondrial Dynamics by the Miro GTPase. Proc. Natl. Acad. Sci. USA 2008, 105, 20728–20733. [Google Scholar] [CrossRef] [Green Version]
  90. Berenguer-Escuder, C.; Grossmann, D.; Massart, F.; Antony, P.; Burbulla, L.F.; Glaab, E.; Imhoff, S.; Trinh, J.; Seibler, P.; Grünewald, A.; et al. Variants in Miro1 Cause Alterations of ER- Mitochondria Contact Sites in Fibroblasts from Parkinson’s Disease Patients. J. Clin. Med. 2019, 8, 2226. [Google Scholar] [CrossRef] [Green Version]
  91. Grossmann, D.; Berenguer-Escuder, C.; Bellet, M.E.; Scheibner, D.; Bohler, J.; Massart, F.; Rapaport, D.; Skupin, A.; Fouquier D’Hérouël, A.; Sharma, M.; et al. Mutations in RHOT1 Disrupt Endoplasmic Reticulum-Mitochondria Contact Sites Interfering with Calcium Homeostasis and Mitochondrial Dynamics in Parkinson’s Disease. Antioxid. Redox Signal. 2019, 31, 1213–1234. [Google Scholar] [CrossRef] [Green Version]
  92. Liu, S.; Sawada, T.; Lee, S.; Yu, W.; Silverio, G.; Alapatt, P.; Millan, I.; Shen, A.; Saxton, W.; Kanao, T.; et al. Parkinson’s Disease-Associated Kinase PINK1 Regulates Miro Protein Level and Axonal Transport of Mitochondria. PLoS Genet. 2012, 8, e1002537. [Google Scholar] [CrossRef] [PubMed]
  93. Birsa, N.; Norkett, R.; Wauer, T.; Mevissen, T.E.T.; Wu, H.C.; Foltynie, T.; Bhatia, K.; Hirst, W.D.; Komander, D.; Plun-Favreau, H.; et al. Lysine 27 Ubiquitination of the Mitochondrial Transport Protein Miro Is Dependent on Serine 65 of the Parkin Ubiquitin Ligase. J. Biol. Chem. 2014, 289, 14569–14582. [Google Scholar] [CrossRef] [Green Version]
  94. Giacomello, M.; Drago, I.; Pizzo, P.; Pozzan, T. Mitochondrial Ca2+ as a Key Regulator of Cell Life and Death. Cell Death Differ. 2007, 14, 1267–1274. [Google Scholar] [CrossRef] [PubMed]
  95. Boehning, D.; Patterson, R.L.; Sedaghat, L.; Glebova, N.O.; Kurosaki, T.; Snyder, S.H. Cytochrome c Binds to Inositol (1,4,5) Trisphosphate Receptors, Amplifying Calcium-Dependent Apoptosis. Nat. Cell Biol. 2003, 5, 1051–1061. [Google Scholar] [CrossRef] [PubMed]
  96. Pinton, P.; Giorgi, C.; Pandolfi, P.P. The Role of PML in the Control of Apoptotic Cell Fate: A New Key Player at ER-Mitochondria Sites. Cell Death Differ. 2011, 18, 1450–1456. [Google Scholar] [CrossRef] [Green Version]
  97. Molina, J.R.; Hayashi, Y.; Stephens, C.; Georgescu, M.M. Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation. Neoplasia 2010, 12, 453–463. [Google Scholar] [CrossRef] [Green Version]
  98. Arismendi-Morillo, G.J.; Castellano-Ramirez, A.V. Ultrastructural Mitochondrial Pathology in Human Astrocytic Tumors: Potentials Implications pro-Therapeutics Strategies. J. Electron. Microsc. 2008, 57, 33–39. [Google Scholar] [CrossRef]
  99. Vlashi, E.; Lagadec, C.; Vergnes, L.; Matsutani, T.; Masui, K.; Poulou, M.; Popescu, R.; della Donna, L.; Evers, P.; Dekmezian, C.; et al. Metabolic State of Glioma Stem Cells and Nontumorigenic Cells. Proc. Natl. Acad. Sci. USA 2011, 108, 16062–16067. [Google Scholar] [CrossRef] [Green Version]
  100. Arismendi-Morillo, G.; Castellano-Ramírez, A.; Seyfried, T.N. Functional and Therapeutic Implications of Mitochondrial Network and Mitochondria-Associated Membranes: The Glioma’s Case. Glioma-Contemp. Diagn. Ther. Approaches 2019. [Google Scholar] [CrossRef] [Green Version]
  101. Moser, J.J.; Fritzler, M.J.; Rattner, J.B. Ultrastructural Characterization of Primary Cilia in Pathologically Characterized Human Glioblastoma Multiforme (GBM) Tumors. BMC Clin. Pathol. 2014, 14, 40. [Google Scholar] [CrossRef] [Green Version]
  102. Moser, J.J.; Fritzler, M.J.; Rattner, J.B. Primary Ciliogenesis Defects Are Associated with Human Astrocytoma/Glioblastoma Cells. BMC Cancer 2009, 9, 448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Carré, M.; André, N.; Carles, G.; Borghi, H.; Brichese, L.; Briand, C.; Braguer, D.; Medicine, U.F.R.; La, U.O. Tubulin Is an Inherent Component of Mitochondrial Membranes That Interacts with the Voltage-Dependent Anion Channel. J. Biol. Chem. 2002, 277, 33664–33669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Puurand, M.; Tepp, K.; Timohhina, N.; Aid, J.; Shevchuk, I.; Chekulayev, V.; Kaambre, T. Tubulin ΒII and ΒIII Isoforms as the Regulators of VDAC Channel Permeability in Health and Disease. Cells 2019, 8, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Maldonado, E.N.; Sheldon, K.L.; Dehart, D.N.; Patnaik, J.; Manevich, Y.; Townsend, D.M.; Bezrukov, S.M.; Rostovtseva, T.K.; Lemasters, J.J. Voltage-Dependent Anion Channels Modulate Mitochondrial Metabolism in Cancer Cells: Regulation by Free Tubulin and Erastin. J. Biol. Chem. 2013, 288, 11920–11929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Strickland, M.; Stoll, E.A. Metabolic Reprogramming in Glioma. Front. Cell Dev. Biol. 2017, 5, 43. [Google Scholar] [CrossRef] [Green Version]
  107. Mucignat-Caretta, C.; Cavaggioni, A.; Redaelli, M.; Malatesta, M.; Zancanaro, C.; Caretta, A. Selective Distribution of Protein Kinase A Regulatory Subunit Rllα in Rodent Gliomas. Neuro-Oncology 2008, 10, 958–967. [Google Scholar] [CrossRef] [Green Version]
  108. Zhao, P.; Li, Q.; Shi, Z.; Li, C.; Wang, L.; Liu, X.; Jiang, C.; Qian, X.; You, Y.; Liu, N.; et al. GSK-3β Regulates Tumor Growth and Angiogenesis in Human Glioma Cells Theseauthorshavecontributedequallytothiswork. Oncotarget 2021, 6, 31901–31915. [Google Scholar] [CrossRef] [Green Version]
  109. Collet, B.; Guitton, N.; Saïkali, S.; Avril, T.; Pineau, C.; Hamlat, A.; Mosser, J.; Quillien, V. Differential Analysis of Glioblastoma Multiforme Proteome by a 2D-DIGE Approach. Proteome Sci. 2011, 9, 16. [Google Scholar] [CrossRef]
  110. Cui, Y.; Li, J.; Weng, L.; Wirbisky, S.E.; Freeman, J.L.; Liu, J.; Liu, Q.; Yuan, X.; Irudayaraj, J. Regulatory Landscape and Clinical Implication of MBD3 in Human Malignant Glioma. Oncotarget 2016, 7, 81698–81714. [Google Scholar] [CrossRef] [Green Version]
  111. Emig, D.; Salomonis, N.; Baumbach, J.; Lengauer, T.; Conklin, B.R.; Albrecht, M. AltAnalyze and DomainGraph: Analyzing and Visualizing Exon Expression Data. Nucleic Acids Res. 2010, 38, 755–762. [Google Scholar] [CrossRef] [Green Version]
  112. Alessio, E.; Buson, L.; Chemello, F.; Peggion, C.; Grespi, F.; Martini, P.; Massimino, M.L.; Pacchioni, B.; Millino, C.; Romualdi, C.; et al. Single Cell Analysis Reveals the Involvement of the Long Non-Coding RNA Pvt1 in the Modulation of Muscle Atrophy and Mitochondrial Network. Nucleic Acids Res. 2019, 47, 1653–1670. [Google Scholar] [CrossRef] [Green Version]
  113. Pagliari, M.; Munari, F.; Toffoletto, M.; Lonardi, S.; Chemello, F.; Codolo, G.; Millino, C.; Della Bella, C.; Pacchioni, B.; Vermi, W.; et al. Helicobacter Pylori Affects the Antigen Presentation Activity of Macrophages Modulating the Expression of the Immune Receptor CD300E through MiR-4270. Front. Immunol. 2017, 8, 1288. [Google Scholar] [CrossRef] [Green Version]
  114. Howe, E.; Holton, K.; Nair, S.; Schlauch, D.; Sinha, R.; Quackenbush, J. MeV: MultiExperiment Viewer. In Biomedical Informatics for Cancer Research; Springer: Berlin/Heidelberg, Germany, 2010; pp. 267–277. ISBN 9781441957122. [Google Scholar]
  115. Mi, H.; Thomas, P. PANTHER Pathway: An Ontology-Based Pathway Database Coupled with Data Analysis Tools. Methods Mol. Biol. 2009, 563, 123–140. [Google Scholar] [CrossRef] [PubMed]
  116. Supek, F.; Bošnjak, M.; Škunca, N.; Šmuc, T. Revigo Summarizes and Visualizes Long Lists of Gene Ontology Terms. PLoS ONE 2011, 6, e21800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Schweizer, A.; Ericsson, M.; Bachi, T.; Griffiths, G.; Hauri, H.P. Characterization of a Novel 63 KDa Membrane Protein. Implications for the Organization of the ER-to-Golgi Pathway. J. Cell Sci. 1993, 104, 671–683. [Google Scholar] [CrossRef] [PubMed]
  118. Harada, T.; Sada, R.; Osugi, Y.; Matsumoto, S.; Matsuda, T.; Hayashi-Nishino, M.; Nagai, T.; Harada, A.; Kikuchi, A. Palmitoylated CKAP4 Regulates Mitochondrial Functions through an Interaction with VDAC2 at ER-Mitochondria Contact Sites. J. Cell Sci. 2020, 133, jcs249045. [Google Scholar] [CrossRef] [PubMed]
  119. Kikuchi, A.; Fumoto, K.; Kimura, H. The Dickkopf1-Cytoskeleton-Associated Protein 4 Axis Creates a Novel Signalling Pathway and May Represent a Molecular Target for Cancer Therapy. Br. J. Pharmacol. 2017, 174, 4651–4665. [Google Scholar] [CrossRef] [Green Version]
  120. Noda, C.; Kimura, H.; Arasaki, K.; Matsushita, M.; Yamamoto, A.; Wakana, Y.; Inoue, H.; Tagaya, M. Valosin-Containing Protein-Interacting Membrane Protein (VIMP) Links the Endoplasmic Reticulum with Microtubules in Concert with Cytoskeleton-Linking Membrane Protein (CLIMP)-63. J. Biol. Chem. 2014, 289, 24304–24313. [Google Scholar] [CrossRef] [Green Version]
  121. Kuznetsov, A.V.; Javadov, S.; Grimm, M.; Margreiter, R.; Ausserlechner, M.J.; Hagenbuchner, J. Crosstalk between Mitochondria and Cytoskeleton in Cardiac Cells. Cells 2020, 9, 222. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Enriched GO terms. (A). GO enrichment from upregulated genes in GB from GSE147352 dataset. (B). GO enrichment from upregulated genes in GB grade IV from GSE79878 dataset. The bubble color indicates the FDR for the GO term (bright red lower FDR). Highly similar GO terms are linked by edges in the graph.
Figure 1. Enriched GO terms. (A). GO enrichment from upregulated genes in GB from GSE147352 dataset. (B). GO enrichment from upregulated genes in GB grade IV from GSE79878 dataset. The bubble color indicates the FDR for the GO term (bright red lower FDR). Highly similar GO terms are linked by edges in the graph.
Biomolecules 12 00567 g001
Figure 2. Differentially expressed genes involved in MERC interactions. Genes associated to MERC interactions are represented according to subcellular localization of coded proteins. Proteins specifically localized in the endoplasmic reticulum are associated with the node labelled as er, those associated with mitochondria are associated with the node labelled with mit, and those localized both in ER on in the mitochondria are associated with the node labelled with er–mitoch. Proteins involved in MERCs according to [104] not specifically associated with ER or mitochondria were linked to a node labelled with int. All nodes are linked to the MERCs node because all are involved in the formation of MERCs. MERCs, ER, mitochondria, and ER–mitochondria nodes are colored with light blue while other nodes are colored according to the log FC expression of control versus high grade glioblastoma. Differently node border is colored according to the log FC expression of control versus glioma. Blue indicates highly expressed in the tumor, yellow or red in the control, and white indicates non-differences.
Figure 2. Differentially expressed genes involved in MERC interactions. Genes associated to MERC interactions are represented according to subcellular localization of coded proteins. Proteins specifically localized in the endoplasmic reticulum are associated with the node labelled as er, those associated with mitochondria are associated with the node labelled with mit, and those localized both in ER on in the mitochondria are associated with the node labelled with er–mitoch. Proteins involved in MERCs according to [104] not specifically associated with ER or mitochondria were linked to a node labelled with int. All nodes are linked to the MERCs node because all are involved in the formation of MERCs. MERCs, ER, mitochondria, and ER–mitochondria nodes are colored with light blue while other nodes are colored according to the log FC expression of control versus high grade glioblastoma. Differently node border is colored according to the log FC expression of control versus glioma. Blue indicates highly expressed in the tumor, yellow or red in the control, and white indicates non-differences.
Biomolecules 12 00567 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Grespi, F.; Vianello, C.; Cagnin, S.; Giacomello, M.; De Mario, A. The Interplay of Microtubules with Mitochondria–ER Contact Sites (MERCs) in Glioblastoma. Biomolecules 2022, 12, 567. https://doi.org/10.3390/biom12040567

AMA Style

Grespi F, Vianello C, Cagnin S, Giacomello M, De Mario A. The Interplay of Microtubules with Mitochondria–ER Contact Sites (MERCs) in Glioblastoma. Biomolecules. 2022; 12(4):567. https://doi.org/10.3390/biom12040567

Chicago/Turabian Style

Grespi, Francesca, Caterina Vianello, Stefano Cagnin, Marta Giacomello, and Agnese De Mario. 2022. "The Interplay of Microtubules with Mitochondria–ER Contact Sites (MERCs) in Glioblastoma" Biomolecules 12, no. 4: 567. https://doi.org/10.3390/biom12040567

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