Next Article in Journal
Health Benefits of Dietary Fiber for the Management of Inflammatory Bowel Disease
Previous Article in Journal
Systemic Inflammation Predicts Alzheimer Pathology in Community Samples without Dementia
Previous Article in Special Issue
Neurological Benefits, Clinical Challenges, and Neuropathologic Promise of Medical Marijuana: A Systematic Review of Cannabinoid Effects in Multiple Sclerosis and Experimental Models of Demyelination
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparative Profiling of TG2 and Its Effectors in Human Relapsing Remitting and Progressive Multiple Sclerosis

by
Damien D. Pearse
1,2,3,4,5,
Andrew B. Hefley
1,
Alejo A. Morales
1 and
Mousumi Ghosh
1,2,5,*
1
The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
2
The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
3
The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
4
The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
5
Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(6), 1241; https://doi.org/10.3390/biomedicines10061241
Submission received: 9 February 2022 / Revised: 6 May 2022 / Accepted: 13 May 2022 / Published: 26 May 2022

Abstract

:
Multiple Sclerosis (MS) is a chronic CNS autoimmune disease characterized by immune-mediated demyelination, axon loss, and disability. Dysregulation of transglutaminase-2 (TG2) has been implicated in disease initiation and progression. Herein, TG2 expression in post-mortem human brain tissue from Relapsing Remitting MS (RRMS) or Progressive MS (PMS) individuals were examined and correlated with the presence of TG2 binding partners and effectors implicated in the processes of inflammation, scar formation, and the antagonism of repair. Tissues from Relapsing-Remitting Multiple Sclerosis (RRMS; n = 6), Progressive Multiple Sclerosis (PMS; n = 5), and non-MS control (n = 6) patients underwent immunohistochemistry for TG2, PLA2, COX-2, FN, CSPG, and HSPG. TG2 was strongly upregulated in active RRMS and PMS lesions, within blood vessels and the perivascular tissue of sclerotic plaques. TG2 colocalization was observed with GFAP+ astrocytes and ECM, including FN, HSPG, and CSPG, which also increased in either RRMS or PMS lesions. Although TG2 was not colocalized with inflammatory mediators COX-2 and PLA2, or the macrophage-microglia marker Iba1, its increased expression correlated with their elevation in active RRMS and PMS lesions. In summary, the correlation of strong TG2 induction in either RRMS or PMS with some of its binding partners but not others implicates potentially different roles for TG2 in disparate MS forms that may warrant further investigation.

1. Introduction

Multiple Sclerosis (MS) is a debilitating and chronic autoimmune neurodegenerative disease of the central nervous system (CNS) that exhibits inflammation mediated demyelination [1,2]. MS leads to diverse neurological deficits, progressive loss of sensorimotor function, and clinical disability [3,4]. The etiology underlying MS pathogenesis has been proposed to involve environmental factors, particularly exposure to the Epstein–Barr virus (EBV) and vitamin D insufficiencies as well as ultraviolet light exposure, smoking, and stress. When combined with unique genetic predispositions related to gender and ethnicity, these factors contribute to MS susceptibility [5,6].
The clinical disease course of MS can be highly variable, with four distinct clinical subtypes identified. In the clinically isolated syndrome (CIS), only a single episode of neurological symptoms resulting from inflammation or demyelination in the CNS might occur. However, most MS patients (>85%) experience an inflammation-induced, relapsing remitting (RRMS) form of the disease that exhibits acute exacerbations with one or more neurological deficits, followed by a partial or complete recovery of symptoms. Whereas RRMS occurs in the initial stage of MS, in the later stages of the disease, around 65% of patients with RRMS will convert to a non-relapsing form, with a progression of neurological symptoms and disability. Secondary progressive MS (SPMS) is associated with a degenerative condition of the neuron–axonal axis. Lastly, 10–15% of MS patients are diagnosed with primary progressive MS (PPMS), where neurological deficits continue to worsen gradually from the disease onset [7].
The immunopathophysiology of MS remains to be fully elucidated, although the dysregulation of both the innate and the adaptive immune response is involved [8,9], leading to the aberrant immune targeting of oligodendrocytes and the myelin sheath. The key non-CNS cellular components of MS pathophysiology include CD4+ Th1 and Th17 cells, CD20+ B cells, CD8+ T cells, monocytes, neutrophils, macrophages, and dendritic cells [10,11]. The interactions of these immune cells with CNS resident glia, microglia, and astrocytes are important for disease pathogenesis [8,9,12]. Acute and progressive phases of MS, however, do exhibit disparities in which immune components are involved, with peripheral immune cells predominating in early disease and resident microglia playing a greater role in progressive disease [9,12]. Further disparities exist in the dysregulation of inherent immunosuppressant control mechanisms [13,14], the integrity of the blood–brain barrier (BBB) [15], and the CNS tissue presence of autoreactive T and B lymphocytes as well as monocytes [16,17,18,19]. Acute RRMS exhibits BBB disruption, immune cell infiltration, and glial reactivity across multifocal plaques of axon demyelination that can then undergo a remission phase of repair and remyelination [20]. Conversely, progressive MS (PMS) disease occurs with compartmentalized, chronic inflammatory lesions exhibiting an intact BBB, demyelination, axon injury, and neurodegeneration that is associated with insufficient repair to reverse continued or persistent functional loss [21]. Despite the advances in identifying the cellular and molecular mechanisms underlying disparate forms of MS and the translation of novel therapeutics to reduce the occurrence, duration, and severity of disease relapses, a cure for MS remains elusive.
Transglutaminase-2 (TG2) is a calcium-dependent, protein-crosslinking enzyme [22,23,24,25,26] that is ubiquitously expressed. TG2 possesses a wide variety of activities, from acting as a GTPase and kinase to a transamidase and scaffolding protein with many substrates across various intracellular compartments, the cell surface, and within the extracellular space. These interactions are important for cell survival [27,28] and the phagocytic function of the macrophages in tissue repair [29,30,31], but can also contribute to increased inflammation [32] and cell-death [33] in neurodegenerative conditions [34], including Huntington disease (HD) [35], Alzheimer disease (AD) [36], Amyotrophic Lateral Sclerosis (ALS) [37], and Multiple Sclerosis (MS) [38,39,40,41]. Recently, our group and others identified TG2 dysregulation in cadaveric MS brain samples and in the CNS of rodents with EAE (experimental autoimmune encephalomyelitis) [38], highlighting its role in the pathogenesis of MS. In EAE, TG2 is expressed in infiltrating monocytes [41], promoting their adhesion and tissue migration into the CNS [41]. Sestito et al. [39] demonstrated that this elevated TG2 in the monocytes of MS patients correlated with the progression of the clinical course of disease in MS [39,42], suggesting that TG2 is an important therapeutic target to alter the disease course in MS. Extracellular TG2 also crosslinks with various extracellular matrix (ECM) molecules such as Fibronectin (FN) [43] and Heparin Sulfate Proteoglycan (HSPG) [44], which are known to mediate inflammation and promote tissue scarring, which is inhibitory to repair [45]. Selective deletion of TG2 in astrocytes has been demonstrated to perturb the production of the chondroitin sulfate proteoglycan (CSPG) NG2 after spinal cord injury (SCI) [46] and to improve neuronal survival when these TG2 depleted astrocytes are co-cultured with neurons in vitro following exposure to cytotoxic stimuli [47]. We recently showed elevated expression of endothelial and neuronal TG2 acutely after EAE in mice and rats and in MS brain tissue [38], where TG2 likely contributes to endothelial inflammation and the formation of sclerotic plaques as well as neuron degeneration [38] and apoptosis via cleaved caspase-3 activity [48]. Though the precise cellular role of TG2 in MS remains to be elucidated, global TG2 knockout or TG2 inhibition is associated with perturbed induction and reduced disease severity in EAE.
In the present study, we sought to examine how TG2 expression may be disparately altered in CNS sclerotic plaques from cadaveric RRMS and PMS patients. We also assessed how TG2 was spatially correlated to the localization of proinflammatory mediators, FN, CSPG, HSPG, cPLA2, and COX-2, which are known to functionally interact with, or are regulated by, TG2.

2. Materials and Methods

Human brain specimens: Human post-mortem brain tissue from RRMS (n = 6), PMS (n = 5), or non-MS controls (n = 6) were obtained from The Human Brain and Spinal Fluid Resource Center (VA West Los Angeles Healthcare Center, Los Angeles, CA, USA). The brain tissue specimens were derived from subjects between the ages of 38 and 82 years and included both genders, though a preponderance of male samples did exist, as documented in Table 1. Brain specimens were examined and the presence of typical demyelinating lesions that were consistent with MS were confirmed by a neuropathologist at The Human Brain and Spinal Fluid Resource Center. A waiver from The University of Miami IRB was obtained. The cadaveric tissue samples did not contain any of the 18 identifiers noted in the HIPPA privacy rule.
Tissue processing: Frozen tissue blocks from cadaveric human brains of non-MS (control) specimens and from the two forms of relapsing remitting and progressive MS were fixed by immersion in 4% paraformaldehyde solution (phosphate buffer, pH 7.0) for a period of 48 h at 4 °C, replacing with fresh paraformaldehyde solution at 24 h. Fixed tissues were washed twice with phosphate buffer and dehydrated in 30% sucrose solution and stored at 4 °C prior to cryosectioning. After rapidly embedding the samples in OCT compound (EMS, #62550-01), cryosectioning was performed. Coronal sections of the tissues (20 µm thickness) were cut using a cryostat and mounted on charged glass slides (TruBond 380 Adhesive Microscope Slides, IHC World, IW-T380c). Slides were then dried at room temperature for at least one hour prior to storage at −20 °C until further immunohistological processing.
Reagents: Table 2 shows the list of different primary antibodies, their source, and the dilutions that were employed in the study for immunohistochemistry.
Histology and immunocytochemistry: For visualization of tissue pathology and to identify plaques of demyelination and lesion activity, cryosectioned tissue sections were stained with Hematoxylin, Eosin and Luxol Fast Blue (LFB) [49]. The stage of the lesion was defined as active when both degraded myelin and Iba1+ immune cells were present in the vicinity of the sclerotic plaques. Inactive lesions were characterized by complete demyelination along with an absence of activated immune cells.
For immunohistochemistry, heat antigen retrieval was first performed on the sectioned tissue, as described previously [50], followed by blocking in 2% bovine serum albumin (BSA) in PBS (phosphate-buffered saline) with 0.5% Triton-X100 for an hour at room temperature. Blocked tissue was subjected to specific primary antibody treatment overnight at room temperature. Primary antibodies employed were validated against their specific target proteins in previously published studies or by the manufacturer. Sudan Black was used to block tissue autofluorescence in pathological specimens to eliminate false positive signals by CNS lipids. Antibody immunoreactivity (IR) was visualized using fluorophore conjugated secondary antibodies. Sectioned tissues with only a secondary antibody added were employed as controls to ensure that the fluorescent signal observed was specific to the primary antibodies employed.
Imaging and analysis: Image acquisition of the stained tissue was carried out using a slide scanner or a confocal laser-scanning microscope (Olympus, Fluoview FV 1000, Center Valley, PA, USA). For batch acquisition of high resolution images of the tissue stained with Hematoxylin, Eosin, and Luxol Fast Blue, the images were acquired using the brightfield batch scan function on the Olympus VS120 Fluorescence and Brightfield Slide Scanner for High-Throughput Image Acquisition. Images of immunostained tissue sections were taken using the fluorescent scan function at 20× magnification. The images were imported and processed using OlyVIA software. The fluorescent scans were performed using DAPI, FITC, TRITC, and CY5 filter cubes. The images of each fluorescent channel were exported in grayscale, pseudo-colored, and overlaid to form a multichannel composite image using Adobe Photoshop 22 (Adobe Systems Inc., San Jose, CA, USA). Immunostained images were exported in Big TIFF format and further processed by performing a 2 × 2 bin of each image in Adobe Photoshop 22 (Adobe Systems Inc., San Jose, CA, USA) for converting the files to a TIFF format for the purpose of size reduction, without altering the clarity and the resolution of the images. To maintain image consistency, all images were processed with the same exposure adjustment of +2.80 and offset adjustment of −0.03.
For acquisition of immunofluorescent images using the confocal laser scanning microscope, images were obtained from at least three randomly selected fields from the region of interest (ROI) within the stained tissue sections. Immunofluorescent intensity was measured to determine the antibody reactivity for each of the target proteins and quantified using Image J software (available online: http://imagej.nih.gov/ij/, accessed on 13 December 2019). The sharpness and the tonal range (smart sharpen, 0.9 pixels) of the Tiff files of the images were normalized using Adobe Photoshop CS2 (Adobe Systems Inc., San Jose, CA, USA). All cadaveric samples were imaged and analyzed according to HSB# ID only by an individual who was blinded to the neuropathological identification and classification of the sample as MS or control. Samples obtained from The Human Brain and Spinal Fluid Resource Center (Los Angeles, CA, USA) were randomly provided based upon number of samples requested. Note that the samples provided did contain an unusually high allocation of males for a condition that clinically shows a greater prevalence in females.
Statistical analysis: Quantitative data obtained from measurements of the immunofluorescent intensity of different staining combinations were plotted as the mean ± standard deviation of the mean (SD). Statistical analysis was performed, employing an analysis of variance (ANOVA) followed by a Tukey test for multiple comparisons between the two forms of MS and the non-MS control group using GraphPad Prism v7.0. (GraphPad Software, Inc., La Jolla, CA, USA). Differences were accepted as statistically significant at * p < 0.05, ** p < 0.01, or *** p < 0.0001. To investigate whether TG2 immunoreactivity levels were related to disease progression and/or the stage of the lesion in patients with the two forms of MS, linear regression analyses were performed between TG2 IR of 3 or more randomly selected lesions from each section/patient and the corresponding immunoreactivity of specific immunopathological markers or pathogenic ECM as stated in each figure. The p-value determined by linear regression and Pearson correlation coefficient (r) are indicated in each of the figures.

3. Results

PMS exhibited significantly higher Iba1 immunoreactivity than RRMS, whereas the strong TG2 induction in both forms was comparable. Comparative pathological assessment was conducted on cadaveric cortical brain tissue samples obtained from 17 individuals, 6 with RRMS, 5 with PMS, and 6 non-MS controls. The clinical characteristics of the three groups are presented in Table 1. Randomized imaging of at least three demyelinated plaques and ensuing histological evaluation was performed on tissue sections stained with Hematoxylin, Eosin, and Luxol Fast Blue. Profound demyelinated lesions were observed in the white matter of both RRMS (Figure 1G–I) and PMS (Figure 1M–O) specimens compared to normal appearing myelin in the non-MS controls (Figure 1A–C). The gross pathological examination revealed similar lesion sizes and morphology between both MS forms, though a significant disparity in immune cell density was identified upon additional immunostaining for the marker Iba1. Cellular immunoreactivity for Iba1 was present at demyelinated white matter lesions, with marked density in perivascular tissues surrounding inflamed, strongly TG2-positive blood vessels. The numbers of Iba1+ cells were significantly greater in PMS specimens (Figure 1P–R) than those of RRMS (Figure 1J–L), with these cells around and within the TG2 immunoreactive tissue areas and blood vessels at the sclerotic lesions. The presence of Iba1 would be indicative of either macrophage CNS tissue infiltration or activation and focal migration of reactive microglia to the lesion. The normal-appearing white matter (NAWM) and analogous tissue regions from the non-MS control exhibited only sparse Iba1 immunoreactivity (Figure 1D–F).
Quantitative evaluation of TG2 immunoreactive density within active sclerotic plaques of confocal images revealed no differences in staining between RRMS (Figure 2D,F) and PMS (Figure 2G,I) samples, although the immunofluorescence intensity was significantly greater than that of the non-MS controls (Figure 2A,C). Comparative regions in the non-MS controls had significantly less TG2 expression, with strong but restricted immunoreactivity only in the blood vessels (Figure 2A,C). In the MS specimens, TG2 in the sclerotic plaques was associated with high accumulation of Iba1+ immune cells in perivascular tissue areas (Figure 2E,F,H,I), with less pronounced Iba1 immunoreactivity in the active lesions of RRMS (Figure 2E,F) compared to the active PMS (Figure 2H,I) lesions. The extent of TG2 expression in the sclerotic plaques in both forms of MS was not uniform and the level of its expression appeared to be dependent on the stage of the lesion. Inactive lesions exhibited lower TG2 immunoreactivity than active lesions in both RRMS (Figure 2J,L) and PMS (Figure 2M,O). TG2 staining appeared patchy within blood vessels and perivascular tissues that also exhibited sparse, low intensity cellular Iba1 immunoreactivity (Figure 2K,L,N,O). Linear regression analysis of the correlation between TG2 and Iba1 immunoreactive density within active lesions of RRMS (Figure 2R) and PMS (Figure 2S) samples showed a significant, positive correlation for both forms.
The lesion levels of degraded myelin correlate with TG2 immunoreactivity in both RRMS and PMS. Degraded myelin was identified in tissue sections using an antibody targeted against the immunogen consisting of YGSLPQKSQRSQDENPVV MBP69-86 synthetic peptide, which recognizes degraded MBP [51]. Pronounced degraded myelin was found within sclerotic lesions of both RRMS (Figure 3A–C) and PMS (Figure 3D–F) samples, particularly within perivascular tissues. Inactive lesions displayed much lower levels of degraded myelin immunoreactivity (Figure 3M) for both RRMS (Figure 3G–I) and PMS (Figure 3J–L). Linear regression analysis of the correlation between TG2 and degraded myelin immunoreactive density within active lesions of RRMS (Figure 3N) and PMS (Figure 3O) samples showed a significant, positive correlation for both forms of MS.
Strong, perivascular TG2 immunoreactivity colocalized with endothelial cells (Figure 4) and GFAP-positive astrocytes (Figure 5) in sclerotic lesions of RRMS and PMS. TG2 levels were robustly increased in the disrupted microvasculatures associated with active lesions only in both forms of MS. In blood vessels probed with CD31 (Figure 4B,E,H) or tomato lectin (Figure 4K,N,Q) (Lycopersicon esculentum), characteristic markers of endothelial cells, TG2 IR (Figure 4C,F,I and L,O,R respectively) was significantly enhanced in MS lesions (Figure 4D–I) compared to the controls (Figure 4A–C,J–L), while TG2 IR was not apparent in inactive or old lesions (data not shown). Additionally, dense GFAP immunoreactivity was found to be localized to perivascular tissue in active lesions in RRMS (Figure 5E,F) and PMS (Figure 5H,I), and was significantly increased compared to the non-MS brain tissue control (Figure 5B,C), as measured quantitatively by fluorescent intensity (Figure 5J). TG2 staining showed robust induction and colocalization with GFAP in both RRMS (Figure 5F) and PMS (Figure 5I), implicating an astrocyte contribution to TG2 upregulation in MS. The elevated expression of TG2 in astrocytes was particularly evident in perivascular regions surrounding active lesions in both forms of MS. Linear regression analysis of the correlation between TG2 and GFAP density within active lesions of RRMS (Figure 5K) and PMS (Figure 5L) showed a significant, positive correlation for both forms of MS.
TG2 immunoreactivity exhibits strong overlap with FN within MS lesions of RRMS and PMS. Compared to non-MS control tissue (Figure 6B,C), FN was strongly up regulated in both RRMS (Figure 6E,F) and PMS (Figure 6H,I), as measured quantitatively by fluorescent intensity (Figure 6J). Colocalization of FN was pronounced with TG2, both within the blood vessels as well as in the surrounding perivascular tissue of RRMS (Figure 6D,F) and PMS (Figure 6G,I) lesions. No differences in FN expression or in its colocalization with TG2 were observed between MS forms. Linear regression analysis of the correlation between TG2 and FN density within active lesions of RRMS (Figure 6K) and PMS (Figure 6L) samples showed a significant, positive correlation for both forms of MS.
Perivascular TG2 colocalizes with CSPGs in active brain lesions after MS. Robust and overlapping TG2 and chondroitin sulfate proteoglycan (CSPG), an axon growth inhibitory extracellular matrix, were observed both within the core and the edge of sclerotic plaques of RRMS (Figure 7D–F) and PMS (Figure 7G–I) specimens compared to a low basal level expression of both in non-MS control tissue (Figure 7A,B). Quantitative measurements of fluorescence intensity showed a marked increase in CSPG levels in both forms of MS, with no significant difference between them (Figure 7J). Linear regression analysis of the correlation between TG2 and CSPG density within active lesions of RRMS (Figure 7K) and PMS (Figure 7L) samples showed a significant, positive correlation for both forms of MS.
Increased HSPG immunoreactivity localizes with TG2 in RRMS lesions. Compared to low basal levels of Heparan Sulfate Proteoglycans (HSPG) in non-MS control brain tissue, its immunoreactivity was significantly increased (Figure 8J) in blood vessels and perivascular tissue after RRMS (Figure 8E,F) compared to the lesions in PMS (Figure 8H,I). HSPG colocalized with TG2 after RRMS (Figure 8F) both in the lesion center as well as around the lesion edge compared to the lesions after PMS (Figure 8I), which showed increased HSPG colocalization with TG2 largely in the lesion center while the non-MS control tissue (Figure 8C) only exhibited low levels of HSPG-TG2 colocalization that was limited to the basal lamina around the blood vessels. Linear regression analysis of the correlation between TG2 and HSPG density within active lesions of RRMS (Figure 8K) and PMS (Figure 8L) samples showed a significant, positive correlation for both forms of MS.
PMS, but not RRMS, exhibited a significant increase in inflammatory phospholipase A2, which correlated with TG2 expression. Compared to non-MS controls (Figure 9A–C), increased levels of the proinflammatory mediator cytosolic-phospho-PLA2Ser505 was detected concurrently with elevated TG2 IR in sclerotic plaques of PMS (Figure 9G–I), but not RRMS (Figure 9D–F), brain tissue. Quantitative analysis of phospho-PLA2 IR revealed a significant increase in PMS lesions compared to non-MS controls (Figure 9J). Linear regression analysis of the correlation between TG2 and PLA2Ser505 immunoreactivity within active lesions of PMS (Figure 9L), but not RRMS (Figure 9K), samples showed a significant, positive correlation.
Sclerotic plaques of RRMS and PMS exhibit elevated COX-2 with increased TG2. Cyclooxygenase-2 (COX-2) immunoreactivity was robustly increased in perivascular tissues of PMS lesions (Figure 10H,I) and to a lesser extent in RRMS (Figure 10E,F) compared to the non-MS brain tissue control (Figure 10B,C), as measured quantitatively by fluorescent intensity (Figure 10J). COX-2 immunoreactivity was cellular and likely associated with macrophages and microglia. TG2 staining also showed robust induction in lesions that correlated, but did not colocalize, with COX-2 in both RRMS (Figure 10D–F) and PMS (Figure 10G–I). Linear regression analysis of the correlation between TG2 and COX-2 density within the active lesions of RRMS (Figure 10K) and PMS (Figure 10L) samples showed a significant, positive correlation for both forms of MS.

4. Discussion

Relapsing remitting and progressive forms of MS share the pathological appearance of perivascular, demyelinating lesions within the CNS but exhibit significant differences in vascular, immunological, and neurodegenerative features. Herein we compared the expression of the multifunctional enzyme TG2 in post-mortem MS brain tissues from RRMS and PMS individuals and correlated TG2 changes to the expression of specific markers of vascular inflammation, glial, and immune cell reactivity, and scar-related ECM deposition. TG2 showed strong induction in both RRMS and PMS within the endothelial cells and intraluminal structures of the microvasculature, as well as extracellularly in perivascular tissue regions and reactive glia, particularly astrocytes. The expression of TG2 in macrophages and microglia, however, was limited in both RRMS and PMS. The increased TG2 expression in both forms of MS correlated with the prominent markers of the MS pathology examined, except for pPLA2Ser50 in RRMS. The present study confirms the putative pathogenic role of TG2 in MS across distinct forms of the disease.
Active lesions exhibited strong TG2 expression and were associated with the CNS accumulation of reactive microglia and macrophages in both forms of MS. The density of these immune cells was more pronounced in PMS compared to RRMS. Active lesions also showed the concurrent induction of other inflammatory mediators, including an activated form of cytosolic p-PLA2Ser505 and inducible COX-2, as well as scar-related ECMs such as FN, CSPG, and HSPG. These ECM molecules displayed significant colocalization with TG2, both within the architecture of the blood vessels and the inflamed perivascular CNS tissue, and within hypertrophied astrocytes surrounding the lesions. These proinflammatory molecules have been previously reported to be potential binding partners of TG2 [22,44,52,53] and provide functional interactions that mediate the intercellular signaling associated with the promotion of neuroinflammation and demyelination.
Under normal physiological conditions, enzymes of the PLA2 superfamily are involved in phospholipid metabolism; however, an increased level of cytosolic PLA2 in the CNS has been shown to induce inflammation-mediated demyelination and is a pathophysiological marker of MS [54,55]. TG2 has been reported to interact with PLA2 and is involved in the post-translational modification of this enzyme to enhance its catalytic activity. Increased PLA2 activity increases the mobilization of arachidonic acid (AA) from the membrane phospholipids [52], which is then catalyzed by the enzyme COX-2 to release eicosanoids such as prostaglandins and leukotrienes, which are known proinflammatory lipid mediators. Although low levels of COX-2 are observed constitutively in the normal CNS, where it is associated with synaptic and neurovascular functions [56], its expression is significantly increased in the presence of an inflammatory stimuli [57]. Prostaglandins (PGs) that have been demonstrated to drive an inflammatory response in various neurodegenerative conditions [58], including MS [59], are generated by inducible COX-2. Previous work has reported that a marked increase in COX-2 occurs during the relapse phase of chronic EAE [59], where it is associated with activated macrophages and microglia surrounding the sclerotic lesions [60]. Similarly, robust cellular COX-2 was expressed in the perivascular CNS tissue of PMS lesions that exhibited strong TG2, but was significantly less prevalent in RRMS lesions. The interaction of TG2–PLA2–COX-2 appears therefore to be one of the key signaling mechanisms involved in inflammation and the MS pathophysiology of the progressive disease form. Studies with cytosolic PLA2-deficient mice have shown resistance to EAE induction [55]. Collectively, these findings indicate that the functional coupling between TG2 and cPLA2 could be a critical inducer of MS pathology.
ECM produced by reactive glia have also been identified as important constituents of MS pathology [61]. TG2 within the microvasculature at the perivascular lesion sites colocalizes extracellularly with FN [38,62], where it fulfills a functional role in disease propagation as an immune cell adhesive scaffold. TG2 has a high binding affinity towards FN and cross links FN to confer increased stability and rigidity during the formation of the matrix [63]. Under normal physiological conditions, the presence of FN is limited only to blood vessels [64]. FN, however, is transiently upregulated during tissue injury and binds other ECM or regulates cell migration and proliferation through its interaction with integrin receptors. In MS lesions, FN has been shown to be a major impediment to remyelination repair by inhibiting oligodendrocyte progenitor cell (OPC) maturation [64,65]. The upregulation of FN appears to be a characteristic response in MS lesions and functions to promote leukocyte migration and impair remyelination, irrespective of the type and stage of MS disease. The vascular TG2–FN scaffold sequesters circulating immune cells at the site of BBB breakdown and allows them to then extravasate into surrounding CNS tissue to potentiate damage, demyelination, axon degeneration, and neuronal loss.
Although the active lesions of RRMS and PMS both exhibited strong vascular and perivascular TG2 immunoreactivity, there was a difference in the upregulation of its potential binding partners between MS forms, with a robust presence of HSPG2 in RRMS but not PMS and alternatively strong phospho-PLA2Ser505 in PMS but not as much in RRMS. A linear correlation between the expression levels of TG2 and the activation of cPLA2 was statistically significant only after PMS and not in the RRMS lesions. The functional significance of this disparity in the disease course is unknown but the availability of TG2 and PLA2 knockout mice would allow for examination of whether the removal of one or the other would have a concurrent effect. Genetic ablation of TG2 [66,67] or PLA2 [55] is associated with the antagonism of EAE induction.
Abnormal tissue remodeling and ECM production in the regions of demyelination of the brain and spinal cord after MS have been widely reported in MS [68,69,70]. ECM deposition of regeneration-inhibitory proteoglycans at the edge of the perivascular lesions has been implicated in lesion expansion, scar formation, abortive axon regeneration, and the inhibition of remyelination repair [61,70]. In the examination of RRMS and PMS brain tissues at lesion sites, increased levels of both FN and CSPG colocalize with strong TG2. CSPG deposition around the lesion edge after MS, in both RRMS and PMS, was observed in the current study and has been also reported elsewhere [71]. CSPG is a growth inhibitory substrate produced primarily by reactive astrocytes and other glia surrounding the lesion [72] and is known to perturb OPC migration and thus antagonize remyelination. CSPGs also prevent OPC maturation and thus in MS are a major impediment for repair. Here we have shown that TG2 and CSPG colocalize within the perivascular tissue in both RRMS and PMS. Although there is no evidence of a direct protein–protein interaction between TG2 and CSPGs, recent work has reported a diminution of CSPG production, when TG2 is selectively knocked out in astrocytes following spinal cord injury [46,73] and that such a manipulation leads to improved functional recovery.
Another ECM that is upregulated with MS is perlecan or HSPG2, a subtype of HSPG, which functions as a reservoir for growth factors, including vascular endothelial growth factor (VEGF), transforming growth factor (TGFβ), and platelet-derived growth factor (PDGF), which are critical in intercellular interactions [74]. Perlecan also plays an important role in maintaining the stability and functional integrity of the BBB [75] and has been implicated as a pro-regenerative substrate for damaged axons [76]. In contrast to these supportive actions, HSPGs have also been associated with the inhibition of myelination [77] that would antagonize repair in MS. We found that perlecan colocalizes with TG2 in active lesions after RRMS but not CPMS, suggesting a differential function of TG2–HSPG interactions across MS disease forms. In addition, endothelial TG2 has been reported to interact with HSPGs and prevent heparin sulfate VEGF-induced angiogenesis [78]. Abnormal angiogenesis is a known pathological component of human MS [79] and is associated with the severity of the disease. Whether TG2–perlecan interactions in RRMS are beneficial, contributing to the repair of the BBB or remyelination repair through the release of VEGF or PDGF during the remission phase of the disease, requires further investigation. Similarly, a lack of TG2–HSPG interaction in the active lesions of PMS may be associated with loss of endogenous repair and disease progression, a phenomenon that would need to be substantiated.
In summary, the present work reveals that in different forms of MS, RRMS and PMS, the differential availability of inflammatory and ECM binding partners of TG2 at the sites of perivascular lesions and strong TG2 induction may confer very different pathological roles for TG2 in disease remission or progression. Further examination of these interactions in animal models of disease and the use of genetic knockout strategies will shed light on whether these differences play important roles in the pathological course of either MS disease form or whether such interactions could be a therapeutically targetable direction for peptide-based approaches. Indeed, the peptide inhibition of TG2–FN interactions has been demonstrated to alter TG2-dependent cell-ECM adhesion and spreading [80].

Author Contributions

D.D.P. and M.G. were involved in the concept and experimental design of this study. A.A.M., A.B.H. and M.G. performed the experiments. D.D.P. and M.G. were involved with data analysis and interpretation of the results, and the writing of the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by The Miami Project to Cure Paralysis and The Buoniconti Fund (M.G. and D.D.P.), The John M. and Jocelyn H.K. Watkins Distinguished Chair in Cell Therapies to D.D.P., and Summer Fellowship Funding (Federal Work Study) to A.B.H.

Institutional Review Board Statement

This study does not constitute Human Subject research but involves the use of only cadaveric samples which do not contain any of the 18 identifiers noted in the privacy rule. Therefore, it is not subject to IRB review under 45 CFR 46.

Informed Consent Statement

Not Applicable.

Data Availability Statement

The study did not report any data.

Acknowledgments

We would like to thank the Human Brain and Spinal Fluid Resource Center, VA West Los Angeles Healthcare Center, Los Angeles, CA, for providing the tissue specimen used in this study, which is sponsored by NINDS/NIMH, National Multiple Sclerosis Society, and Department of Veterans Affairs. We want to thank Yan Shi at the Miami Project Imaging Core for her assistance with confocal imaging. We are also grateful to Marcia Boulina for her generosity in sharing the VS120 Slide Scanner housed at the University of Miami Miller School of Medicine Analytical Imaging Core Facility and supported by NIH grant 1S10OD023579-01.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dobson, R.; Giovannoni, G. Multiple sclerosis—A review. Eur. J. Neurol. 2019, 26, 27–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Reich, D.S.; Lucchinetti, C.F.; Calabresi, P.A. Multiple Sclerosis. N. Engl. J. Med. 2018, 378, 169–180. [Google Scholar] [CrossRef] [PubMed]
  3. Miehm, J.D.; Buonaccorsi, J.; Lim, J.; Sato, S.; Rajala, C.; Averill, J.; Khalighinejad, F.; Ionete, C.; Jones, S.L.; Kent, J.A.; et al. Sensorimotor function in progressive multiple sclerosis. Mult. Scler. J. Exp. Transl. Clin. 2020, 6, 2055217320934835. [Google Scholar] [CrossRef] [PubMed]
  4. Racosta, J.M.; Kimpinski, K.; Morrow, S.A.; Kremenchutzky, M. Autonomic dysfunction in multiple sclerosis. Auton. Neurosci. 2015, 193, 1–6. [Google Scholar] [CrossRef]
  5. Abbatemarco, J.R.; Fox, R.J.; Li, H.; Bermel, R.A.; Ontaneda, D. Vitamin D Levels and Visual System Measurements in Progressive Multiple Sclerosis: A Cross-sectional Study. Int. J. MS Care 2021, 23, 53–58. [Google Scholar] [CrossRef]
  6. Ghasemi, N.; Razavi, S.; Nikzad, E. Multiple Sclerosis: Pathogenesis, Symptoms, Diagnoses and Cell-Based Therapy. Cell J. 2017, 19, 1–10. [Google Scholar]
  7. Miller, D.H.; Leary, S.M. Primary-progressive multiple sclerosis. Lancet Neurol. 2007, 6, 903–912. [Google Scholar] [CrossRef]
  8. van Langelaar, J.; Rijvers, L.; Smolders, J.; van Luijn, M.M. B and T Cells Driving Multiple Sclerosis: Identity, Mechanisms and Potential Triggers. Front. Immunol. 2020, 11, 760. [Google Scholar] [CrossRef]
  9. Disano, K.D.; Linzey, M.R.; Royce, D.B.; Pachner, A.R.; Gilli, F. Differential neuro-immune patterns in two clinically relevant murine models of multiple sclerosis. J. Neuroinflammation 2019, 16, 109. [Google Scholar] [CrossRef] [Green Version]
  10. Ransohoff, R.M. Immune-cell crosstalk in multiple sclerosis. Nature 2018, 563, 194–195. [Google Scholar] [CrossRef] [Green Version]
  11. Wu, G.F.; Alvarez, E. The immunopathophysiology of multiple sclerosis. Neurol. Clin. 2011, 29, 257–278. [Google Scholar] [CrossRef] [Green Version]
  12. Hemmer, B.; Kerschensteiner, M.; Korn, T. Role of the innate and adaptive immune responses in the course of multiple sclerosis. Lancet Neurol. 2015, 14, 406–419. [Google Scholar] [CrossRef]
  13. Fletcher, J.M.; Lalor, S.J.; Sweeney, C.M.; Tubridy, N.; Mills, K.H.G. T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin. Exp. Immunol. 2010, 162, 1–11. [Google Scholar] [CrossRef]
  14. Danikowski, K.M.; Jayaraman, S.; Prabhakar, B.S. Regulatory T cells in multiple sclerosis and myasthenia gravis. J. Neuroinflammation 2017, 14, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Minagar, A.; Alexander, J.S. Blood-brain barrier disruption in multiple sclerosis. Mult. Scler. 2003, 9, 540–549. [Google Scholar] [CrossRef] [PubMed]
  16. Arneth, B. Contributions of T cells in multiple sclerosis: What do we currently know? J. Neurol. 2021, 268, 4587–4593. [Google Scholar] [CrossRef]
  17. Lehmann-Horn, K.; Kronsbein, H.C.; Weber, M.S. Targeting B cells in the treatment of multiple sclerosis: Recent advances and remaining challenges. Ther. Adv. Neurol. Disord. 2013, 6, 161–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Luo, C.; Jian, C.; Liao, Y.; Huang, Q.; Wu, Y.; Liu, X.; Zou, D.; Wu, Y. The role of microglia in multiple sclerosis. Neuropsychiatr. Dis. Treat. 2017, 13, 1661–1667. [Google Scholar] [CrossRef] [Green Version]
  19. Voet, S.; Prinz, M.; van Loo, G. Microglia in Central Nervous System Inflammation and Multiple Sclerosis Pathology. Trends. Mol. Med. 2019, 25, 112–123. [Google Scholar] [CrossRef]
  20. Lassmann, H. Pathogenic Mechanisms Associated With Different Clinical Courses of Multiple Sclerosis. Front. Immunol. 2018, 9, 3116. [Google Scholar] [CrossRef]
  21. Correale, J.; Gaitán, M.I.; Ysrraelit, M.C.; Fiol, M.P. Progressive multiple sclerosis: From pathogenic mechanisms to treatment. Brain 2017, 140, 527–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Belkin, A.M. Extracellular TG2: Emerging functions and regulation. FEBS J. 2011, 278, 4704–4716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tatsukawa, H.; Furutani, Y.; Hitomi, K.; Kojima, S. Transglutaminase 2 has opposing roles in the regulation of cellular functions as well as cell growth and death. Cell Death. Dis. 2016, 7, e2244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lorand, L.; Graham, R.M. Transglutaminases: Crosslinking enzymes with pleiotropic functions. Nat. Rev. Mol. Cell Biol. 2003, 4, 140–156. [Google Scholar] [CrossRef] [PubMed]
  25. Bailey, C.D.; Tucholski, J.; Johnson, G.V. Transglutaminases in neurodegenerative disorders. Prog. Exp. Tumor. Res. 2005, 38, 139–157. [Google Scholar] [PubMed]
  26. Martin, A.; De Vivo, G.; Ricotta, M.; Iannuzzi, M.; Gentile, V. Transglutaminases as possible therapeutic targets in neurodegenerative diseases. Recent Pat. CNS Drug Discov. 2010, 5, 195–202. [Google Scholar] [CrossRef]
  27. Eckert, R.L.; Kaartinen, M.T.; Nurminskaya, M.; Belkin, A.M.; Colak, G.; Johnson, G.V.; Mehta, K. Transglutaminase regulation of cell function. Physiol. Rev. 2014, 94, 383–417. [Google Scholar] [CrossRef] [Green Version]
  28. Nurminskaya, M.V.; Belkin, A.M. Cellular functions of tissue transglutaminase. Int. Rev. Cell Mol. Biol. 2012, 294, 1–97. [Google Scholar]
  29. Sestito, C.; Brevé, J.J.; Bol, J.G.; Wilhelmus, M.M.; Drukarch, B.; van Dam, A.-M. Tissue Transglutaminase contributes to myelin phagocytosis in interleukin-4-treated human monocyte-derived macrophages. Cytokine 2020, 128, 155024. [Google Scholar] [CrossRef]
  30. Sun, H.; Kaartinen, M.T. Transglutaminases in Monocytes and Macrophages. Med. Sci. 2018, 6, 115. [Google Scholar] [CrossRef] [Green Version]
  31. Giera, S.; Luo, R.; Ying, Y.; Ackerman, S.D.; Jeong, S.; Stoveken, H.M.; Folts, C.J.; Welsh, C.A.; Tall, G.G.; Stevens, B.; et al. Microglial transglutaminase-2 drives myelination and myelin repair via GPR56/ADGRG1 in oligodendrocyte precursor cells. eLife 2018, 7, e33385. [Google Scholar] [CrossRef] [PubMed]
  32. Ientile, R.; Curro, M.; Caccamo, D. Transglutaminase 2 and neuroinflammation. Amino Acids 2015, 47, 19–26. [Google Scholar] [CrossRef] [PubMed]
  33. Tatsukawa, H.; Hitomi, K. Role of Transglutaminase 2 in Cell Death, Survival, and Fibrosis. Cells 2021, 10, 1842. [Google Scholar] [CrossRef]
  34. Keillor, J.W.; Johnson, G.V.W. Transglutaminase 2 as a therapeutic target for neurological conditions. Expert Opin. Targets 2021, 25, 721–731. [Google Scholar] [CrossRef] [PubMed]
  35. Mastroberardino, P.G.; Piacentini, M. Type 2 transglutaminase in Huntington’s disease: A double-edged sword with clinical potential. J. Intern. Med. 2010, 268, 419–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Wilhelmus, M.M.; de Jager, M.; Bakker, E.N.T.P.; Drukarch, B. Tissue transglutaminase in Alzheimer’s disease: Involvement in pathogenesis and its potential as a therapeutic target. J. Alzheimers Dis. 2014, 42 (Suppl. 3), S289–S303. [Google Scholar] [CrossRef]
  37. Oono, M.; Okado-Matsumoto, A.; Shodai, A.; Ido, A.; Ohta, Y.; Abe, K.; Ayaki, T.; Ito, H.; Takahashi, R.; Taniguchi, N.; et al. Transglutaminase 2 accelerates neuroinflammation in amyotrophic lateral sclerosis through interaction with misfolded superoxide dismutase 1. J. Neurochem. 2014, 128, 403–418. [Google Scholar] [CrossRef] [Green Version]
  38. Pearse, D.D.; Otero, P.A.; Diaz, A.; Pan, X.; Ghosh, M. Neuronal and Endothelial Transglutaminase-2 Expression during Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Neuroscience 2021, 461, 140–154. [Google Scholar] [CrossRef]
  39. Sestito, C.; Leurs, C.E.; Steenwijk, M.D.; Brevé, J.J.; Twisk, J.W.; Wilhelmus, M.M.; Drukarch, B.; Teunissen, C.E.; van Dam, A.-M.; Killestein, J. Tissue Transglutaminase Expression Associates With Progression of Multiple Sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e998. [Google Scholar] [CrossRef]
  40. Chrobok, N.L.; Bol, J.G.J.M.; Jongenelen, C.A.; Brevé, J.J.P.; Alaoui, S.E.; Wilhelmus, M.M.M.; Drukarch, B.; van Dam, A. Characterization of Transglutaminase 2 activity inhibitors in monocytes in vitro and their effect in a mouse model for multiple sclerosis. PLoS ONE 2018, 13, e0196433. [Google Scholar]
  41. Chrobok, N.L.; Bol, J.G.J.M.; Wilhelmus, M.M.M.; Drukarch, B.; Van Dam, A.-M. Tissue Transglutaminase Appears in Monocytes and Macrophages but Not in Lymphocytes in White Matter Multiple Sclerosis Lesions. J. Neuropathol. Exp. Neurol. 2019, 78, 492–500. [Google Scholar] [CrossRef] [PubMed]
  42. Sestito, C.; Brevé, J.J.P.; van Eggermond, M.C.J.A.; Killestein, J.; Teunissen, C.E.; van Rossum, J.; Wilhelmus, M.M.M.; Drukarch, B.; van den Elsen, P.J.; van Dam, A. Monocyte-derived tissue transglutaminase in multiple sclerosis patients: Reflecting an anti-inflammatory status and function of the cells? J. Neuroinflammation 2017, 14, 257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Soluri, M.F.; Boccafoschi, F.; Cotella, D.; Moro, L.; Forestieri, G.; Autiero, I.; Cavallo, L.; Oliva, R.; Griffin, M.; Wang, Z.; et al. Mapping the minimum domain of the fibronectin binding site on transglutaminase 2 (TG2) and its importance in mediating signaling, adhesion, and migration in TG2-expressing cells. FASEB J. 2019, 33, 2327–2342. [Google Scholar] [CrossRef] [Green Version]
  44. Furini, G.; Verderio, E.A.M. Spotlight on the Transglutaminase 2-Heparan Sulfate Interaction. Med. Sci. 2019, 7, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. O’Callaghan, P.; Zhang, X.; Li, J.P. Heparan Sulfate Proteoglycans as Relays of Neuroinflammation. J. Histochem. Cytochem. 2018, 66, 305–319. [Google Scholar] [CrossRef] [Green Version]
  46. Elahi, A.; Emerson, J.; Rudlong, J.; Keillor, J.W.; Salois, G.; Visca, A.; Girardi, P.; Johnson, G.V.; Pröschel, C. Deletion or Inhibition of Astrocytic Transglutaminase 2 Promotes Functional Recovery after Spinal Cord Injury. Cells 2021, 10, 2942. [Google Scholar] [CrossRef] [PubMed]
  47. Yunes-Medina, L.; Paciorkowski, A.; Nuzbrokh, Y.; Johnson, G.V. Depletion of transglutaminase 2 in neurons alters expression of extracellular matrix and signal transduction genes and compromises cell viability. Mol. Cell Neurosci. 2018, 86, 72–80. [Google Scholar] [CrossRef]
  48. Yoo, J.-O.; Lim, Y.-C.; Kim, Y.-M.; Ha, K.-S. Transglutaminase 2 promotes both caspase-dependent and caspase-independent apoptotic cell death via the calpain/Bax protein signaling pathway. J. Biol. Chem. 2012, 287, 14377–14388. [Google Scholar] [CrossRef] [Green Version]
  49. Führmann, T.; Ghosh, M.; Otero, A.; Goss, B.; Dargaville, T.R.; Pearse, D.D.; Dalton, P.D. Peptide-functionalized polymeric nanoparticles for active targeting of damaged tissue in animals with experimental autoimmune encephalomyelitis. Neurosci. Lett. 2015, 602, 126–132. [Google Scholar] [CrossRef]
  50. Ghosh, M.; Xu, Y.; Pearse, D.D. Cyclic AMP is a key regulator of M1 to M2a phenotypic conversion of microglia in the presence of Th2 cytokines. J. Neuroinflammation 2016, 13, 9. [Google Scholar] [CrossRef] [Green Version]
  51. Liu, H.; Shiryaev, S.A.; Chernov, A.V.; Kim, Y.; Shubayev, I.; Remacle, A.G.; Baranovskaya, S.; Golubkov, V.S.; Strongin, A.Y.; Shubayev, V.I. Immunodominant fragments of myelin basic protein initiate T cell-dependent pain. J. Neuroinflammation 2012, 9, 119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Currò, M.; Ferlazzo, N.; Risitano, R.; Condello, S.; Vecchio, M.; Caccamo, D.; Ientile, R. Transglutaminase 2 and phospholipase A(2) interactions in the inflammatory response in human Thp-1 monocytes. Amino Acids 2014, 46, 759–766. [Google Scholar] [CrossRef] [PubMed]
  53. Cardoso, I.; Østerlund, E.C.; Stamnaes, J.; Iversen, R.; Andersen, J.T.; Jørgensen, T.J.D.; Sollid, L.M. Dissecting the interaction between transglutaminase 2 and fibronectin. Amino Acids 2017, 49, 489–500. [Google Scholar] [CrossRef] [Green Version]
  54. Kalyvas, A.; David, S. Cytosolic phospholipase A2 plays a key role in the pathogenesis of multiple sclerosis-like disease. Neuron 2004, 41, 323–335. [Google Scholar] [CrossRef] [Green Version]
  55. Marusic, S.; Leach, M.W.; Pelker, J.W.; Azoitei, M.L.; Uozumi, N.; Cui, J.; Shen, M.W.; Declercq, C.M.; Miyashiro, J.S.; Carito, B.A.; et al. Cytosolic phospholipase A2 alpha-deficient mice are resistant to experimental autoimmune encephalomyelitis. J. Exp. Med. 2005, 202, 841–851. [Google Scholar] [CrossRef] [PubMed]
  56. Yang, H.; Chen, C. Cyclooxygenase-2 in synaptic signaling. Curr. Pharm. Des. 2008, 14, 1443–1451. [Google Scholar] [CrossRef] [Green Version]
  57. Dubois, R.N.; Abramson, S.B.; Crofford, L.; Gupta, R.A.; Simon, L.S.; van de Putte, L.B.; Lipsky, P.E. Cyclooxygenase in biology and disease. FASEB J. 1998, 12, 1063–1073. [Google Scholar] [CrossRef] [Green Version]
  58. Minghetti, L. Cyclooxygenase-2 (COX-2) in inflammatory and degenerative brain diseases. J. Neuropathol. Exp. Neurol. 2004, 63, 901–910. [Google Scholar] [CrossRef] [Green Version]
  59. Ayoub, S.S.; Wood, E.G.; Hassan, S.-U.; Bolton, C. Cyclooxygenase expression and prostaglandin levels in central nervous system tissues during the course of chronic relapsing experimental autoimmune encephalomyelitis (EAE). Inflamm. Res. 2011, 60, 919–928. [Google Scholar] [CrossRef]
  60. Yiangou, Y.; Facer, P.; Durrenberger, P.; Chessell, I.P.; Naylor, A.; Bountra, C.; Banati, R.R.; Anand, P. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006, 6, 12. [Google Scholar] [CrossRef] [Green Version]
  61. Sobel, R.A. The extracellular matrix in multiple sclerosis: An update. Braz. J. Med. Biol. Res. 2001, 34, 603–609. [Google Scholar] [CrossRef] [PubMed]
  62. van Strien, M.E.; Brevé, J.J.P.; Fratantoni, S.; Schreurs, M.W.J.; Bol, J.G.J.M.; Jongenelen, C.A.M.; Drukarch, B.; van Dam, A. Astrocyte-derived tissue transglutaminase interacts with fibronectin: A role in astrocyte adhesion and migration? PLoS ONE 2011, 6, e25037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Nelea, V.; Nakano, Y.; Kaartinen, M.T. Size distribution and molecular associations of plasma fibronectin and fibronectin crosslinked by transglutaminase 2. Protein. J. 2008, 27, 223–233. [Google Scholar] [CrossRef] [PubMed]
  64. Stoffels, J.M.J.; de Jonge, J.C.; Stancic, M.; Nomden, A.; van Strien, M.E.; Ma, D.; Šišková, Z.; Maier, O.; Ffrench-Constant, C.; Franklin, R.J.M.; et al. Fibronectin aggregation in multiple sclerosis lesions impairs remyelination. Brain 2013, 136, 116–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Šišková, Z.; Baron, W.; de Vries, H.; Hoekstra, D. Fibronectin impedes "myelin" sheet-directed flow in oligodendrocytes: A role for a beta 1 integrin-mediated PKC signaling pathway in vesicular trafficking. Mol. Cell Neurosci. 2006, 33, 150–159. [Google Scholar] [CrossRef] [PubMed]
  66. Oh, K.; Park, H.-B.; Seo, M.W.; Byoun, O.-J.; Lee, D.-S. Transglutaminase 2 exacerbates experimental autoimmune encephalomyelitis through positive regulation of encephalitogenic T cell differentiation and inflammation. Clin. Immunol. 2012, 145, 122–132. [Google Scholar] [CrossRef] [PubMed]
  67. van Strien, M.E.; de Vries, H.E.; Chrobok, N.L.; Bol, J.G.J.M.; Breve, J.J.P.; van der Pol, S.M.P.; Kooij, G.; van Buul, J.D.; Karpuj, M.; Steinman, L.; et al. Tissue Transglutaminase contributes to experimental multiple sclerosis pathogenesis and clinical outcome by promoting macrophage migration. Brain Behav. Immun. 2015, 50, 141–154. [Google Scholar] [CrossRef]
  68. de Jong, J.M.; Wang, P.; Oomkens, M.; Baron, W. Remodeling of the interstitial extracellular matrix in white matter multiple sclerosis lesions: Implications for remyelination (failure). J. Neurosci. Res. 2020, 98, 1370–1397. [Google Scholar] [CrossRef] [Green Version]
  69. Yahn, S.L.; Li, J.; Goo, I.; Gao, H.; Brambilla, R.; Lee, J.K. Fibrotic scar after experimental autoimmune encephalomyelitis inhibits oligodendrocyte differentiation. Neurobiol. Dis. 2020, 134, 104674. [Google Scholar] [CrossRef]
  70. Karsdal, M.A.; Nielsen, M.J.; Sand, J.M.; Henriksen, K.; Genovese, F.; Bay-Jensen, A.-C.; Smith, V.; Adamkewicz, J.I.; Christiansen, C.; Leeming, D.J. Extracellular matrix remodeling: The common denominator in connective tissue diseases. Possibilities for evaluation and current understanding of the matrix as more than a passive architecture, but a key player in tissue failure. Assay Drug Dev. Technol. 2013, 11, 70–92. [Google Scholar] [CrossRef] [Green Version]
  71. Lau, L.W.; Keough, M.B.; Haylock-Jacobs, S.; Cua, R.; Döring, A.; Sloka, S.; Stirling, D.P.; Rivest, S.; Yong, V.W. Chondroitin sulfate proteoglycans in demyelinated lesions impair remyelination. Ann. Neurol. 2012, 72, 419–432. [Google Scholar] [CrossRef] [PubMed]
  72. Sobel, R.A.; Ahmed, A.S. White matter extracellular matrix chondroitin sulfate/dermatan sulfate proteoglycans in multiple sclerosis. J. Neuropathol. Exp. Neurol. 2001, 60, 1198–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Monteagudo, A.; Feola, J.; Natola, H.; Ji, C.; Pröschel, C.; Johnson, G.V. Depletion of astrocytic transglutaminase 2 improves injury outcomes. Mol. Cell Neurosci. 2018, 92, 128–136. [Google Scholar] [CrossRef] [PubMed]
  74. Lord, M.S.; Chuang, C.Y.; Melrose, J.; Davies, M.J.; Iozzo, R.V.; Whitelock, J.M. The role of vascular-derived perlecan in modulating cell adhesion, proliferation and growth factor signaling. Matrix Biol. 2014, 35, 112–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Roberts, J.; Kahle, M.P.; Bix, G.J. Perlecan and the blood-brain barrier: Beneficial proteolysis? Front. Pharm. 2012, 3, 155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Shen, Y. Traffic lights for axon growth: Proteoglycans and their neuronal receptors. Neural. Regen. Res. 2014, 9, 356–361. [Google Scholar] [CrossRef] [PubMed]
  77. van Horssen, J.; Bö, L.; Dijkstra, C.D.; de Vries, H.E. Extensive extracellular matrix depositions in active multiple sclerosis lesions. Neurobiol. Dis. 2006, 24, 484–491. [Google Scholar] [CrossRef]
  78. Beckouche, N.; Bignon, M.; Lelarge, V.; Mathivet, T.; Pichol-Thievend, C.; Berndt, S.; Hardouin, J.; Garand, M.; Ardidie-Robouant, C.; Barret, A.; et al. The interaction of heparan sulfate proteoglycans with endothelial transglutaminase-2 limits VEGF165-induced angiogenesis. Sci. Signal. 2015, 8, ra70. [Google Scholar] [CrossRef]
  79. Lengfeld, J.; Cutforth, T.; Agalliu, D. The role of angiogenesis in the pathology of multiple sclerosis. Vasc. Cell 2014, 6, 23. [Google Scholar] [CrossRef] [Green Version]
  80. Hang, J.; Zemskov, E.A.; Lorand, L.; Belkin, A.M. Identification of a novel recognition sequence for fibronectin within the NH2-terminal beta-sandwich domain of tissue transglutaminase. J. Biol. Chem. 2005, 280, 23675–23683. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Comparative gross pathology of cortical lesions after RRMS and PMS. Brain tissue sections from control non-MS (A) and after RRMS (G), and PMS (M) were stained with Hematoxylin, Eosin, and Luxol Fast Blue (A,G,M) to delineate white matter perivascular lesions with demyelination. An adjacent tissue section from each sample was subjected to immunohistochemistry (D,J,P, respectively) using antibodies towards TG2 (Red) and the macrophage-microglia specific marker Iba1(Green). Scale bars = 1000 µm. Close up views of specific regions of H&E/LFB stained section (A,G,M) are shown in (B,C,H,I,N,O), respectively, while close up views of specific regions of Iba1 and TG2 immunostained sections of (D,J,P) are shown in (E,F,K,L,Q,R), respectively. Magnified views of microglia and macrophages can be seen in Iba1+/TG2 double stained brain sections from control (S), RRMS (T), and PMS (U) patients. The control tissue from non-MS patients shows a microglial morphology of a typical resting phenotype that is highly branched and has small cell bodies. Stained section from RRMS patients showed microglial cells with an activated phenotype surrounding active lesions with the cells having an ameboid morphology with larger cell bodies and short processes. Tissue sections from PMS patients showed highly dense regions of activated microglia within and around active lesions exhibiting shorter processes and larger cell bodies. The density of activated microglia and macrophages surrounding active lesions in tissue sections from PMS patients appeared significantly higher than that from RRMS patients.
Figure 1. Comparative gross pathology of cortical lesions after RRMS and PMS. Brain tissue sections from control non-MS (A) and after RRMS (G), and PMS (M) were stained with Hematoxylin, Eosin, and Luxol Fast Blue (A,G,M) to delineate white matter perivascular lesions with demyelination. An adjacent tissue section from each sample was subjected to immunohistochemistry (D,J,P, respectively) using antibodies towards TG2 (Red) and the macrophage-microglia specific marker Iba1(Green). Scale bars = 1000 µm. Close up views of specific regions of H&E/LFB stained section (A,G,M) are shown in (B,C,H,I,N,O), respectively, while close up views of specific regions of Iba1 and TG2 immunostained sections of (D,J,P) are shown in (E,F,K,L,Q,R), respectively. Magnified views of microglia and macrophages can be seen in Iba1+/TG2 double stained brain sections from control (S), RRMS (T), and PMS (U) patients. The control tissue from non-MS patients shows a microglial morphology of a typical resting phenotype that is highly branched and has small cell bodies. Stained section from RRMS patients showed microglial cells with an activated phenotype surrounding active lesions with the cells having an ameboid morphology with larger cell bodies and short processes. Tissue sections from PMS patients showed highly dense regions of activated microglia within and around active lesions exhibiting shorter processes and larger cell bodies. The density of activated microglia and macrophages surrounding active lesions in tissue sections from PMS patients appeared significantly higher than that from RRMS patients.
Biomedicines 10 01241 g001
Figure 2. TG2 immunoreactivity is significantly upregulated in active lesions of MS brain tissue. Compared to non-MS brain tissue (AC), which exhibited low basal levels of TG2 expression (A,C) and a sparse population of Iba1+ immune cells (B), TG2 expression was robustly increased in active lesions of both RRMS (D,F) and PMS (G,I). A dense cellular immunoreactivity for Iba1+ activated microglia and macrophages was also observed in and around the lesions (E,H). TG2 and Iba1-positive immune cell density were both significantly lower in inactive lesions of both RRMS (JL) and PMS (MO). Quantitative assessment of TG2 (P) and Iba1 (Q) immunoreactivity in active lesions of RRMS and PMS brain tissues revealed significantly increased expression levels compared to non-MS brain tissue. A positive linear correlation was observed between TG2 immunoreactivity and the density of Iba1-positive immune cells surrounding active lesions in both RRMS (R) and PMS (S). Scale bar = 40 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05; ** p < 0.01, determined using a 1-way analysis of variance and post hoc Tukey test. The correlation between the immunodensity of Iba1 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (R,S). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 2. TG2 immunoreactivity is significantly upregulated in active lesions of MS brain tissue. Compared to non-MS brain tissue (AC), which exhibited low basal levels of TG2 expression (A,C) and a sparse population of Iba1+ immune cells (B), TG2 expression was robustly increased in active lesions of both RRMS (D,F) and PMS (G,I). A dense cellular immunoreactivity for Iba1+ activated microglia and macrophages was also observed in and around the lesions (E,H). TG2 and Iba1-positive immune cell density were both significantly lower in inactive lesions of both RRMS (JL) and PMS (MO). Quantitative assessment of TG2 (P) and Iba1 (Q) immunoreactivity in active lesions of RRMS and PMS brain tissues revealed significantly increased expression levels compared to non-MS brain tissue. A positive linear correlation was observed between TG2 immunoreactivity and the density of Iba1-positive immune cells surrounding active lesions in both RRMS (R) and PMS (S). Scale bar = 40 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05; ** p < 0.01, determined using a 1-way analysis of variance and post hoc Tukey test. The correlation between the immunodensity of Iba1 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (R,S). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g002
Figure 3. TG2 expression correlates with degraded myelin in active lesions after MS. Active lesions in brain tissues from both RRMS (AC) and PMS (DF) showed elevated levels of TG2 (A,C and D,F) respectively. TG2 correlated with levels of degraded myelin surrounding sclerotic plaques of both forms of MS (B,C and E,F, respectively). Inactive lesions from RRMS (GI) and PMS (JL) exhibited reduced levels of TG2 and degraded myelin. Quantitative assessment of TG2 levels in sclerotic plaques of both RRMS and PMS brain tissue exhibited significant differences in TG2 expression levels between active and inactive lesions (M). Scale bar = 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05; ** p < 0.01, determined using a 1-way analysis of variance and post hoc Tukey test. Degraded myelin immunoreactivity showed a significant, positive correlation with the extent of TG2 expression in lesioned tissue after both RRMS (N) and PMS (O). The correlation between levels of degraded myelin and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (N,O). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 3. TG2 expression correlates with degraded myelin in active lesions after MS. Active lesions in brain tissues from both RRMS (AC) and PMS (DF) showed elevated levels of TG2 (A,C and D,F) respectively. TG2 correlated with levels of degraded myelin surrounding sclerotic plaques of both forms of MS (B,C and E,F, respectively). Inactive lesions from RRMS (GI) and PMS (JL) exhibited reduced levels of TG2 and degraded myelin. Quantitative assessment of TG2 levels in sclerotic plaques of both RRMS and PMS brain tissue exhibited significant differences in TG2 expression levels between active and inactive lesions (M). Scale bar = 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05; ** p < 0.01, determined using a 1-way analysis of variance and post hoc Tukey test. Degraded myelin immunoreactivity showed a significant, positive correlation with the extent of TG2 expression in lesioned tissue after both RRMS (N) and PMS (O). The correlation between levels of degraded myelin and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (N,O). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g003
Figure 4. TG2 immunoreactivity is robustly upregulated in endothelial cells within active lesions of MS brain tissue. Compared to non-MS brain tissue (Top panel; AC and Bottom panel; JL), which showed low levels of basal TG2 expression (A,C,J,L) in the endothelial cells labelled with CD31 (Top panel, B,E,H) or tomato lectin (Bottom panel, K,N,Q), TG2 expression was robustly increased in the active lesions of both forms of MS; RRMS (Top panel D,F and Bottom panel M,O) and PMS (Top panel, G,I and Bottom panel P,R). Scale bar = 50 µm.
Figure 4. TG2 immunoreactivity is robustly upregulated in endothelial cells within active lesions of MS brain tissue. Compared to non-MS brain tissue (Top panel; AC and Bottom panel; JL), which showed low levels of basal TG2 expression (A,C,J,L) in the endothelial cells labelled with CD31 (Top panel, B,E,H) or tomato lectin (Bottom panel, K,N,Q), TG2 expression was robustly increased in the active lesions of both forms of MS; RRMS (Top panel D,F and Bottom panel M,O) and PMS (Top panel, G,I and Bottom panel P,R). Scale bar = 50 µm.
Biomedicines 10 01241 g004
Figure 5. TG2 expressing sclerotic lesions are surrounded by reactive astrocytes. Compared to non-MS brain tissue (AC), sclerotic lesions with elevated levels of TG2 showed an increase in reactive astrocytic marker glial fibrillary acidic protein (GFAP, red) surrounding the plaques in brain tissue after both RRMS (DF) and PMS (GI). The edge of the sclerotic plaques showed overlapping of GFAP and TG2 (green) immunoreactivity. Scale bar represents 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance is indicated by a ** p < 0.001; *** p < 0.0001, determined using a 1-way analysis of variance and post hoc Tukey test. Quantitative measurement of total fluorescence intensity indicated a significant increase in GFAP immunoreactivity in both RRMS (n = 6) and PMS (n = 5) sclerotic plaques (J) compared to non-MS brain tissue (n = 6). GFAP immunoreactivity in lesioned tissue showed a significant positive correlation with TG2 expression in active lesions of both RRMS (K) and PMS (L). The correlation between GFAP+ expression and TG2 IR in lesions from all patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 5. TG2 expressing sclerotic lesions are surrounded by reactive astrocytes. Compared to non-MS brain tissue (AC), sclerotic lesions with elevated levels of TG2 showed an increase in reactive astrocytic marker glial fibrillary acidic protein (GFAP, red) surrounding the plaques in brain tissue after both RRMS (DF) and PMS (GI). The edge of the sclerotic plaques showed overlapping of GFAP and TG2 (green) immunoreactivity. Scale bar represents 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance is indicated by a ** p < 0.001; *** p < 0.0001, determined using a 1-way analysis of variance and post hoc Tukey test. Quantitative measurement of total fluorescence intensity indicated a significant increase in GFAP immunoreactivity in both RRMS (n = 6) and PMS (n = 5) sclerotic plaques (J) compared to non-MS brain tissue (n = 6). GFAP immunoreactivity in lesioned tissue showed a significant positive correlation with TG2 expression in active lesions of both RRMS (K) and PMS (L). The correlation between GFAP+ expression and TG2 IR in lesions from all patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g005
Figure 6. Comparative assessment of TG2 and FN co-expression in normal and MS brain tissues. The expression levels of FN and TG2 in sclerotic plaques of brain tissue after RRMS (n = 6) and PMS (n = 5) were analyzed and compared to non-MS brain tissue (n = 6) using double fluorescent immunohistochemistry. TG2 (green) and FN (red) colocalized (yellow) around blood vessels. Representative images of active lesions from demyelinated regions of tissue samples from the two forms of MS were acquired and compared to non-MS tissue samples (AC). Elevated levels of FN immunoreactivity overlapped in regions that exhibited increased TG2 expression, which was more abundant in the active lesion core after both RRMS (DF) and PMS (GI). Baseline expression levels of FN in non-MS brain tissue were significantly lower (J). Scale bar = 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance is indicated by a ** p < 0.001; *** p < 0.0001, determined using a 1-way analysis of variance and post hoc Tukey test. FN expression showed a significant positive correlation to the extent of TG2 IR in active lesions after both RRMS (K) and PMS (L). The correlation between FN expression and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 6. Comparative assessment of TG2 and FN co-expression in normal and MS brain tissues. The expression levels of FN and TG2 in sclerotic plaques of brain tissue after RRMS (n = 6) and PMS (n = 5) were analyzed and compared to non-MS brain tissue (n = 6) using double fluorescent immunohistochemistry. TG2 (green) and FN (red) colocalized (yellow) around blood vessels. Representative images of active lesions from demyelinated regions of tissue samples from the two forms of MS were acquired and compared to non-MS tissue samples (AC). Elevated levels of FN immunoreactivity overlapped in regions that exhibited increased TG2 expression, which was more abundant in the active lesion core after both RRMS (DF) and PMS (GI). Baseline expression levels of FN in non-MS brain tissue were significantly lower (J). Scale bar = 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance is indicated by a ** p < 0.001; *** p < 0.0001, determined using a 1-way analysis of variance and post hoc Tukey test. FN expression showed a significant positive correlation to the extent of TG2 IR in active lesions after both RRMS (K) and PMS (L). The correlation between FN expression and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g006
Figure 7. CSPG levels were upregulated with increased TG2 IR within active sclerotic lesions of both RRMS and PMS brain tissues. TG2 immunofluorescence in control, non-MS brain tissue exhibited restricted basal expression (A), which was largely found within blood vessels and overlapped with very low levels of CSPG immunoreactivity (CS56 antibody, B,C). Conversely, there was robust and overlapping accumulation of TG2 and CSPG, both within the core and the edge of sclerotic plaques in brain tissue from both RRMS (DF) and PMS (GI). High-magnification image scale bar represents 50 µm. Quantitative measurement of total fluorescence intensity of CS-56 immunoreactivity indicated a significant increase in the levels of CSPG in both RRMS (n = 6) and PMS (n = 5) sclerotic plaques (J) compared to the low expression levels measured in non-MS brain tissue (n = 6). No significant difference was observed in CSPG immunoreactivity within the active sclerotic lesions between RRMS and PMS. Statistical significance is indicated by a ** p < 0.01; determined using a 1-way analysis of variance and post hoc Tukey test. Error bars are mean ± SD. CSPG expression showed a significant positive correlation with TG2 IR in active lesions after both RRMS (K) and PMS (L). The correlation between the expression levels of CS-56 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 7. CSPG levels were upregulated with increased TG2 IR within active sclerotic lesions of both RRMS and PMS brain tissues. TG2 immunofluorescence in control, non-MS brain tissue exhibited restricted basal expression (A), which was largely found within blood vessels and overlapped with very low levels of CSPG immunoreactivity (CS56 antibody, B,C). Conversely, there was robust and overlapping accumulation of TG2 and CSPG, both within the core and the edge of sclerotic plaques in brain tissue from both RRMS (DF) and PMS (GI). High-magnification image scale bar represents 50 µm. Quantitative measurement of total fluorescence intensity of CS-56 immunoreactivity indicated a significant increase in the levels of CSPG in both RRMS (n = 6) and PMS (n = 5) sclerotic plaques (J) compared to the low expression levels measured in non-MS brain tissue (n = 6). No significant difference was observed in CSPG immunoreactivity within the active sclerotic lesions between RRMS and PMS. Statistical significance is indicated by a ** p < 0.01; determined using a 1-way analysis of variance and post hoc Tukey test. Error bars are mean ± SD. CSPG expression showed a significant positive correlation with TG2 IR in active lesions after both RRMS (K) and PMS (L). The correlation between the expression levels of CS-56 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g007
Figure 8. TG2 and HSPG immunoreactivity overlap in sclerotic lesions after RRMS. In contrast to non-MS control brain tissue, which exhibited lower expression levels of TG2 and HSPG in the basal lamina of the blood vessels (AC), sclerotic lesions after RRMS (DF) show pronounced and overlapping reactivity of HSPG and TG2 immunofluorescence both in the center and the edge of the sclerotic lesions. The extent of HSPG expression in lesions after PMS was not as robustly increased (GI) compared to the RRMS lesions and was mostly limited to the lesion center. Quantitative assessment of HSPG immunoreactivity from active lesions of the two forms of MS indicated significant upregulation in the levels of HSPG only after RRMS and not PMS compared to non-MS controls (J). Scale bar = 30 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05, determined using a 1-way analysis of variance and post hoc Tukey test. The extent of HSPG expression was proportional to the lesion associated TG2 levels and showed a significant positive correlation to the intensity of TG2 IR in sclerotic lesions after both RRMS (L) and CPMS (K). The correlation between the levels of HSPG and TG2 in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 8. TG2 and HSPG immunoreactivity overlap in sclerotic lesions after RRMS. In contrast to non-MS control brain tissue, which exhibited lower expression levels of TG2 and HSPG in the basal lamina of the blood vessels (AC), sclerotic lesions after RRMS (DF) show pronounced and overlapping reactivity of HSPG and TG2 immunofluorescence both in the center and the edge of the sclerotic lesions. The extent of HSPG expression in lesions after PMS was not as robustly increased (GI) compared to the RRMS lesions and was mostly limited to the lesion center. Quantitative assessment of HSPG immunoreactivity from active lesions of the two forms of MS indicated significant upregulation in the levels of HSPG only after RRMS and not PMS compared to non-MS controls (J). Scale bar = 30 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05, determined using a 1-way analysis of variance and post hoc Tukey test. The extent of HSPG expression was proportional to the lesion associated TG2 levels and showed a significant positive correlation to the intensity of TG2 IR in sclerotic lesions after both RRMS (L) and CPMS (K). The correlation between the levels of HSPG and TG2 in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g008
Figure 9. Lesions show increased phospho-PLA2Ser505 immunoreactivity in brain tissue with TG2 after PMS. pPLA2Ser505 levels were elevated with an increase in the expression of TG2 in active lesions after PMS. Most of the PLA2 immunoreactivity was found in the lesion edge, while being absent in the lesion core. The non-MS control brain tissue (AC) did not show pPLA2Ser50 IR (B,C) compared to brain tissue after RRMS (DF), which showed only modest increase in the levels of pPLA2Ser50 (E,F) compared to the control tissue. Sclerotic lesions after PMS (GI) exhibited a robust increase in pPLA2Ser50 (H,I) expression. Quantitative assessment of pPLA2Ser50 immunoreactivity indicated significant upregulation in the levels of pPLA2Ser50 only after PMS (J). Scale bar = 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05, determined using a 1-way analysis of variance and post hoc Tukey test. pPLA2Ser50 expression showed a significant positive correlation with TG2 IR in active lesions only after PMS (L) and not after RRMS (K). The correlation between the expression levels of pPLA2Ser50 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 9. Lesions show increased phospho-PLA2Ser505 immunoreactivity in brain tissue with TG2 after PMS. pPLA2Ser505 levels were elevated with an increase in the expression of TG2 in active lesions after PMS. Most of the PLA2 immunoreactivity was found in the lesion edge, while being absent in the lesion core. The non-MS control brain tissue (AC) did not show pPLA2Ser50 IR (B,C) compared to brain tissue after RRMS (DF), which showed only modest increase in the levels of pPLA2Ser50 (E,F) compared to the control tissue. Sclerotic lesions after PMS (GI) exhibited a robust increase in pPLA2Ser50 (H,I) expression. Quantitative assessment of pPLA2Ser50 immunoreactivity indicated significant upregulation in the levels of pPLA2Ser50 only after PMS (J). Scale bar = 50 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05, determined using a 1-way analysis of variance and post hoc Tukey test. pPLA2Ser50 expression showed a significant positive correlation with TG2 IR in active lesions only after PMS (L) and not after RRMS (K). The correlation between the expression levels of pPLA2Ser50 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g009
Figure 10. MS lesions show increased COX-2 immunoreactivity with TG2 in brain tissue after MS. Inducible COX-2 (red) levels were elevated in sclerotic lesions associated with TG2 (green) after MS. The non-MS control brain tissue (AC) exhibited low, basal levels of COX-2 IR (B,C) compared to affected regions of brain tissue after RRMS (DF), which showed a modest increase in COX-2 that was proportional to the level of the cellular density around the active lesion (E,F). Sclerotic lesions in PMS (GI) exhibited a robust COX-2 expression (H,I) that was similarly localized but much higher in density (H). Quantitative assessment of COX-2 IR indicated significant upregulation only after PMS (J). Scale bar = 40 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05, *** p < 0.0001 determined using a 1-way analysis of variance and post hoc Tukey test. COX-2 expression showed a significant positive correlation to the extent of TG2 IR in active lesions after both RRMS (K) and PMS (L). The correlation between the expression levels of COX2 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Figure 10. MS lesions show increased COX-2 immunoreactivity with TG2 in brain tissue after MS. Inducible COX-2 (red) levels were elevated in sclerotic lesions associated with TG2 (green) after MS. The non-MS control brain tissue (AC) exhibited low, basal levels of COX-2 IR (B,C) compared to affected regions of brain tissue after RRMS (DF), which showed a modest increase in COX-2 that was proportional to the level of the cellular density around the active lesion (E,F). Sclerotic lesions in PMS (GI) exhibited a robust COX-2 expression (H,I) that was similarly localized but much higher in density (H). Quantitative assessment of COX-2 IR indicated significant upregulation only after PMS (J). Scale bar = 40 µm. Data were quantified and expressed as value of the mean plus the standard deviation (SD). Statistical significance indicated a * p < 0.05, *** p < 0.0001 determined using a 1-way analysis of variance and post hoc Tukey test. COX-2 expression showed a significant positive correlation to the extent of TG2 IR in active lesions after both RRMS (K) and PMS (L). The correlation between the expression levels of COX2 and TG2 IR in the lesions from all the patients in each of the two forms of MS is shown in (K,L). The p-value determined by linear regression and Pearson correlation coefficient (r) is indicated for each of the analyses.
Biomedicines 10 01241 g010
Table 1. Source of tissue specimens from control and multiple sclerosis patients employed in the study.
Table 1. Source of tissue specimens from control and multiple sclerosis patients employed in the study.
HSB#
Patient ID
Age (years)GenderMicroscopic Description of Lesion from the Neuropathological ReportPost-Mortem Interval (hours)Clinical Diagnosis
342262MaleSection of periventricular white matter showed demyelination characterized by a variably decreased axonal density (80–100%) and complete demyelination. There was severe associated gliosis and oligodendrocyte loss but no associated macrophage activity or perivascular lymphocytic cuffing.
Presence of chronic MS plaque formation.
11.75MS/RRMS
389153MaleSection of periventricular white matter showed plaque formation with up to 100% axonal loss and 100% demyelination, near complete loss of oligodendrocytes, and associated gliosis was prominent along with scattered evidence of macrophage activity and perivascular lymphocytic cuffing
Presence of chronic/active multiple sclerosis plaque formation and active multiple sclerosis plaque formation.
25.3MS/CPMS, depression, post-traumatic stress disorder, degenerative spine disease
505367MaleSection of periventricular white matter showed plaque formation with up to 100% axonal loss and 100% demyelination There was near complete oligodendrocyte loss and prominent associated gliosis. There was no evidence of macrophage activity or perivascular lymphocytic cuffing
Evidence of chronic multiple sclerosis plaque formation.
24.9MS/secondary progressive MS, migraine, history of stroke, transient ischemic attack (TIA), Chronic Obstructive Pulmonary Disease (COPD), pneumonia, aspiration, meningitis, ataxia, chronic UTI
509560MaleSection of periventricular white matter showed irregular but fairly well-defined area of extensive myelin loss and relative preservation of axons with scant perivascular mononuclear infiltrates. There was associated gliosis at the periphery of the lesion.8.1MS, RRMS, sclerosis, heart attack
510260FemaleSection of periventricular white matter showed fairly well-defined area of demyelination and relative preservation of axons with scattered macrophages.14.7MS, RRMS, depression, celiac disease, headache. Neurogenic bladder, chronic UTI, osteoporosis, insomnia
512379FemaleSection of periventricular white matter displayed somewhat ill-defined areas of myelin pallor with relative axonal preservation.24.8MS/RRMS, congestive heart failure, neuralgia, trigeminal, asthma
513981MaleSection of periventricular white matter showed myelin loss with relative axonal preservation consistent with chronic inactive plaque of multiple sclerosis.
Evidence of demyelinating lesions consistent with clinical history of multiple sclerosis.
36.8MS/RRMS, dysphagia, paraplegic, neurogenic bladder, diabetes type 11, hypertension, hyperlipidemia, myocardial infarction (Ml), atherosclerosis
515463MaleSection of periventricular white matter showed irregular but fairly well-defined area of extensive myelin loss and relative preservation of axons with rare perivascular mononuclear infiltrates. There was associated mild gliosis at the periphery of the lesion.12.6MS/RRMS, optic neuritis
516082MaleSections of periventricular white matter showed plaque formation with up to 100% axonal loss and 100% demyelination with near-complete oligodendrocyte loss and associated gliosis. There was no evidence of macrophage activity or perivascular lymphocytic cuffing.
Chronic multiple sclerosis plaque formation.
9.8MS/RRMS, stroke/CVA, pneumonia, aspiration, MRSA (methicillin-resistant Staphylococcus aureus), chronic UTI
517052FemaleSections from the lateral angle of the lateral ventricle showed an old demyelination plaque surrounded by hypercellular white matter.
Presence of old demyelination plaque surrounded by hypercellular white matter.
25.0MS, CPMS, optic neuritis, depression
527038FemaleOld demyelination plaques were seen in the sections of posterior cingulate gyrus and pons.
Presence of old demyelination plaques.
17.5MS, CPMS, asthma, depression, pain, paraplegic, ataxia (cerebellar)
430784MaleNormal Appearing White Matter (NAWM)11.8CA, stomach, renal failure, acute, COPD (control, non-MS)
430870MaleNAWM11.8Coronary heart disease, leukemia, diabetes type I, myocardial infarction (control, non-MS)
429480MaleNAWM19.2CA, pancreas, hypertension (control, non-MS)
463159MaleNAWM20.2COPD, pulmonary emphysema, congestive heart failure, tobacco abuse, atrial fibrillation, hypertension (control, non-MS)
507283MaleNAWM19.5COPD, seizure disorder (clinical only), atrial fibrillation (control, non-MS)
461549MaleNAWM15CA, colon with metastasis to liver, depression (control, non-MS)
The demographic data were obtained from The Human Brain and Spinal Fluid Resource Center (Los Angeles, CA).
Table 2. Primary antibodies used for immunohistochemistry.
Table 2. Primary antibodies used for immunohistochemistry.
Primary AntibodyManufacturerCatalog NumberAntibody HostDilution Used
TGM2 pAb (CUB 7402)Thermo FisherMA5-12739Mouse1:100
Rb pAb to Transglutaminase-2Abcamab421Rabbit1:100
Anti IBA1, Rabbit (for ICC)Wako/Fuji019-19741Rabbit1:1000
Chicken Polyclonal to IBA1EncorCPCA-IBA1Chicken1:1000
Goat pAb to IBA1Abcamab5076Goat1:500
Myelin Basic ProteinEncorCPCA-MBPChicken1:2500
Degraded Myelin Basic ProteinMilliporeAB5864Rabbit1:1000
Glial Fibrillary Acidic ProteinDakoZ0334Rabbit1:500
Anti-Glial Fibrillary Acidic ProteinMilliporeAB5541Chicken1:250
Anti-Fibronectin antibodySigma F3648Rabbit1:200
Monoclonal Anti-Chondroitin Sulfate (Clone CS-56)SigmaC8035 (SAB4200696)Mouse1:200
Anti-Heparan Sulfate Proteoglycan, (Perlecan), clone 5D7-2E4SigmaMABT 12Mouse1:200
Anti-phospho-c-PLA2 (pSer505)SigmaSAB4503812 Rabbit1:100
Anti-COX-2/Cyclooxygenase 2Abcamab15191Rabbit1:100
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pearse, D.D.; Hefley, A.B.; Morales, A.A.; Ghosh, M. Comparative Profiling of TG2 and Its Effectors in Human Relapsing Remitting and Progressive Multiple Sclerosis. Biomedicines 2022, 10, 1241. https://doi.org/10.3390/biomedicines10061241

AMA Style

Pearse DD, Hefley AB, Morales AA, Ghosh M. Comparative Profiling of TG2 and Its Effectors in Human Relapsing Remitting and Progressive Multiple Sclerosis. Biomedicines. 2022; 10(6):1241. https://doi.org/10.3390/biomedicines10061241

Chicago/Turabian Style

Pearse, Damien D., Andrew B. Hefley, Alejo A. Morales, and Mousumi Ghosh. 2022. "Comparative Profiling of TG2 and Its Effectors in Human Relapsing Remitting and Progressive Multiple Sclerosis" Biomedicines 10, no. 6: 1241. https://doi.org/10.3390/biomedicines10061241

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