Elsevier

Cellular Signalling

Volume 68, April 2020, 109527
Cellular Signalling

KRIT1 loss-mediated upregulation of NOX1 in stromal cells promotes paracrine pro-angiogenic responses

https://doi.org/10.1016/j.cellsig.2020.109527Get rights and content

Highlights

  • KRIT1 loss in stromal cells dysregulates oxidative stress and inflammatory pathways.

  • KRIT1 loss in fibroblasts promotes angiogenic responses.

  • NOX-1 increase in KRIT1−/− fibroblasts controls both VEGF and PGE2 production.

  • NOX-1 upregulation in KRIT1−/− stromal cells regulates endothelial activation.

Abstract

Cerebral cavernous malformation (CCM) is a cerebrovascular disorder of proven genetic origin characterized by abnormally dilated and leaky capillaries occurring mainly in the central nervous system, with a prevalence of 0.3–0.5% in the general population. Genetic studies have identified causative mutations in three genes, CCM1/KRIT1, CCM2 and CCM3, which are involved in the maintenance of vascular homeostasis. However, distinct studies in animal models have clearly shown that CCM gene mutations alone are not sufficient to cause CCM disease, but require additional contributing factors, including stochastic events of increased oxidative stress and inflammation. Consistently, previous studies have shown that up-regulation of NADPH oxidase-mediated production of reactive oxygen species (ROS) in KRIT1 deficient endothelium contributes to the loss of microvessel barrier function.

In this study, we demonstrate that KRIT1 loss-of-function in stromal cells, such as fibroblasts, causes the up-regulation of NADPH oxidase isoform 1 (NOX1) and the activation of inflammatory pathways, which in turn promote an enhanced production of proangiogenic factors, including vascular endothelial growth factor (VEGF) and prostaglandin E2 (PGE2). Furthermore and importantly, we show that conditioned media from KRIT1 null fibroblasts induce proliferation, migration, matrix metalloproteinase 2 (MMP2) activation and VE-cadherin redistribution in wild type human endothelial cells.

Taken together, our results demonstrate that KRIT1 loss-of-function in stromal cells affects the surrounding microenvironment through a NOX1-mediated induction and release of angiogenic factors that are able to promote paracrine proangiogenic responses in human endothelial cells, thus pointing to a novel role for endothelial cell-nonautonomous effects of KRIT1 mutations in CCM pathogenesis, and opening new perspectives for disease prevention and treatment.

Introduction

Cerebral cavernous malformations (CCMs), also known as cavernous angioma or cavernoma, are vascular anomalies typically found in the brain and spinal cord with a prevalence of 0.3%–0.5%. CCMs are mulberry-like, thin-walled sinusoidal capillaries lacking normal vessel structural components, including pericytes and astrocytes, and often surrounded by hemosiderin deposits and gliosis [[1], [2], [3]]. These vascular lesions can develop anywhere in the body, but signs and symptoms generally appear only when they occur in brain and spinal cord, where they account for 5–15% of all vascular malformations.

CCMs can occur as single or multiple lesions (even in the hundreds order), with size ranging from a few millimeters to a few centimeters. Despite the high prevalence of CCM lesions, approximately only 30% of affected people develop clinical symptoms, including recurrent headaches, neurological deficits, seizures, stroke, and intracerebral hemorrhage (ICH); however, the majority of CCM lesions remain clinically silent during most of the host's lifetime [3].

CCM is a disease of proven genetic origin that can arise sporadically or may be inherited as an autosomal dominant condition with incomplete penetrance and variable clinical expressivity [4]. The sporadic form (sCCM) accounts for up to 80% of cases, whereas the familial form (fCCM) accounts for at least 20% of cases. Genetic studies have identified three genes associated to CCMs: KRIT1 (CCM1), MGC4607 (CCM2) and PDCD10 (CCM3), which account for about 50%, 20% and 10% of the cases, respectively. The remaining 20% are likely associated to undetected genetic alterations of CCM genes [5]. Many different CCM mutations have been identified, most of which typical of a single family. A different clinical penetrance between the CCM genes (60–88% for KRIT1, up to 100% for CCM2, and 63% for PDCD10) [6], and a large variability of severity of CCM lesions even among family members carrying the same germline mutation have been observed. However, distinct studies in animal models have clearly shown that mutations of CCM genes are not sufficient to cause CCM disease, suggesting that additional factors can contribute to CCM disease pathogenesis [5,7]. Consistently, DNA sequencing analysis of surgically-resected CCM lesions from autosomal dominant CCM patients have identified somatic mutations at very low frequencies, suggesting that the minority of cells in the mature CCM harbor these mutations [8,9].

In the central nervous system (CNS) the endothelium is part of complex units, called neurovascular units (NVU), where it is in close contact with other cell types (pericytes, astrocytes and neurons). All components of this unit interact with each other in a multidimensional process in which mediators released from multiple cells engage distinct signaling pathways and effector systems in a highly orchestrated manner and safeguard the integrity of the structure itself by regulating immune response, angiogenesis, vasculogenesis, oligodendrogenesis, neuroprotection and neuroplasticity [[10], [11], [12]].

To date, the effect of the KRIT1 loss on NVU is not known. However, recent observations outline the importance of microenvironment in the development of vascular lesions observed in fCCM. For instance, Louvi et al., demonstrated in an animal model that CCM3 neural deletion has cell nonautonomous effects resulting in the formation of multiple vascular lesions that closely resemble human cavernomas. Consistently, in a very recent paper Malinverno et al., showed that vascular lesions originate from clonal expansion of few CCM3 KO endothelial cells that attract sourrounding wild-type endothelial cells thus contributing to cavernoma growth [3,13,14].

Although the effective mechanisms through which loss of CCM proteins leads to vascular malformations remain to be comprehensively defined, in recent years it has been demonstrated that these proteins exert pleiotropic effects, related to their role in the regulation of multiple molecules and mechanisms involved in angiogenesis, cellular response to oxidative stress, inflammation, cell-cell and cell-matrix adhesion, and cytoskeleton dynamics [3,15].

Reactive oxygen species (ROS) are produced by the activity of a wide array of cellular enzymes, including NADPH oxidases (NOX), enzymes of the mitochondrial respiratory chain, xanthine oxidases, cytochrome p450 monooxygenases, lipoxygenases and cyclooxygenases, which can be induced by a variety of endogenous and exogenous chemical and physical stimuli [16]. The NOX family of enzymes produces ROS as their sole function, and are becoming recognized as key modulators of signal transduction pathways with a physiological role under acute stress and a pathological role after excessive activation under chronic stress. ROS produced by NOX proteins are now recognized to play essential roles in the regulation of cytoskeletal remodeling, gene expression, proliferation, differentiation, migration, and cell death. The NOX isoforms (NOX1–5, and DUOX1/2) differ in their regulation, tissue and subcellular localization and even ROS products [17,18]. NOX1, NOX2, NOX4, and NOX5 are expressed in endothelium, vascular smooth muscle cells, fibroblasts, or perivascular adipocytes. While NOX1/NOX2 promote the development of endothelial dysfunction, hypertension, and inflammation, NOX4 may play a role in protecting the vasculature during stress; however, when its activity is increased, it may be detrimental [19]. Recently NOX1 has been involved in several brain diseases [20], and has been also described to play a role in cancer by inducing tumor progression and angiogenesis through the regulation of vascular endothelial growth factor (VEGF) expression [[21], [22], [23]].

Previously, we demonstrated that KRIT1 loss affects the intracellular redox homeostasis and results in increased ROS production through distinct mechanisms, including Forkhead box protein O1 (FoxO1) and superoxide dismutase (SOD) downregulation, NOX4 upregulation, and abnormal antioxidant responses, suggesting a novel pathogenetic mechanism whereby CCM disease may result from impaired endothelial cell defenses to microenvironmental oxidative stress events [4,16,[24], [25], [26], [27], [28], [29], [30]].

Herein, we show that KRIT1 loss-of-function in fibroblasts induces the upregulation of NOX1, which in turn can trigger a paracrine proangiogenic response in wild type endothelial cells through increased production and release of angiogenic growth factors, suggesting a novel important role for endothelial cell-nonautonomous effects of KRIT1 mutations in CCM disease pathogenesis, and pointing to NOX1 as a major regulator of these effects and as a new potential therapeutic target.

Section snippets

Cell culture

Wild-type (K+/+) and KRIT1 knock-out (K−/−) mouse embryonic fibroblast (MEF) cell lines were established from KRIT1+/+ and KRIT1−/− E8.5 mouse embryos, respectively [28]. KRIT1−/− MEFs re-expressing KRIT1 (K9/6) were obtained as previously reported [28]. Cells were cultured at 37 °C and 5% CO2 in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), with 4500 mg/l glucose and 100 U/ml penicillin/streptomycin (Euroclone, Milan, Italy).

Human Umbilical Vein

Conditioned medium from KRIT1−/− fibroblasts induces pro-angiogenic responses in wild type human endothelial cells

Accumulated evidences demonstrate that KRIT1 protein plays an important role in modulating different molecular pathways involved in the angiogenic process, whereas KRIT1 loss induces alteration of endothelial cell-cell and cell-extracellular matrix (ECM) adhesion, and enhanced vascular permeability [3,36,37]. However, recent evidence in conditional knockout (cKO) animal models has clearly shown that loss-of-function mutations in CCM genes alone are not sufficient for the development of CCM

Discussion

CCM lesions are typically found in the CNS and are characterized by dilated and leaky capillaries that are devoid of normal vessel structural components.

Several lines of evidence show that the KRIT1 protein is involved in different physiological aspects of endothelial biology, including vascular development, modulation of different redox-sensitive signaling pathways, and maintenance of endothelial barrier homeostasis, as well as that the absence of KRIT1 in endothelial cells induces an

Conclusions

Our findings provide novel insights into CCM pathogenesis, and suggest novel promising therapeutic options for CCM prevention and treatment.

Currently, the only therapy available for CCM is surgical excision or radiological destruction of the lesions. While several compounds are being investigated in preclinical studies, only a few agents have reached clinical testing and to date CCM has no pharmacological options [52]. Several studies have been conducted to identify new CCM-related cellular

Funding

This work was supported by the Telethon Foundation (grant GGP15219) to SFR and LT and MIUR (Progetto Dipartimento di Eccellenza 2018-2022) to LT and FF.

Acknowledgements

The authors are grateful to the Italian Research Network for Cerebral Cavernous Malformation (CCM Italia, http://www.ccmitalia.unito.it), and the Associazione Italiana Angiomi Cavernosi (AIAC, http://www.ccmitalia.unito.it/aiac) for fundamental collaboration and support.

References (54)

  • R.P. Brandes et al.

    Nox family NADPH oxidases: molecular mechanisms of activation

    Free Radic. Biol. Med.

    (2014)
  • J.M. Zabramski et al.

    Propranolol treatment of cavernous malformations with symptomatic hemorrhage

    World Neurosurg.

    (2016)
  • I.A. Awad et al.

    Cavernous angiomas: deconstructing a neurosurgical disease

    J. Neurosurg.

    (2019)
  • A. Padarti et al.

    Recent advances in cerebral cavernous malformation research

    Vessel Plus.

    (2018)
  • S.F. Retta et al.

    Oxidative stress and inflammation in cerebral cavernous malformation disease pathogenesis: Two sides of the same coin

    Int. J. Biochem. Cell Biol.

    (2016)
  • C. Antognelli et al.

    KRIT1 loss-of-function induces a chronic Nrf2-mediated adaptive homeostasis that sensitizes cells to oxidative stress: implication for cerebral cavernous malformation disease

    Free Radic. Biol. Med.

    (2018)
  • H. Choquet et al.

    Genetics of cerebral cavernous malformations: current status and future prospects

    J. Neurosurg. Sci.

    (2015)
  • I.J.H.M. de Vos et al.

    Review of familial cerebral cavernous malformations and report of seven additional families

    Am. J. Med. Genet. A

    (2017)
  • E. Trapani et al.

    Cerebral cavernous malformation (CCM) disease: from monogenic forms to genetic susceptibility factors

    J. Neurosurg. Sci.

    (2015)
  • D.A. McDonald et al.

    Lesions from patients with sporadic cerebral cavernous malformations harbor somatic mutations in the CCM genes: evidence for a common biochemical pathway for CCM pathogenesis

    Hum. Mol. Genet.

    (2014)
  • J. Gault et al.

    Cerebral cavernous malformations

    Neurosurgery.

    (2009)
  • L. Huang et al.

    Astrocyte signaling in the neurovascular unit after central nervous system injury

    Int. J. Mol. Sci.

    (2019)
  • A. Louvi et al.

    Loss of cerebral cavernous malformation 3 (Ccm3) in neuroglia leads to CCM and vascular pathology

    Proc. Natl. Acad. Sci.

    (2011)
  • M. Malinverno et al.

    Endothelial cell clonal expansion in the development of cerebral cavernous malformations

    Nat. Commun.

    (2019)
  • K.M. Draheim et al.

    Cerebral cavernous malformation proteins at a glance

    J. Cell Sci.

    (2014)
  • L. Goitre et al.

    Molecular crosstalk between integrins and cadherins: do reactive oxygen species set the talk?

    J. Signal Transduct.

    (2012)
  • G. Teixeira et al.

    Therapeutic potential of NADPH oxidase 1/4 inhibitors

    Br. J. Pharmacol.

    (2017)
  • Cited by (15)

    • CCM proteins are key players in redox signaling and oxidative stress regulation in Cerebral Cavernous Malformations

      2023, Modulation of Oxidative Stress: Biochemical, Physiological and Pharmacological Aspects
    • Polymorphisms in genes related to oxidative stress and inflammation: Emerging links with the pathogenesis and severity of Cerebral Cavernous Malformation disease

      2021, Free Radical Biology and Medicine
      Citation Excerpt :

      Over the 20 years since their identification, the characterization of the physiopathological functions of the three known causative genes of CCM disease, KRIT1 (CCM1), CCM2 and CCM3, has provided fundamental insights into the understanding of major pathogenetic mechanisms and the development of therapeutic strategies [11,43]. In particular, accumulated findings in cellular and animal models have clearly demonstrated that loss-of-function mutations of CCM genes cause pleiotropic effects by affecting multiple redox-sensitive signaling pathways and mechanisms implicated in cellular homeostasis and defenses against oxidative stress and inflammation [10,26–33,38,41,269]. In fact, whereas the evidence for a major involvement of CCM proteins in redox homeostasis and signaling continues to grow [39], it is now becoming clear that the direct effects of CCM gene mutations are not sufficient to cause overt disease, but only sensitize cells to additional critical determinants of CCM pathogenesis, such as oxidative stress and inflammatory events [10,11,34,42,250].

    • KRIT1 as a possible new player in melanoma aggressiveness

      2020, Archives of Biochemistry and Biophysics
      Citation Excerpt :

      Bands of gelatinase activity appeared as transparent areas against a blue background. Gelatinase activity was then evaluated by quantitative densitometry [15,42]. A375 cells were seeded in 96-well microplates (2.0 × 104 cells/well) in DMEM 10% FBS and after 12 and 18 and hours they were fixed for 10 min with 70% EtOH and colored with Coomassie Blue solution (10% Acetic acid, 10% ethanol, and 0,25% Blue Coomassie).

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    2

    CCM Italia research network (www.ccmitalia.unito.it).

    View full text