26
|
Romano E, Rosa I, Fioretto BS, Matucci-Cerinic M, Manetti M. Circulating Neurovascular Guidance Molecules and Their Relationship with Peripheral Microvascular Impairment in Systemic Sclerosis. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071056. [PMID: 35888144 PMCID: PMC9316343 DOI: 10.3390/life12071056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex connective tissue disease whose earliest clinical manifestations are microvascular tone dysregulation and peripheral microcirculatory abnormalities. Following previous evidence of an association between circulating neurovascular guidance molecules and SSc disturbed angiogenesis, here, we measured the levels of soluble neuropilin 1 (sNRP1), semaphorin 3E (Sema3E), and Slit2 by enzyme-linked immunosorbent assay in serum samples from a large case series of 166 SSc patients vs. 110 healthy controls. We focused on their possible correlation with vascular disease clinical features and applied logistic regression analysis to determine which of them could better reflect disease activity and severity. Our results demonstrate that, in SSc: (i) sNRP1 is significantly decreased, with lower sNRP1 serum levels correlating with the severity of nailfold videocapillaroscopy (NVC) abnormalities and the presence of ischemic digital ulcers (DUs); (ii) both Sema3E and Slit2 are increased, with Sema3E better reflecting early NVC abnormalities; and (iii) higher Sema3E correlates with the absence of DUs, while augmented Slit2 associates with the presence of DUs. Receiver operator characteristics curve analysis revealed that both circulating sNRP1 and Sema3E show a moderate diagnostic accuracy. Moreover, logistic regression analysis allowed to identify sNRP1 and Sema3E as more suitable independent biomarkers reflecting the activity and severity of SSc-related peripheral microvasculopathy.
Collapse
|
27
|
Russo E, Carboni D, Baldi S, Fioretto B, Romano E, El Aoufy K, Ramazzotti M, Rosa I, Lepri G, DI Gloria L, Bruni C, Melchiorre D, Guiducci S, Manetti M, Matucci-Cerinic M, Amedei A, Bellando Randone S. POS0487 THE CROSSTALK OF THE SKIN-ORAL-GUT MICROBIOME AXIS IN LIMITED AND DIFFUSE CUTANEOUS SYSTEMIC SCLEROSIS. Ann Rheum Dis 2022. [DOI: 10.1136/annrheumdis-2022-eular.4630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BackgroundSystemic sclerosis (SSc) is a complex and still unclear rare disease. Microbiota has recently emerged as an important environmental factor in SSc pathogenesis, either at gut, oral and skin level.ObjectivesTo investigate the role of microbiota in SSc subsets, focusing on the skin-oral-gut microbiota axis and serum and fecal free fatty acids (FFA) profile.MethodsTwenty-six consecutive SSc patients (22 females) (mean disease duration (SD): 13 ± 6.91 yrs), classified according to the ACR/EULAR2013 criteria, were enrolled. Demographic, clinical and laboratory data were recorded. Gastrointestinal symptoms were investigated with UCLA GIT-2.0-questionnaire. Fecal, unstimulated saliva and superficial epidermal samples were collected. Microbiota was assessed through 16S ribosomal RNA Next Generation gene-sequencing analysis. Gas Cromatography-Mass Spettroscopy was used to measure FFAs in serum and fecal samples.ResultsThirteen patients had limited cutaneous SSc (lcSSc), 13 diffuse cutaneous (dcSSc). The two subsets displayed a different cutaneous and fecal microbiota profile. In detail, the class of cutaneous Sphingobacteria was significantly higher in lcSSc (p<0.05), while the phylum of Lentisphaerae, the family of Victivallaceae and the genus of Victivallis were significantly higher in fecal samples of lcSSc (all p<0.05). A significant increase of fecal propionic acid was observed in lcSSc patients (p<0.05). Moreover, all fecal medium-chain FAs and hexanoic acids were significantly higher in lcSSc (p<0.05 and p<0.001, respectively). The analysis of serum FFAs levels showed an increase of valeric and octanoic acids in lcSSc (both p<0.05). A negative correlation between UCLA-GIT-2.0 total score and fecal octanoic acid (rho=-0.61; p=0.03), and a positive correlation with serum propionic acid (rho=0.55; p=0.05) was found in lcSSc.ConclusionOur findings show a different microbiota signature in the skin and gut, and a different FFAs profile in lcSSc and dcSSc. Such a differential regulation of microbiota composition and bacterial metabolite production suggests different dynamics of skin-oral-gut microbiota axis in SSc subsets. This data could be useful to develop personalized therapies targeting gastrointestinal and skin involvement.Disclosure of InterestsNone declared.
Collapse
|
28
|
Romano E, Rosa I, Fioretto BS, Giuggioli D, Manetti M, Matucci-Cerinic M. AB0132 STIMULATION OF SOLUBLE GUANYLATE CYCLASE (sGC) FOSTERS ANGIOGENESIS AND BLUNTS ENDOTHELIAL-TO-MESENCHYMAL TRANSITION (EndoMT) OF SYSTEMIC SCLEROSIS (SSc) DERMAL MICROVASCULAR ENDOTHELIAL CELLS. Ann Rheum Dis 2022. [DOI: 10.1136/annrheumdis-2022-eular.2362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BackgroundIn SSc, early abnormalities in microvessel morphology and angiogenic impairment in parallel advance with the development of tissue fibrosis orchestrated by myofibroblasts. Increasing evidence suggests that the EndoMT process, in which endothelial cells transdifferentiate into profibrotic myofibroblasts, may take centre stage in SSc pathogenesis [1,2]. sGC is an enzyme regulating cell growth/proliferation and vascular tone/remodelling by catalysing the production of cyclic guanosine monophosphate. Previous studies reported that sGC stimulation inhibits TGFβ-induced fibroblast-to-myofibroblast differentiation and collagen synthesis by blocking non-canonical ERK-dependent TGFβ signalling, and that sCG stimulators (sGCS) may exert antifibrotic effects in experimental models of fibrotic disorders.ObjectivesTo investigate the possible modulatory effects of sGC stimulation on impaired angiogenesis and EndoMT of SSc dermal microvascular endothelial cells (SSc-MVECs).MethodsTo evaluate the effects of treatment with sGCS on endothelial cell viability/proliferation, 5 lines of SSc-MVECs and 5 lines of healthy dermal MVECs (H-MVECs) were challenged with sGCS (here MK-2947) and assayed with both annexin V/PI flow cytometry and WST-1. To analyse the modulation of angiogenesis by sGCS, SSc-MVECs were challenged with MK-2947 and subsequently tested for wound healing and capillary-like tube formation capabilities. To study the effects of MK-2947 on EndoMT, the same cells were assayed for the expression of endothelial and mesenchymal/myofibroblast markers by quantitative real-time PCR, western blotting and immunofluorescence, as well as for their contractile ability by collagen gel contraction assay. Phosphorylation of ERK1/2 was assessed by western blotting.ResultsTreatment with MK-2947 did not affect viability/proliferation of H-MVECs, while it significantly increased the proliferation of SSc-MVECs (p<0.001 vs. basal). Compared to basal condition, the MK-2947 challenge ameliorated both wound healing capability (p<0.001) and angiogenic performance (number of nodes: p<0.01; segments: p<0.001; meshes: p<0.01; and junctions: p<0.001) of SSc-MVECs. Upon stimulation of sGC, SSc-MVECs exhibited increased gene expression of proangiogenic matrix metalloproteinase (MMP)-9 (p<0.05) and decreased expression of both antiangiogenic MMP-12 (p<0.05) and pentraxin-3 (p<0.001) respect to basal SSc-MVECs. A significant increase in both gene and protein expression of the endothelial markers CD31 and VE-cadherin, and a parallel decrease in the expression of the mesenchymal/myofibroblast markers α-SMA, S100A4, and type I collagen were found in MK-2947-treated SSc-MVECs. MK-2947 also downregulated the EndoMT-driving transcription factor SNAIL1 in SSc-MVECs. Stimulation with MK-2947 was able to significantly counteract the intrinsic ability of myofibroblast-like SSc-MVECs to contract collagen gels (p<0.001) and effectively reduce phosphorylated-ERK1/2 protein levels (p<0.01) respect to basal cells.ConclusionStimulation of sGC effectively ameliorates the angiogenic performance and blunts the pathogenic myofibroblast-like profibrotic phenotype of SSc-MVECs.References[1]Manetti M, et al. Endothelial-to-mesenchymal transition contributes to endothelial dysfunction and dermal fibrosis in systemic sclerosis. Ann Rheum Dis. 2017;76:924–34.[2]Romano E, et al. New insights into profibrotic myofibroblast formation in systemic sclerosis: when the vascular wall becomes the enemy. Life (Basel). 2021;11:610.Disclosure of InterestsEloisa Romano: None declared, Irene Rosa: None declared, Bianca Saveria Fioretto: None declared, Dilia Giuggioli: None declared, Mirko Manetti Speakers bureau: has received consulting fees or honorarium from MSD, Marco Matucci-Cerinic Speakers bureau: has received consulting fees or honorarium from Actelion, Janssen, Inventiva, Bayer, Biogen, Boehringer, CSL Behring, Corbus, Galapagos, Mitsubishi, Samsung, Regeneron, Acceleron, MSD, Chemomab, Lilly, Pfizer, Roche, Grant/research support from: has received consulting fees or honorarium from Actelion, Janssen, Inventiva, Bayer, Biogen, Boehringer, CSL Behring, Corbus, Galapagos, Mitsubishi, Samsung, Regeneron, Acceleron, MSD, Chemomab, Lilly, Pfizer, Roche
Collapse
|
29
|
Rosa I, Ibba‐Manneschi L, Guasti D, Perigli G, Faussone‐Pellegrini M, Manetti M. Morphologic evidence of telocytes in human thyroid stromal tissue. J Cell Mol Med 2022; 26:2477-2481. [PMID: 35307951 PMCID: PMC8995439 DOI: 10.1111/jcmm.17282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022] Open
Abstract
Despite the evidence accumulated over the past decade that telocytes (TCs) are a distinctive, though long neglected, cell entity of the stromal microenvironment of several organs of the human body, to date their localization in the endocrine glands remains almost unexplored. This study was therefore undertaken to examine the presence and characteristics of TCs in normal human thyroid stromal tissue through an integrated morphologic approach featuring light microscopy and ultrastructural analysis. TCs were first identified by immunohistochemistry that revealed the existence of an intricate network of CD34+ stromal cells spread throughout the thyroid interfollicular connective tissue. Double immunofluorescence allowed to clearly differentiate CD34+ stromal cells lacking CD31 immunoreactivity from neighbour CD31+ microvascular structures, and the evidence that these stromal cells coexpressed CD34 and platelet‐derived growth factor receptor α further strengthened their identification as TCs. Transmission electron microscopy confirmed the presence of stromal cells ultrastructurally identifiable as TCs projecting their characteristic cytoplasmic processes (i.e., telopodes) into the narrow interstitium between thyroid follicles and blood microvessels, where telopodes intimately surrounded the basement membrane of thyrocytes. Collectively, these morphologic findings provide the first comprehensive demonstration that TCs are main constituents of the human thyroid stroma and lay the necessary groundwork for further in‐depth studies aimed at clarifying their putative implications in glandular homeostasis and pathophysiology.
Collapse
|
30
|
Manetti M, Rosa I, Fioretto BS, Matucci-Cerinic M, Romano E. Decreased Serum Levels of SIRT1 and SIRT3 Correlate with Severity of Skin and Lung Fibrosis and Peripheral Microvasculopathy in Systemic Sclerosis. J Clin Med 2022; 11:1362. [PMID: 35268452 PMCID: PMC8910971 DOI: 10.3390/jcm11051362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Systemic sclerosis (SSc, scleroderma) is a severe autoimmune connective tissue disease characterized by widespread peripheral microvasculopathy, and progressive cutaneous and visceral fibrosis, leading to significant organ dysfunction. Sirtuins (SIRTs) are a family of NAD-dependent protein deacetylases with pleiotropic effects on a variety of biological processes, including metabolism, cell survival, and aging. In the last decades, increasing studies have explored the contribution of SIRTs to the pathogenesis of SSc, highlighting a significant antifibrotic effect of both SIRT1 and SIRT3. On these bases, the aim of this study was to measure circulating SIRT1 and SIRT3 levels by enzyme-linked immune-sorbent assay in a well-characterized cohort of SSc patients (n = 80) and healthy controls (n = 71), focusing on their possible association with disease clinical features, and their potential as biomarkers reflecting SSc activity and severity. Significantly decreased serum levels of both SIRT1 and SIRT3 were found in SSc patients compared to controls. In SSc, the reduction in circulating SIRT1 and SIRT3 associated with a greater extent of cutaneous fibrosis, presence of interstitial lung disease, and worse pulmonary function. Serum SIRT1 and SIRT3 decrease also correlated with the severity of nailfold microvascular damage, with SIRT3 levels being additionally related to the occurrence of digital ulcers. The levels of these two proteins showed a direct correlation with one another in the circulation of SSc patients. Of the two SIRTs, serum SIRT3 was found to better reflect disease activity and severity in a logistic regression analysis model. Our findings suggest that serum SIRT1 and SIRT3 may represent novel potential biomarkers of increased risk for a more severe, life-threatening SSc disease course.
Collapse
|
31
|
Carretti G, Mirandola D, Maestrini F, Sequi L, Germano S, Muraca MG, Miccinesi G, Manetti M, Marini M. Quality of life improvement in breast cancer survivors affected by upper limb lymphedema through a novel multiperspective physical activity methodology: a monocentric pilot study. Breast Cancer 2022; 29:437-449. [PMID: 35025064 DOI: 10.1007/s12282-021-01322-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/05/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Chronic lymphedema causes psychophysical sequelae jeopardizing quality of life (QoL) of breast cancer (BC) survivors, and lack of effective therapies represents a major challenge for healthcare professionals. Structured adapted physical activity (APA) may represent an effective strategy to attenuate cancer treatment-related impairments and improve QoL. Here, we describe the effects of a specific APA intervention based on a novel multiperspective methodology in counteracting lymphedema-related morphofunctional alterations and improving QoL of BC survivors. METHODS BC survivors with chronic moderate/severe lymphedema attending the Cancer Rehabilitation Center in Florence were assessed before and after 8-week APA. The protocol consisted of both APA specialist-supervised and self-leaded sessions using a tailor-designed proprioceptive board. Body mass index, bioimpedance parameters, indirect upper limb volume measurement, and ultrasonography were performed. Wrist flexion/extension and hand strength functional tests were also executed. QoL, depression/anxiety and pain intensity were evaluated by ULL27, HADS, distress thermometer and NRS questionnaires, respectively. RESULTS Although bioimpedance, ultrasound and volumetric measures remained mostly unchanged, wrist mobility, pain perception, depression, and QoL were all significantly ameliorated after APA. CONCLUSIONS Our findings suggest that a multidisciplinary treatment approach involving APA professionals should be employed in the management of BC-related lymphedema to improve patient psychophysical outcomes and QoL.
Collapse
|
32
|
Manetti M. Comment on: Iontophoresis of treprostinil promotes wound healing in a murine model of scleroderma-related ulcers. Rheumatology (Oxford) 2022; 61:e156-e157. [PMID: 35015845 DOI: 10.1093/rheumatology/keac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 12/23/2021] [Indexed: 11/13/2022] Open
|
33
|
Marini M, Tani A, Manetti M, Sgambati E. Overview of sialylation status in human nervous and skeletal muscle tissues during aging. Acta Histochem 2021; 123:151813. [PMID: 34753032 DOI: 10.1016/j.acthis.2021.151813] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022]
Abstract
Sialic acids (Sias) are a large and heterogeneous family of electronegatively charged nine-carbon monosaccharides containing a carboxylic acid and are mostly found as terminal residues in glycans of glycoproteins and glycolipids such as gangliosides. They are linked to galactose or N-acetylgalactosamine via α2,3 or α2,6 linkage, or to other Sias via α2,8 or more rarely α2,9 linkage, resulting in mono, oligo and polymeric forms. Given their characteristics, Sias play a crucial role in a multitude of human tissue biological processes in physiological and pathological conditions, ranging from development and growth to adult life until aging. Here, we review the sialylation status in human adult life focusing on the nervous and skeletal muscle tissues, which both display significant structural and functional changes during aging, strongly impacting on the whole human body and, therefore, on the quality of life. In particular, this review highlights the fundamental roles played by different types of glycoconjugates Sias in several cellular biological processes in the nervous and skeletal muscle tissues during adult life, also discussing how changes in Sia content during aging may contribute to the physiological decline of physical and nervous functions and to the development of age-related degenerative pathologies. Based on our current knowledge, further in-depth investigations could help to develop novel prophylactic strategies and therapeutic approaches that, by maintaining and/or restoring the correct sialylation status in the nervous and skeletal muscle tissues, could contribute to aging slowing and the prevention of age-related pathologies.
Collapse
|
34
|
Romano E, Rosa I, Fioretto BS, Kosalka-Wegiel J, Sticchi E, Bellando-Randone S, Manetti M, Matucci-Cerinic M. A candidate gene study reveals association between a variant of the SRp55 splicing factor gene and systemic sclerosis. Clin Exp Rheumatol 2021; 40:1921-1925. [DOI: 10.55563/clinexprheumatol/mpgq0y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022]
|
35
|
Romano E, Rosa I, Fioretto BS, Matucci-Cerinic M, Manetti M. New Insights into Profibrotic Myofibroblast Formation in Systemic Sclerosis: When the Vascular Wall Becomes the Enemy. Life (Basel) 2021; 11:610. [PMID: 34202703 PMCID: PMC8307837 DOI: 10.3390/life11070610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
In systemic sclerosis (SSc), abnormalities in microvessel morphology occur early and evolve into a distinctive vasculopathy that relentlessly advances in parallel with the development of tissue fibrosis orchestrated by myofibroblasts in nearly all affected organs. Our knowledge of the cellular and molecular mechanisms underlying such a unique relationship between SSc-related vasculopathy and fibrosis has profoundly changed over the last few years. Indeed, increasing evidence has suggested that endothelial-to-mesenchymal transition (EndoMT), a process in which profibrotic myofibroblasts originate from endothelial cells, may take center stage in SSc pathogenesis. While in arterioles and small arteries EndoMT may lead to the accumulation of myofibroblasts within the vessel wall and development of fibroproliferative vascular lesions, in capillary vessels it may instead result in vascular destruction and formation of myofibroblasts that migrate into the perivascular space with consequent tissue fibrosis and microvessel rarefaction, which are hallmarks of SSc. Besides endothelial cells, other vascular wall-resident cells, such as pericytes and vascular smooth muscle cells, may acquire a myofibroblast-like synthetic phenotype contributing to both SSc-related vascular dysfunction and fibrosis. A deeper understanding of the mechanisms underlying the differentiation of myofibroblasts inside the vessel wall provides the rationale for novel targeted therapeutic strategies for the treatment of SSc.
Collapse
|
36
|
Rosa I, Marini M, Manetti M. Telocytes: An Emerging Component of Stem Cell Niche Microenvironment. J Histochem Cytochem 2021; 69:795-818. [PMID: 34165348 DOI: 10.1369/00221554211025489] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Telocytes (TCs) are newly identified interstitial cells characterized by thin and long cytoplasmic processes, called telopodes, which exhibit a distinctive moniliform shape and, often, a sinuous trajectory. Telopodes typically organize in intricate networks within the stromal space of most organs, where they communicate with neighboring cells by means of specialized cell-to-cell junctions or shedding extracellular vesicles. Hence, TCs are generally regarded as supporting cells that help in the maintenance of local tissue homeostasis, with an ever-increasing number of studies trying to explore their functions both in physiological and pathological conditions. Notably, TCs appear to be part of stem cell (SC) niches in different organs, including the intestine, skeletal muscle, heart, lung, and skin. Indeed, growing evidence points toward a possible implication of TCs in the regulation of the activity of tissue-resident SCs and in shaping the SC niche microenvironment, thus contributing to tissue renewal and repair. Here, we review how the introduction of TCs into the scientific literature has deepened our knowledge of the stromal architecture focusing on the intestine and skeletal muscle, two organs in which the recently unveiled unique relationship between TCs and SCs is currently in the spotlight as potential target for tissue regenerative purposes.
Collapse
|
37
|
Chiari M, Barone S, Bombini A, Calzolai G, Carraresi L, Castelli L, Czelusniak C, Fedi ME, Gelli N, Giambi F, Giardi F, Giuntini L, Lagomarsino S, Liccioli L, Lucarelli F, Manetti M, Massi M, Mazzinghi A, Nava S, Ottanelli P, Sciortino S, Ruberto C, Sodi L, Taccetti F, Mandò PA. LABEC, the INFN ion beam laboratory of nuclear techniques for environment and cultural heritage. EUROPEAN PHYSICAL JOURNAL PLUS 2021; 136:472. [PMID: 33968561 PMCID: PMC8086225 DOI: 10.1140/epjp/s13360-021-01411-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/09/2021] [Indexed: 05/28/2023]
Abstract
The LABEC laboratory, the INFN ion beam laboratory of nuclear techniques for environment and cultural heritage, located in the Scientific and Technological Campus of the University of Florence in Sesto Fiorentino, started its operational activities in 2004, after INFN decided in 2001 to provide our applied nuclear physics group with a large laboratory dedicated to applications of accelerator-related analytical techniques, based on a new 3 MV Tandetron accelerator. The new accelerator greatly improved the performance of existing Ion Beam Analysis (IBA) applications (for which we were using since the 1980s an old single-ended Van de Graaff accelerator) and in addition allowed to start a novel activity of Accelerator Mass Spectrometry (AMS), in particular for 14C dating. Switching between IBA and AMS operation became very easy and fast, which allowed us high flexibility in programming the activities, mainly focused on studies of cultural heritage and atmospheric aerosol composition, but including also applications to biology, geology, material science and forensics, ion implantation, tests of radiation damage to components, detector performance tests and low-energy nuclear physics. This paper describes the facilities presently available in the LABEC laboratory, their technical features and some success stories of recent applications.
Collapse
|
38
|
Romano E, Rosa I, Fioretto BS, Cerinic MM, Manetti M. The Role of Pro-fibrotic Myofibroblasts in Systemic Sclerosis: from Origin to Therapeutic Targeting. Curr Mol Med 2021; 22:209-239. [PMID: 33823766 DOI: 10.2174/0929867328666210325102749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/02/2021] [Accepted: 03/09/2021] [Indexed: 11/22/2022]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex connective tissue disorder characterized by multisystem clinical manifestations resulting from immune dysregulation/autoimmunity, vasculopathy and, most notably, progressive fibrosis of the skin and internal organs. In recent years, it has emerged that the main drivers of SSc-related tissue fibrosis are myofibroblasts, a type of mesenchymal cells with both the extracellular matrix-synthesizing features of fibroblasts and the cytoskeletal characteristics of contractile smooth muscle cells. The accumulation and persistent activation of pro-fibrotic myofibroblasts during SSc development and progression result into elevated mechanical stress and reduced matrix plasticity within the affected tissues and may be ascribed to a reduced susceptibility of these cells to pro-apoptotic stimuli, as well as their increased formation from tissue-resident fibroblasts or transition from different cell types. Given the crucial role of myofibroblasts in SSc pathogenesis, finding the way to inhibit myofibroblast differentiation and accumulation by targeting their formation, function and survival may represent an effective approach to hamper the fibrotic process or even halt or reverse established fibrosis. In this review, we discuss the role of myofibroblasts in SSc-related fibrosis, with a special focus on their cellular origin and the signaling pathways implicated in their formation and persistent activation. Furthermore, we provide an overview of potential therapeutic strategies targeting myofibroblasts that may be able to counteract fibrosis in this pathological condition.
Collapse
|
39
|
Mencucci R, Favuzza E, Becatti M, Tani A, Mazzantini C, Vignapiano R, Fiorillo C, Pellegrini-Giampietro D, Manetti M, Marini M, Landucci E. Co-expression of the SARS-CoV-2 entry receptors ACE2 and TMPRSS2 in healthy human conjunctiva. Exp Eye Res 2021; 205:108527. [PMID: 33667466 PMCID: PMC7921718 DOI: 10.1016/j.exer.2021.108527] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
The purpose of this study was to evaluate the expression of the SARS-CoV-2 receptors ACE2 and TMPRSS2 in an immortalized human conjunctival epithelial cell line and in healthy human conjunctiva excised during ocular surgery, using Western blot, confocal microscopy and immunohistochemistry. The Western blot showed that ACE2 and TMPRSS2 proteins were expressed in human immortalized conjunctival cells, and this was confirmed by confocal microscopy images, that demonstrated a marked cellular expression of the viral receptors and their co-localization on the cell membranes. Healthy conjunctival samples from 11 adult patients were excised during retinal detachment surgery. We found the expression of ACE2 and TMPRSS2 in all the conjunctival surgical specimens analyzed and their co-localization in the superficial conjunctival epithelium. The ACE2 Western blot levels and immunofluorescence staining for ACE2 were variable among specimens. These results suggest the susceptibility of the conjunctival epithelium to SARS-CoV-2 infection, even though with a possible interindividual variability.
Collapse
|
40
|
Bandinelli F, Benucci M, Salaffi F, Manetti M, Infantino M, Damiani A, Manfredi M, Grossi V, Matucci A, Li Gobbi F, Marin G. Do new and old biomarkers of early undifferentiated arthritis correlate with Arthritis Impact Measurement Scales? Clin Exp Rheumatol 2021; 39:79-83. [DOI: 10.55563/clinexprheumatol/nqqx5k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/24/2020] [Indexed: 11/13/2022]
|
41
|
Bruni C, Guignabert C, Manetti M, Cerinic MM, Humbert M. The multifaceted problem of pulmonary arterial hypertension in systemic sclerosis. THE LANCET. RHEUMATOLOGY 2021; 3:e149-e159. [PMID: 38279370 DOI: 10.1016/s2665-9913(20)30356-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 01/16/2023]
Abstract
Cardiopulmonary complications are a leading cause of death in systemic sclerosis. Pulmonary hypertension in particular carries a high mortality and morbidity burden. Patients with systemic sclerosis can suffer from all of the clinical groups of pulmonary hypertension, particularly pulmonary arterial hypertension and pulmonary hypertension related to interstitial lung disease. Despite a similar pathogenetic background with idiopathic pulmonary arterial hypertension, different mechanisms determine a worse prognostic outcome for patients with systemic sclerosis. In this Viewpoint, we will consider the link between pathogenetic and potential therapeutic targets for the treatment of pulmonary hypertension in the context of systemic sclerosis, with a focus on the current unmet needs, such as the importance of early screening and detection, the absence of agreed criteria to distinguish pulmonary arterial hypertension with interstitial lung disease from pulmonary hypertension due to lung fibrosis, and the need for a holistic treatment approach to target all the vascular, immunological, and inflammatory components of the disease.
Collapse
|
42
|
Rosa I, Romano E, Fioretto BS, Matucci-Cerinic M, Manetti M. Adipose-derived stem cells: Pathophysiologic implications vs therapeutic potential in systemic sclerosis. World J Stem Cells 2021; 13:30-48. [PMID: 33584978 PMCID: PMC7859990 DOI: 10.4252/wjsc.v13.i1.30] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) residing in the stromal vascular fraction (SVF) of white adipose tissue are recently emerging as an alternative tool for stem cell-based therapy in systemic sclerosis (SSc), a complex connective tissue disorder affecting the skin and internal organs with fibrotic and vascular lesions. Several preclinical and clinical studies have reported promising therapeutic effects of fat grafting and autologous SVF/ADSC-based local treatment for facial and hand cutaneous manifestations of SSc patients. However, currently available data indicate that ADSCs may represent a double-edged sword in SSc, as they may exhibit a pro-fibrotic and anti-adipogenic phenotype, possibly behaving as an additional pathogenic source of pro-fibrotic myofibroblasts through the adipocyte-to-myofibroblast transition process. Thus, in the perspective of a larger employ of SSc-ADSCs for further therapeutic applications, it is important to definitely unravel whether these cells present a comparable phenotype and similar immunosuppressive, anti-inflammatory, anti-fibrotic and pro-angiogenic properties in respect to healthy ADSCs. In light of the dual role that ADSCs seem to play in SSc, this review will provide a summary of the most recent insights into the preclinical and clinical studies employing SVF and ADSCs for the treatment of the disease and, at the same time, will focus on the main findings highlighting the possible involvement of these stem cells in SSc-related fibrosis pathogenesis.
Collapse
|
43
|
Andreucci E, Margheri F, Peppicelli S, Bianchini F, Ruzzolini J, Laurenzana A, Fibbi G, Bruni C, Bellando-Randone S, Guiducci S, Romano E, Manetti M, Matucci-Cerinic M, Calorini L. Glycolysis-derived acidic microenvironment as a driver of endothelial dysfunction in systemic sclerosis. Rheumatology (Oxford) 2021; 60:4508-4519. [PMID: 33471123 DOI: 10.1093/rheumatology/keab022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Systemic sclerosis (SSc) is an autoimmune disease characterized by peripheral vasculopathy and skin and internal organ fibrosis. Accumulating evidence underlines a close association between a metabolic reprogramming of activated fibroblasts and fibrosis. This prompted us to determine the metabolism of SSc dermal fibroblasts and the effect on the vasculopathy characterizing the disease. METHODS Seahorse XF96 Extracellular Flux Analyzer was exploited to evaluate SSc fibroblast metabolism. In vitro invasion and capillary morphogenesis assays were used to determine the angiogenic ability of endothelial cells (EC). Immunofluorescence, flow cytometer and real time PCR techniques provided evidence of the molecular mechanism behind the impaired vascularization that characterizes SSc patients. RESULTS SSc fibroblasts, compared with control, showed a boosted glycolytic metabolism with increased lactic acid release and subsequent extracellular acidification, that in turn was found to impair EC invasion and organization in capillary-like networks without altering cell viability. A molecular link between extracellular acidosis and endothelial dysfunction was identified as acidic EC up-regulated MMP-12 which cleaves and inactivates uPAR, impairing angiogenesis in SSc. Moreover, the acidic environment was found to induce the loss of endothelial markers and the acquisition of mesenchymal-like features in EC, thus promoting the endothelial-to-mesenchymal transition (EndoMT) process that contributes to both capillary rarefaction and tissue fibrosis in SSc. CONCLUSION This study disclosed a liaison among the metabolic reprogramming of SSc dermal fibroblasts, extracellular acidosis and endothelial dysfunction that may contribute to the impairment and loss of peripheral capillary networks in SSc disease.
Collapse
|
44
|
Rosa I, Faussone-Pellegrini MS, Romano E, Ibba-Manneschi L, Matucci-Cerinic M, Manetti M. Impairment in the telocyte/CD34 + stromal cell network in human rheumatoid arthritis synovium. J Cell Mol Med 2020; 25:2274-2278. [PMID: 33350073 PMCID: PMC7882959 DOI: 10.1111/jcmm.16225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/25/2020] [Accepted: 12/03/2020] [Indexed: 12/19/2022] Open
Abstract
Telocytes (TCs)/CD34+ stromal cells have recently emerged as peculiar interstitial cells detectable in a variety of organs throughout the human body. TCs are typically arranged in networks establishing unique spatial relationships with neighbour cells and likely contributing to the maintenance of tissue homeostasis by both cell‐to‐cell contacts and releasing extracellular vesicles. Hence, TC defects are being increasingly reported in different pathologies, such as chronic inflammatory and fibrotic conditions. In this regard, TCs/CD34+ stromal cells have been shown to constitute an intricate interstitial network in the subintimal area of the normal human synovial membrane, but whether they are altered in chronic synovitis has yet to be explored. We therefore undertook a morphologic study to compare the distribution of TCs/CD34+ stromal cells between normal synovium and chronically inflamed synovium from patients with rheumatoid arthritis (RA) by using CD34 immunohistochemistry and CD31/CD34 double immunofluorescence. CD34 immunostaining revealed that, at variance with normal synovium, the inflamed and hyperplastic RA synovial tissue was nearly or even completely devoid of TCs/CD34+ stromal cells. Double immunofluorescence confirmed that almost all CD34+ tissue components detectable in RA synovium were blood vessels coexpressing CD31, while a widespread network of CD31−/CD34+ TCs was clearly evident in the whole sublining layer of normal synovium. In the context of the emerging diverse roles of TCs/CD34+ stromal cells in the regulation of tissue homeostasis and structure, the remarkable impairment in their networks herein uncovered in RA synovium may suggest important pathophysiologic implications that will be worth investigating further.
Collapse
|
45
|
Rosa I, Romano E, Fioretto BS, Manetti M. The contribution of mesenchymal transitions to the pathogenesis of systemic sclerosis. Eur J Rheumatol 2020; 7:S157-S164. [PMID: 31922472 PMCID: PMC7647682 DOI: 10.5152/eurjrheum.2019.19081] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/25/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic sclerosis (SSc) is a multifaceted connective tissue disease characterized by widespread vasculopathy and autoimmune reactions that evolve into progressive interstitial, perivascular, and vessel wall fibrosis that affects the skin and multiple internal organs. Such an uncontrolled fibrotic process gradually disrupts the physiologic architecture of the affected tissues and frequently leads to significant organ dysfunction, thus representing a major cause of death in SSc patients. The main fibrosis orchestrators in SSc are represented by chronically activated myofibroblasts, a peculiar population of mesenchymal cells combining the extracellular matrix-synthesizing features of fibroblasts with cytoskeletal characteristics of contractile smooth muscle cells. Multiple lines of evidence support the notion that profibrotic myofibroblasts may derive not only from the activation of tissue resident fibroblasts but also from a variety of additional cell types, including pericytes, epithelial cells, vascular endothelial cells and preadipocytes/adipocytes. Here we overview an emerging picture that espouses that several cell transitional processes may be novel essential contributors to the pool of profibrotic myofibroblasts in SSc, potentially representing new suitable targets for therapeutic purposes. An in-depth dissection of the multiple origins of myofibroblasts and the underlying molecular mechanisms may be crucial in the process of deciphering the cellular bases of fibrosis persistence and refractoriness to the treatment and, therefore, may help in developing more effective and personalized therapeutic opportunities for SSc patients.
Collapse
|
46
|
Romano E, Rosa I, Fioretto BS, Lucattelli E, Innocenti M, Ibba-Manneschi L, Matucci-Cerinic M, Manetti M. A Two-Step Immunomagnetic Microbead-Based Method for the Isolation of Human Primary Skin Telocytes/CD34+ Stromal Cells. Int J Mol Sci 2020; 21:ijms21165877. [PMID: 32824287 PMCID: PMC7461544 DOI: 10.3390/ijms21165877] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
Telocytes (TCs), commonly referred to as TCs/CD34+ stromal cells, are a peculiar type of interstitial cells with distinctive morphologic traits that are supposed to exert several biological functions, including tissue homeostasis regulation, cell-to-cell signaling, immune surveillance, and reparative/regenerative effects. At present, the majority of studies investigating these cells are mainly descriptive and focus only on their morphology, with a consequent paucity of functional data. To gain relevant insight into the possible functions of TCs, in vitro analyses are clearly required, but currently, the protocols for TC isolation are only at the early stages and not fully standardized. In the present in vitro study, we describe a novel methodology for the purification of human primary skin TCs through a two-step immunomagnetic microbead-based cell separation (i.e., negative selection for CD31 followed by positive selection for CD34) capable of discriminating these cells from other connective tissue-resident cells on the basis of their different immunophenotypic features. Our experiments clearly demonstrated that the proposed method allows a selective purification of cells exhibiting the peculiar TC morphology. Isolated TCs displayed very long cytoplasmic extensions with a moniliform silhouette (telopodes) and presented an immunophenotypic profile (CD31−/CD34+/PDGFRα+/vimentin+) that unequivocally differentiates them from endothelial cells (CD31+/CD34+/PDGFRα−/vimentin+) and fibroblasts (CD31−/CD34−/PDGFRα+/vimentin+). This novel methodology for the isolation of TCs lays the groundwork for further research aimed at elucidating their functional properties and possible translational applications, especially in the field of regenerative medicine.
Collapse
|
47
|
Avanoǧlu Güler A, Rossi FW, Bellando-Randone S, Prevete N, Tufan A, Manetti M, de Paulis A, Matucci-Cerinic M. The Role of Endogenous Eicosapentaenoic Acid and Docosahexaenoic Acid-Derived Resolvins in Systemic Sclerosis. Front Immunol 2020; 11:1249. [PMID: 32636845 PMCID: PMC7318896 DOI: 10.3389/fimmu.2020.01249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/18/2020] [Indexed: 12/19/2022] Open
Abstract
Resolvins, the member of specialized pro-resolving mediators, are produced from omega-3 polyunsaturated fatty acids as a response to an acute inflammatory process in that termination and resolution of inflammation. In the acute inflammation, these lipid mediators limit polymorphonuclear cells infiltration, proinflammatory cytokine production; promote efferocytosis, and regulate several cell types being important roles in innate and adaptive immunity. Any dysregulation or defect of the resolution phase result in prolonged, persistent inflammation and eventually fibrosis. Resolvins are implicated in the development of various chronic autoimmune diseases. Systemic sclerosis (SSc) is a very complicated, chronic autoimmune disorder proceeding with vasculopathy, inflammation, and fibrosis. Dysregulation of innate and adaptive immunity is another important contributing factor in the pathogenesis of SSc. In this review, we will focus on the different roles of this new family of lipid mediators, characterized by the ability to prevent the spread of inflammation and its chronicity in various ways and how they can control the development of fibrotic diseases like SSc.
Collapse
|
48
|
Romano E, Manetti M, Kosalka-Wegiel J, Fioretto BS, Rosa I, Sticchi E, Guiducci S, Bellando-Randone S, Ibba-Manneschi L, Matucci-Cerinic M. SAT0282 ASSOCIATION BETWEEN A VARIANT OF THE SRP55 SPLICING FACTOR GENE AND SYSTEMIC SCLEROSIS IN AN ITALIAN POPULATION. Ann Rheum Dis 2020. [DOI: 10.1136/annrheumdis-2020-eular.1707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background:In systemic sclerosis (SSc), alternative splicing of the last exon (exon 8) of vascular endothelial growth factor (VEGF)-A pre-mRNA is a key element in the switch from proangiogenic to antiangiogenic VEGF-A isoforms. The mRNA-binding protein serine/arginine protein 55 (SRp55, also known as SFRS6) is a key regulatory splicing factor that promotes distal splice-site selection in the exon 8 region of VEGF-A pre-mRNA and subsequent upregulation of the exon 8b-containing VEGF165b antiangiogenic isoform. Overexpression of both VEGF165b and SRp55 has been implicated in SSc-related angiogenesis impairment and peripheral vascular damage. Moreover, differential splicing of the VEGF-A gene has been shown to be critical for development of pulmonary fibrosis. Of note, previous studies reported the lack of sequence variations in the VEGF-A alternatively spliced region, while a single nucleotide polymorphism (SNP) in theSRp55gene (rs2235611) has been associated with susceptibility to disturbed ocular angiogenesis in proliferative diabetic retinopathy.Objectives:This case-control pilot study examined the possible implication ofSRp55rs2235611 SNP in the genetic predisposition to SSc susceptibility and clinical phenotype.Methods:A total population of 872 white Italian individuals (414 SSc patients, 458 controls) was studied. All patients were classified as limited and diffuse cutaneous SSc (lcSSc and dcSSc, respectively) and were clinically evaluated for the presence of autoantibodies (anticentromere, anti-Scl70 antibodies), pulmonary fibrosis and digital ulcers. TheSRp55rs2235611 SNP was genotyped by TaqMan Real-Time PCR.Results:SRp55rs2235611 genotype distribution and allele frequency were similar in SSc and healthy controls, though a trend toward significance was observed for genotype distribution (p=0.07). TheSRp55rs2235611 AA genotype significantly influenced the predisposition to SSc (OR 2.55, 95% CI 1.11 to 5.57, p = 0.03), and to both lcSSc (OR 2.80, 95% CI 1.16 to 6.84, p = 0.02) and dcSSc (OR 3.42, 95% CI 1.20 to 9.72, p = 0.02) subtypes. TheSRp55rs2235611 A minor allele and AA genotype showed a significant risk association with susceptibility to SSc-related pulmonary fibrosis (A allele: OR 1.39, 95% CI 1.00 to 1.93, p = 0.046; AA genotype: OR 3.95, 95% CI 1.48 to 10.54, p = 0.006). A trend toward an association between the AA genotype and anti-Scl70 antibody-positive SSc was also found (OR 2.82, 95% CI 0.95 to 8.37, p = 0.06). Both rs2235611 A allele and AA genotype were significantly associated with the SSc subset without digital ulcers (A allele: OR 1.33, 95% CI 1.01 to 1.75, p = 0.04; AA genotype: OR 3.26, 95% CI 1.32 to 8.03, p = 0.01).Conclusion:TheSRp55rs2235611 polymorphism is associated with susceptibility to SSc and, in particular, with SSc-related pulmonary fibrosis and peripheral vascular phenotype, consistent with a role of VEGF-A pre-mRNA alternative splicing in the development of pulmonary fibrosis and impairment of angiogenesis. Further replication studies are warranted to confirm our findings in independent SSc cohorts.Disclosure of Interests:Eloisa Romano: None declared, Mirko Manetti: None declared, Joanna Kosalka-Wegiel: None declared, Bianca Saveria Fioretto: None declared, Irene Rosa: None declared, Elena Sticchi: None declared, Serena Guiducci: None declared, Silvia Bellando-Randone: None declared, Lidia Ibba-Manneschi: None declared, Marco Matucci-Cerinic Grant/research support from: Actelion, MSD, Bristol-Myers Squibb, Speakers bureau: Acetelion, Lilly, Boehringer Ingelheim
Collapse
|
49
|
Fioretto BS, Rosa I, Romano E, Wang Y, Guiducci S, Zhang G, Manetti M, Matucci-Cerinic M. The contribution of epigenetics to the pathogenesis and gender dimorphism of systemic sclerosis: a comprehensive overview. Ther Adv Musculoskelet Dis 2020; 12:1759720X20918456. [PMID: 32523636 PMCID: PMC7236401 DOI: 10.1177/1759720x20918456] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/15/2020] [Indexed: 02/05/2023] Open
Abstract
Systemic sclerosis (SSc) is a life-threatening connective tissue disorder of unknown etiology characterized by widespread vascular injury and dysfunction, impaired angiogenesis, immune dysregulation and progressive fibrosis of the skin and internal organs. Over the past few years, a new trend of investigations is increasingly reporting aberrant epigenetic modifications in genes related to the pathogenesis of SSc, suggesting that, besides genetics, epigenetics may play a pivotal role in disease development and clinical manifestations. Like many other autoimmune diseases, SSc presents a striking female predominance, and even if the reason for this gender imbalance has yet to be completely understood, it appears that the X chromosome, which contains many gender and immune-related genes, could play a role in such gender-biased prevalence. Besides a short summary of the genetic background of SSc, in this review we provide a comprehensive overview of the most recent insights into the epigenetic modifications which underlie the pathophysiology of SSc. A particular focus is given to genetic variations in genes located on the X chromosome as well as to the main X-linked epigenetic modifications that can influence SSc susceptibility and clinical phenotype. On the basis of the most recent advances, there is realistic hope that integrating epigenetic data with genomic, transcriptomic, proteomic and metabolomic analyses may provide in the future a better picture of their functional implications in SSc, paving the right way for a better understanding of disease pathogenesis and the development of innovative therapeutic approaches.
Collapse
|
50
|
Marini M, Tani A, Manetti M, Sgambati E. Characterization and distribution of sialic acids in human testicular seminoma. Acta Histochem 2020; 122:151532. [PMID: 32143917 DOI: 10.1016/j.acthis.2020.151532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022]
Abstract
Aberrant content of sialic acids (Sias) has been observed in various human cancer types in different organs. Sias have been implicated in cancerous transformation, invasiveness and metastasis, and in the escaping of cancer cells from immune surveillance. Indeed, Sias are commonly regarded as important biomarkers to distinguish cancer cells from their healthy counterparts. However, scarce and not exhaustive investigations have been performed on Sia content in testicular cancers and, in particular, in seminoma, one of the most common malignant testicular tumors. Hence, the aim of this study was to investigate the content and distribution of Sias with different glycosidic linkage, namely α2,3 and α2,6 galactose- or N-acetyl-galactosamine-linked Sias and polymeric Sia (polySia), in the germinal and stromal components of human testes affected by seminoma compared to normal testicular tissue. Structural changes in seminoma tissue were examined using hematoxylin-eosin staining. α2,3 and α2,6 linked Sias were evaluated by lectin histochemistry (Maackia amurensis agglutinin (MAA) and Sambucus nigra agglutinin (SNA)), while confocal immunofluorescence was used for polySia detection. Histopathological findings in seminoma tissue included loss of seminiferous tubules replaced by clusters of uniform polygonal cells with a clear cytoplasm, bundles of fibrotic tissue, numerous microvessels and some atrophic tubules. The content of α2,3 and α2,6 linked Sias was lost in almost all seminoma components respect to normal tissue, with the exception of microvessels in which it was higher. On the contrary, polySia level was increased in all the seminoma components compared to normal testicular tissue. Our findings suggest that an aberrant content of different Sias might have important and differential roles in seminoma development and progression. In particular, polySia might be implicated in seminoma progression by promoting cancer invasiveness and regulating the cross-talk between cancer cells, reactive stroma and vessels. Thus, the possibility that polySia might represent an important biomarker for seminoma deserves further investigation.
Collapse
|