A proteomics approach to further highlight the altered inflammatory condition in Rett syndrome

https://doi.org/10.1016/j.abb.2020.108660Get rights and content

Highlights

  • RTT fibroblasts show a well distinct proteomic profile.

  • RTT proteins profile shows an over-representation of immune function and inflammatory pathways.

  • Inflammasome abnormalities are also evident in RTT.

Abstract

Rett syndrome (RTT) is a progressive neurodevelopmental disorder caused by mutations in the X-linked MECP2 gene. RTT patients show multisystem disturbances associated with perturbed redox homeostasis and inflammation, which appear as possible key factors in RTT pathogenesis. In this study, using primary dermal fibroblasts from control and RTT subjects, we performed a proteomic analysis that, together with data mining approaches, allowed us to carry out a comprehensive characterization of RTT cellular proteome. Functional and pathway enrichment analyses showed that differentially expressed proteins in RTT were mainly enriched in biological processes related to immune/inflammatory responses. Overall, by using proteomic data mining as supportive approach, our results provide a detailed insight into the molecular pathways involved in RTT immune dysfunction that, causing tissue and organ damage, can increase the vulnerability of affected patients to unknown endogenous factors or infections.

Introduction

Rett syndrome (RTT; OMIM 312750), a severe neurodevelopmental disorder, which predominantly affects females (about 1 per 10,000–15,000 live births), is mainly caused by sporadic loss-of-function mutations in the X-linked methyl-CpG-binding protein 2 gene (MECP2) [[1], [2], [3]]. Affected patients display prominent neurologic hallmarks, such as a rapid deterioration of the acquired psychomotor skills (i.e., stereotyped hand movements, absent or very limited speech, ataxia), seizures, and autistic-like behavior, which appear after a period of 6–18-months normal development [4,5]. In addition, common co-morbidities comprise sleep disturbances, breathing and cardiac problems, numerous gastrointestinal disorders, scoliosis, and osteopenia [[6], [7], [8], [9], [10]]. Rett syndrome affects different tissues and organs. It is linked to an abnormal muscular tone [11] and a vasomotor instability especially in the lower limbs [12], to a neurodevelopmental arrest followed by a regression phase where patients lose acquired language and motor skills and exhibit intellectual disability and hand stereotypies [[13], [14], [15]] and to a subclinical myocardial dysfunction [16]. Moreover, Rett patients frequently have marked decreases in bone mineral density leading to osteoporotic fractures [17]. Despite the well-known genetic background at the basis of Rett syndrome, the complete pathogenic mechanisms linking MeCP2 deficiency to the symptoms of this broad-spectrum pathology remain still unclear [18].

Other factors can contribute to the complexity of the disease, indeed an aberrant immune response, an impaired redox homeostasis [[19], [20], [21]], a perturbed cholesterol metabolism [22,23], an altered mitochondrial bioenergetics [24], and a subclinical inflammatory status [[25], [26], [27]] were shown to play a key role in RTT pathogenesis and progression. The condition defined as “OxInflammation”, consisting in a harmful cycle between the redox imbalance and the inflammatory status, has been observed at cellular and systemic levels in both RTT patients and animal models [19].

NF-κB is a redox-sensitive transcription factor involved in innate and adaptive immune responses, as well as in inflammation. Dysregulated NF-κB signaling cascade has been associated with the pathogenesis of several diseases including RTT [28,29]. Indeed, in a recent study our group demonstrated an increase of NF-κB p65 nuclear translocation coupled with high interleukin-1β (IL-1β) transcript levels, and a constitutive NLRP3 inflammasome activation (a multi-protein complex involved in innate immune responses) in primary dermal fibroblasts isolated from RTT patients [30]. Along with other evidence [28], this finding corroborates the important role of this pleiotropic transcription factor in the disorder, even in a non-immune cell type, whose involvement in pathological conditions characterized by chronic inflammation is now ever-growing [[31], [32], [33], [34], [35]].

In this study, using an ex-vivo model, i.e. primary dermal fibroblasts from RTT patients, and a proteomic data mining approach, we were able to confirm the involvement of inflammatory condition in RTT. Therefore, our results on one hand demonstrated that fibroblasts are a good model to investigate RTT [36]; on the other hand, they could be translated to immune cells, thus helping to justify the chronic inflammatory state and immune impairment observed in this disorder [26,37].

In this context, proteome analysis focusing on differentially expressed proteins between RTT and control cells is an effective supportive tool to better explore proteins related to inflammatory network. In particular, we have applied a proteomic approach in order to further examine the dysregulated activation of immune system, which may be responsible for damage tissues and organs, increasing the vulnerability of RTT patients to unknown endogenous factors or infections.

Section snippets

Subjects population

The study included two female patients with clinical diagnosis of classic RTT and MECP2 mutations (sample Rett 1 mutation: R255X, sample Rett 2 mutation:R168X) and two healthy subjects (age: 24 ± 12, and 34 ± 8, respectively). RTT diagnosis and inclusion/exclusion criteria were based on the revised RTT nomenclature consensus [38]. All the patients were admitted to the Child Neuropsychiatry Unit of the “Azienda Ospedaliera Universitaria Senese” (AOUS, Siena, Italy). Both RTT patients have

Results

The latest research on RTT has clearly highlighted how a low-grade chronic inflammatory condition, fueled by abnormal immune response, can play a part in the multisystem manifestations observed in RTT patients [19,28]. In our recent study, fibroblasts isolated from skin biopsies of RTT patients showed atypical function of NF-κB p65 pathway and altered expression of NLRP3 inflammasome machinery [30], proving to be a suitable cellular model able to recapitulate, in vitro, the phenotypic features

Discussion

In recent years, a chronic low-grade inflammatory state has emerged as a prominent phenotypic hallmark in RTT. An altered profile of circulating proinflammatory mediators associated with irregularities in cellular immune function including aberrant NLRP3 inflammasome activity appears to contribute to the broad spectrum of metabolic abnormalities recognized in RTT [19,28,30,54]. In an attempt to explain the molecular and cellular mechanisms behind the atypical immune function of RTT, we have

Conclusions

Taken together, all these findings reinforce the concept of the intricate molecular sequelae resulting from the mutation of a single gene such as MECP2. In particular, we found that RTT fibroblasts show a well distinct proteomic profile compared to control cells, highlighting once again the complex biological nature of MeCP2 deficient cells. To determine whether inflammation is an important component that also characterizes the RTT cellular proteome, we focused our attention on proteins linked

Limitations of the study

An eventual limitation of our study is the number of samples analyzed although considering that RTT is a rare disease and that the isolation of fibroblasts derives from a skin biopsy, which is a very invasive procedure and not easy to have access. In addition, considering the complex techniques and expensive consumables, it would not feasible to analyze a large set of patients. Another limitation would be the use of cell not fully immune competent to study inflammation, but we have previously

Declaration of competing interest

The authors declare no competing interests.

Acknowledgments

The authors thank Fondazione Umberto Veronesi (FUV) for the fellowship supporting VC.

References (87)

  • A. Pecorelli et al.

    Cytokines profile and peripheral blood mononuclear cells morphology in Rett and autistic patients

    Cytokine

    (2016)
  • Q. Zhang et al.

    30 Years of NF-κB: a blossoming of relevance to human pathobiology

    Cell

    (2017)
  • A. Pecorelli et al.

    Altered inflammasome machinery as a key player in the perpetuation of Rett syndrome oxinflammation

    Redox Biol.

    (2020)
  • Y. Lin et al.

    Sodium-deoxycholate-assisted tryptic digestion and identification of proteolytically resistant proteins

    Anal. Biochem.

    (2008)
  • K.J. Livak et al.

    Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method

    Methods

    (2001)
  • V. Cicaloni et al.

    Proteomic profiling reveals mitochondrial alterations in Rett syndrome

    Free Radic. Biol. Med.

    (2020)
  • A. Pecorelli et al.

    13-HODE, 9-HODE and ALOX15 as potential players in Rett syndrome OxInflammation

    Free Radic. Biol. Med.

    (2019)
  • J. Moscat et al.

    p62 at the crossroads of autophagy, apoptosis, and cancer

    Cell

    (2009)
  • L. Huang et al.

    β-catenin promotes NLRP3 inflammasome activation via increasing the association between NLRP3 and ASC

    Mol. Immunol.

    (2020)
  • R.E. Amir et al.

    Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2

    Nat. Genet.

    (1999)
  • A. Anderson et al.

    Twenty years of surveillance in Rett syndrome: what does this tell us?

    Orphanet J. Rare Dis.

    (2014)
  • B. Hagberg

    Clinical manifestations and stages of Rett syndrome

    Ment. Retard. Dev. Disabil. Res. Rev.

    (2002)
  • D.G. Glaze et al.

    Rett's syndrome: characterization of respiratory patterns and sleep

    Ann. Neurol.

    (1987)
  • C. Ellaway et al.

    Prolonged QT interval in Rett syndrome

    Arch. Dis. Child.

    (1999)
  • A. Jefferson et al.

    Clinical guidelines for management of bone health in Rett syndrome based on expert consensus and available evidence

    PloS One

    (2016)
  • V. Faundez et al.

    Molecular systems biology of neurodevelopmental disorders, Rett syndrome as an archetype

    Front. Integr. Neurosci.

    (2019)
  • Y. Nomura et al.

    Motor symptoms of the Rett syndrome: abnormal muscle tone, posture, locomotion and stereotyped movement

    Brain Dev.

    (1992)
  • E. Frullanti et al.

    Analysis of the phenotypes in the Rett networked database

    Int. J. Genom.

    (2019)
  • M. Zappella et al.

    Preserved speech variants of the Rett syndrome: molecular and clinical analysis

    Am. J. Med. Genet.

    (2001)
  • C. De Felice et al.

    Subclinical myocardial dysfunction in Rett syndrome

    Eur. Heart J. Cardiovasc. Imag.

    (2012)
  • C. Caffarelli et al.

    Teriparatide in the treatment of recurrent fractures in a Rett patient

    Clin. Cases Miner. Bone Metab.

    (2015)
  • G. Valacchi et al.

    OxInflammation: from subclinical condition to pathological biomarker

    Front. Physiol.

    (2018)
  • M. Segatto et al.

    Cholesterol metabolism is altered in Rett syndrome: a study on plasma and primary cultured fibroblasts derived from patients

    PloS One

    (2014)
  • A. Cortelazzo et al.

    Subclinical inflammatory status in Rett syndrome

    Mediat. Inflamm.

    (2014)
  • K. Sharma et al.

    MeCP2 in central nervous system glial cells: current updates

    Acta Neurobiol. Exp.

    (2018)
  • A. Pecorelli et al.

    Compromised immune/inflammatory responses in Rett syndrome

    Free Radical Biology and Medicine

    (2020)
  • L.A. Bautista-Hernández et al.

    Fibroblasts: the unknown sentinels eliciting immune responses against microorganisms

    Eur. J. Microbiol. Immunol. (Bp)

    (2017)
  • S.G. Dakin

    Inflammation or damage: fibroblasts decide

    Sci. Transl. Med.

    (2019)
  • A. Hamada et al.

    Trained immunity carried by non-immune cells

    Front. Microbiol.

    (2018)
  • C.D. Richards

    Innate immune cytokines, fibroblast phenotypes, and regulation of extracellular matrix in lung

    J. Interferon Cytokine Res.

    (2017)
  • S. Van Linthout et al.

    Crosstalk between fibroblasts and inflammatory cells

    Cardiovasc. Res.

    (2014)
  • V. Cordone et al.

    The complexity of Rett syndrome models: primary fibroblasts as a disease-in-a-dish reliable approach

    Drug Discovery Today: Disease Models

    (2019)
  • S. Leoncini et al.

    Cytokine dysregulation in MECP2- and CDKL5-related Rett syndrome: relationships with aberrant redox homeostasis, inflammation, and ω-3 PUFAs

    Oxid. Med. Cell Longev.

    (2015)
  • Cited by (5)

    • Ox-inflammasome involvement inneuroinflammation

      2023, Free Radical Biology and Medicine
    • The constitutive activation of TLR4-IRAK1- NFκB axis is involved in the early NLRP3 inflammasome response in peripheral blood mononuclear cells of Rett syndrome patients

      2022, Free Radical Biology and Medicine
      Citation Excerpt :

      We recently confirmed these findings by a proteomic approach on fibroblasts isolated from RTT patients. In this work among the over 2000 proteins identified, over 1200 proteins were common between control and RTT patients and the majority were clustered in the inflammatory responses [50]. The absence of MeCP2 in PBMCs and THP-1 made deficient with a lentiviral shRNA MeCP2 vector resulted in elevated expression of tumor necrosis factor alpha (TNFα), IL-6, and IL-3, due to the involvement of NF-κB signalling [51].

    View full text