100 results on '"Retta SF"'
Search Results
2. KRIT1/CCM1 loss-of-function impairs advanced glycation endproducts receptors and detoxification and worsens fructose diet-induced hepatic dysmetabolism
- Author
-
Mastrocola, R, Cento, As, Fornelli, C, Zotta, A, Perrelli, A, and Retta, Sf
- Subjects
Cerebral Cavernous Malformation (CCM) Disease ,Redox Signaling ,KRIT1 ,Advanced Glycation End-Products (AGEs) ,Hepatic Dysmetabolism ,High Fructose Diet - Published
- 2019
3. Crosstalk Between Redox Signaling and Autophagy in Cerebral Cavernous Malformation Disease: From Basic Mechanisms to Therapeutic Strategies
- Author
-
Retta, Sf
- Subjects
Cerebral Cavernous Malformation (CCM) Disease ,Redox Signaling ,Oxidative Stress ,KRIT1 ,Autophagy - Published
- 2019
4. Multifunctional platinum@bsa-rapamycin nanocarriers for combinatorial therapy of cerebral cavernous malformation disease
- Author
-
Perrelli, A, De Luca, E, Pedone, D, Moglianetti, M, Fornelli, C, Zotta, A, Pulcini, D, Pompa, Pp, and Retta, Sf
- Subjects
Cerebral Cavernous Malformation (CCM) Disease ,Redox Signaling ,Oxidative Stress ,KRIT1 ,Autophagy ,Nanotechnology ,Multifunctional Platinum Nanocarriers ,Combinatorial Therapeutic Strategies - Published
- 2019
5. KRIT1/CCM1 loss-of-function affects sphingolipid metabolism inducing hepatosteatosis and predisposing to liver fibrosis and insulin-resistance
- Author
-
Mastrocola, R, Cento, As, Fornelli, C, Zotta, A, Perrelli, A, and Retta, Sf
- Subjects
Cerebral Cavernous Malformation (CCM) Disease ,Redox Signaling ,KRIT1 ,Sphingolipid Metabolism ,Hepatosteatosis ,Liver fibrosis ,Insulin-Resistance - Published
- 2019
6. Cerebral cavernous malformations: surgical or conservative management?
- Author
-
Fontanella, M, Cornali, C, Garbossa, D, Spena, G, Doglietto, F, Panciani, P, Zenga, F, Bacigaluppi, S, Retta, SF, and Ducati, A
- Subjects
surgery ,ddc: 610 ,conservative treatment ,610 Medical sciences ,Medicine ,cavernoma - Abstract
Objective: Microsurgical excision of cerebral cavernous malformations (CCM) is often routinely performed in patients with related epilepsy or even in asymptomatic cases to prevent hemorrhage. Few prospective studies have reliably demonstrated that surgery can statistically and clinically improve the[for full text, please go to the a.m. URL], 66. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC)
- Published
- 2015
- Full Text
- View/download PDF
7. EndMT contributes to the onset and progression of cerebral cavernous malformations
- Author
-
Maddaluno L, Rudini N, Cuttano R, Bravi L, Giampietro C, Corada M, Ferrarini L, Orsenigo F, Papa E, Boulday G, Tournier-Lasserve E, Chapon F, Richichi C, Retta SF, Lampugnani MG, and Dejana E.
- Published
- 2013
8. Inhibition of PI3K induces Rac activation and membrane ruffling in proto-Dbl exressing cells
- Author
-
Vanni, C, Visco, V, Mancini, P, Parodi, A, Ottaviano, C, Ognibene, M, Manazza, Ad, Retta, Sf, Varesio, L, Torrisi, Mr, and Eva, A.
- Published
- 2006
9. Yeast as a model for Ras signalling
- Author
-
Trabalzini, L, Retta, SF, Tisi, R, Belotti, F, Martegani, E, TISI, RENATA ANITA, BELOTTI, FIORELLA, MARTEGANI, ENZO, Trabalzini, L, Retta, SF, Tisi, R, Belotti, F, Martegani, E, TISI, RENATA ANITA, BELOTTI, FIORELLA, and MARTEGANI, ENZO
- Abstract
For centuries yeast species have been popular hosts for classical biotechnology processes, such as baking, brewing, and wine making, and more recently for recombinant proteins production, thanks to the advantages of unicellular organisms (i.e., ease of genetic manipulation and rapid growth) together with the ability to perform eukaryotic posttranslational modifications. Moreover, yeast cells have been used for few decades as a tool for identifying the genes and pathways involved in basic cellular processes such as the cell cycle, aging, and stress response.In the budding yeast S. cerevisiae the Ras/cAMP/PKA pathway is directly involved in the regulation of metabolism, cell growth, stress resistance, and proliferation in response to the availability of nutrients and in the adaptation to glucose, controlling cytosolic cAMP levels and consequently the cAMP-dependent protein kinase (PKA) activity.Moreover, Ras signalling has been identified in several pathogenic yeasts as a key controller for virulence, due to its involvement in yeast morphogenesis. Nowadays, yeasts are still useful for Ras-like proteins investigation, both as model organisms and as a test tube to study variants of heterologous Ras-like proteins.
- Published
- 2014
10. Methods to study the Ras2 protein activation state and the subcellular localization of Ras-GTP in saccharomyces cerevisiae
- Author
-
Trabalzini, L, Retta, SF, Colombo, S, Martegani, E, COLOMBO, SONIA, MARTEGANI, ENZO, Trabalzini, L, Retta, SF, Colombo, S, Martegani, E, COLOMBO, SONIA, and MARTEGANI, ENZO
- Abstract
Ras proteins were highly conserved during evolution. They function as a point of convergence for different signalling pathways in eukaryotes and are involved in a wide range of cellular responses (shift from gluconeogenic to fermentative growth, breakdown of storage carbohydrates, stress resistance, growth control and determination of life span, morphogenesis and development, and others). These proteins are members of the small GTPase superfamily, which are active in the GTP-bound form and inactive in the GDP-bound form. Given the importance of studies on the Ras protein activation state to understand the detailed mechanism of Ras-mediated signal transduction, we provide here a simple, sensitive, and reliable method, based on the high affinity interaction of Ras-GTP with the Ras binding domain (RBD) of Raf1, to measure the level of Ras2-GTP on total Ras2 in Saccharomyces cerevisiae. Moreover, to study the localization of Ras-GTP in vivo in single S. Cerevisiae cells, we expressed a probe consisting of a GFP fusion with a trimeric Ras Binding Domain of Raf1 (eGFP-RBD3), which was proven to be a useful live-cell biosensor for Ras-GTP in mammalian cells. © 2014 Springer Science+Business Media, LLC.
- Published
- 2014
11. Yeast as a model for Ras signalling
- Author
-
Fiorella Belotti, Renata Tisi, Enzo Martegani, Trabalzini, L, Retta, SF, Tisi, R, Belotti, F, and Martegani, E
- Subjects
Candida albican ,Cell growth ,ved/biology ,ved/biology.organism_classification_rank.species ,pseudohyphal growth ,Morphogenesis ,Saccharomyces cerevisiae ,Cell cycle ,Biology ,BIO/11 - BIOLOGIA MOLECOLARE ,Yeast ,Cell biology ,mating ,Signal transduction ,Protein kinase A ,Model organism ,Gene - Abstract
For centuries yeast species have been popular hosts for classical biotechnology processes, such as baking, brewing, and wine making, and more recently for recombinant proteins production, thanks to the advantages of unicellular organisms (i.e., ease of genetic manipulation and rapid growth) together with the ability to perform eukaryotic posttranslational modifications. Moreover, yeast cells have been used for few decades as a tool for identifying the genes and pathways involved in basic cellular processes such as the cell cycle, aging, and stress response. In the budding yeast S. cerevisiae the Ras/cAMP/PKA pathway is directly involved in the regulation of metabolism, cell growth, stress resistance, and proliferation in response to the availability of nutrients and in the adaptation to glucose, controlling cytosolic cAMP levels and consequently the cAMP-dependent protein kinase (PKA) activity. Moreover, Ras signalling has been identified in several pathogenic yeasts as a key controller for virulence, due to its involvement in yeast morphogenesis. Nowadays, yeasts are still useful for Ras-like proteins investigation, both as model organisms and as a test tube to study variants of heterologous Ras-like proteins.
- Published
- 2014
12. Methods to Study the Ras2 Protein Activation State and the Subcellular Localization of Ras-GTP in Saccharomyces cerevisiae
- Author
-
Enzo Martegani, Sonia Colombo, Trabalzini, L, Retta, SF, Colombo, S, and Martegani, E
- Subjects
Fluorescence microscopy ,biology ,cAMP/PKA pathway ,Saccharomyces cerevisiae ,BIO/11 - BIOLOGIA MOLECOLARE ,biology.organism_classification ,Subcellular localization ,Yeast ,Transport protein ,Cell biology ,Small G protein ,Protein structure ,Biochemistry ,Active Ra ,Small GTPase ,Ras2 ,Signal transduction ,Nutrient ,Binding domain - Abstract
Ras proteins were highly conserved during evolution. They function as a point of convergence for different signalling pathways in eukaryotes and are involved in a wide range of cellular responses (shift from gluconeogenic to fermentative growth, breakdown of storage carbohydrates, stress resistance, growth control and determination of life span, morphogenesis and development, and others). These proteins are members of the small GTPase superfamily, which are active in the GTP-bound form and inactive in the GDP-bound form. Given the importance of studies on the Ras protein activation state to understand the detailed mechanism of Ras-mediated signal transduction, we provide here a simple, sensitive, and reliable method, based on the high affinity interaction of Ras-GTP with the Ras binding domain (RBD) of Raf1, to measure the level of Ras2-GTP on total Ras2 in Saccharomyces cerevisiae. Moreover, to study the localization of Ras-GTP in vivo in single S. Cerevisiae cells, we expressed a probe consisting of a GFP fusion with a trimeric Ras Binding Domain of Raf1 (eGFP-RBD3), which was proven to be a useful live-cell biosensor for Ras-GTP in mammalian cells. © 2014 Springer Science+Business Media, LLC.
- Published
- 2013
- Full Text
- View/download PDF
13. Identification of galectin-3 as a novel potential prognostic/predictive biomarker and therapeutic target for cerebral cavernous malformation disease.
- Author
-
Kar S, Perrelli A, Bali KK, Mastrocola R, Kar A, Khan B, Gand L, Nayak A, Hartmann C, Kunz WS, Samii A, Bertalanffy H, and Retta SF
- Published
- 2023
- Full Text
- View/download PDF
14. KRIT1: A Traffic Warden at the Busy Crossroads Between Redox Signaling and the Pathogenesis of Cerebral Cavernous Malformation Disease.
- Author
-
Perrelli A, Ferraris C, Berni E, Glading AJ, and Retta SF
- Subjects
- Humans, Endothelial Cells metabolism, Signal Transduction, Oxidation-Reduction, Inflammation, Microtubule-Associated Proteins metabolism, KRIT1 Protein metabolism, Hemangioma, Cavernous, Central Nervous System genetics, Hemangioma, Cavernous, Central Nervous System metabolism
- Abstract
Significance: KRIT1 (Krev interaction trapped 1) is a scaffolding protein that plays a critical role in vascular morphogenesis and homeostasis. Its loss-of-function has been unequivocally associated with the pathogenesis of Cerebral Cavernous Malformation (CCM), a major cerebrovascular disease of genetic origin characterized by defective endothelial cell-cell adhesion and ensuing structural alterations and hyperpermeability in brain capillaries. KRIT1 contributes to the maintenance of endothelial barrier function by stabilizing the integrity of adherens junctions and inhibiting the formation of actin stress fibers. Recent Advances: Among the multiple regulatory mechanisms proposed so far, significant evidence accumulated over the past decade has clearly shown that the role of KRIT1 in the stability of endothelial barriers, including the blood-brain barrier, is largely based on its involvement in the complex machinery governing cellular redox homeostasis and responses to oxidative stress and inflammation. KRIT1 loss-of-function has, indeed, been demonstrated to cause an impairment of major redox-sensitive mechanisms involved in spatiotemporal regulation of cell adhesion and signaling, which ultimately leads to decreased cell-cell junction stability and enhanced sensitivity to oxidative stress and inflammation. Critical Issues: This review explores the redox mechanisms that influence endothelial cell adhesion and barrier function, focusing on the role of KRIT1 in such mechanisms. We propose that this supports a novel model wherein redox signaling forms the common link between the various pathogenetic mechanisms and therapeutic approaches hitherto associated with CCM disease. Future Directions: A comprehensive characterization of the role of KRIT1 in redox control of endothelial barrier physiology and defense against oxy-inflammatory insults will provide valuable insights into the development of precision medicine strategies. Antioxid. Redox Signal. 38, 496-528.
- Published
- 2023
- Full Text
- View/download PDF
15. Multidrug-Loaded Lipid Nanoemulsions for the Combinatorial Treatment of Cerebral Cavernous Malformation Disease.
- Author
-
Perrelli A, Bozza A, Ferraris C, Osella S, Moglia A, Mioletti S, Battaglia L, and Retta SF
- Abstract
Cerebral cavernous malformation (CCM) or cavernoma is a major vascular disease of genetic origin, whose main phenotypes occur in the central nervous system, and is currently devoid of pharmacological therapeutic strategies. Cavernomas can remain asymptomatic during a lifetime or manifest with a wide range of symptoms, including recurrent headaches, seizures, strokes, and intracerebral hemorrhages. Loss-of-function mutations in KRIT1/CCM1 are responsible for more than 50% of all familial cases, and have been clearly shown to affect cellular junctions, redox homeostasis, inflammatory responses, and angiogenesis. In this study, we investigated the therapeutic effects of multidrug-loaded lipid nanoemulsions in rescuing the pathological phenotype of CCM disease. The pro-autophagic rapamycin, antioxidant avenanthramide, and antiangiogenic bevacizumab were loaded into nanoemulsions, with the aim of reducing the major molecular dysfunctions associated with cavernomas. Through Western blot analysis of biomarkers in an in vitro CCM model, we demonstrated that drug-loaded lipid nanoemulsions rescue antioxidant responses, reactivate autophagy, and reduce the effect of pro-angiogenic factors better than the free drugs. Our results show the importance of developing a combinatorial preventive and therapeutic approach to reduce the risk of lesion formation and inhibit or completely revert the multiple hallmarks that characterize the pathogenesis and progression of cavernomas.
- Published
- 2023
- Full Text
- View/download PDF
16. Distant Recurrence of a Cerebral Cavernous Malformation in the Vicinity of a Developmental Venous Anomaly: Case Report of Local Oxy-Inflammatory Events.
- Author
-
Bianconi A, Salvati LF, Perrelli A, Ferraris C, Massara A, Minardi M, Aruta G, Rosso M, Massa Micon B, Garbossa D, and Retta SF
- Subjects
- Humans, Child, Preschool, Neoplasm Recurrence, Local, Magnetic Resonance Imaging, Brain pathology, Mutation, Hemangioma, Cavernous, Central Nervous System diagnostic imaging, Hemangioma, Cavernous, Central Nervous System genetics, Hemangioma, Cavernous, Central Nervous System surgery
- Abstract
Background: Cerebral cavernous malformations (CCMs) are a major type of cerebrovascular lesions of proven genetic origin that occur in either sporadic (sCCM) or familial (fCCM) forms, the latter being inherited as an autosomal dominant condition linked to loss-of-function mutations in three known CCM genes. In contrast to fCCMs, sCCMs are rarely linked to mutations in CCM genes and are instead commonly and peculiarly associated with developmental venous anomalies (DVAs), suggesting distinct origins and common pathogenic mechanisms., Case Report: A hemorrhagic sCCM in the right frontal lobe of the brain was surgically excised from a symptomatic 3 year old patient, preserving intact and pervious the associated DVA. MRI follow-up examination performed periodically up to 15 years after neurosurgery intervention demonstrated complete removal of the CCM lesion and no residual or relapse signs. However, 18 years after surgery, the patient experienced acute episodes of paresthesia due to a distant recurrence of a new hemorrhagic CCM lesion located within the same area as the previous one. A new surgical intervention was, therefore, necessary, which was again limited to the CCM without affecting the pre-existing DVA. Subsequent follow-up examination by contrast-enhanced MRI evidenced a persistent pattern of signal-intensity abnormalities in the bed of the DVA, including hyperintense gliotic areas, suggesting chronic inflammatory conditions., Conclusions: This case report highlights the possibility of long-term distant recurrence of hemorrhagic sCCMs associated with a DVA, suggesting that such recurrence is secondary to focal sterile inflammatory conditions generated by the DVA.
- Published
- 2022
- Full Text
- View/download PDF
17. Heterozygous Loss of KRIT1 in Mice Affects Metabolic Functions of the Liver, Promoting Hepatic Oxidative and Glycative Stress.
- Author
-
Mastrocola R, Aimaretti E, Ferreira Alves G, Cento AS, Fornelli C, Dal Bello F, Ferraris C, Goitre L, Perrelli A, and Retta SF
- Subjects
- Animals, Antioxidants, Glucose, Glycogen, KRIT1 Protein, Liver, Mice, Oxidative Stress genetics, Insulins, NF-E2-Related Factor 2 genetics
- Abstract
KRIT1 loss-of-function mutations underlie the pathogenesis of Cerebral Cavernous Malformation (CCM), a major vascular disease affecting the central nervous system (CNS). However, KRIT1 is also expressed outside the CNS and modulates key regulators of metabolic and oxy-inflammatory pathways, including the master transcription factor FoxO1, suggesting a widespread functional significance. Herein, we show that the KRIT1/FoxO1 axis is implicated in liver metabolic functions and antioxidative/antiglycative defenses. Indeed, by performing comparative studies in KRIT1 heterozygous (KRIT1
+/- ) and wild-type mice, we found that KRIT1 haploinsufficiency resulted in FoxO1 expression/activity downregulation in the liver, and affected hepatic FoxO1-dependent signaling pathways, which are markers of major metabolic processes, including gluconeogenesis, glycolysis, mitochondrial respiration, and glycogen synthesis. Moreover, it caused sustained activation of the master antioxidant transcription factor Nrf2, hepatic accumulation of advanced glycation end-products (AGEs), and abnormal expression/activity of AGE receptors and detoxifying systems. Furthermore, it was associated with an impairment of food intake, systemic glucose disposal, and plasma levels of insulin. Specific molecular alterations detected in the liver of KRIT1+/- mice were also confirmed in KRIT1 knockout cells. Overall, our findings demonstrated, for the first time, that KRIT1 haploinsufficiency affects glucose homeostasis and liver metabolic and antioxidative/antiglycative functions, thus inspiring future basic and translational studies.- Published
- 2022
- Full Text
- View/download PDF
18. Next-Generation Sequencing Advances the Genetic Diagnosis of Cerebral Cavernous Malformation (CCM).
- Author
-
Benedetti V, Canzoneri R, Perrelli A, Arduino C, Zonta A, Brusco A, and Retta SF
- Abstract
Cerebral Cavernous Malformation (CCM) is a cerebrovascular disease of genetic origin that predisposes to seizures, focal neurological deficits and fatal intracerebral hemorrhage. It may occur sporadically or in familial forms, segregating as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. Its pathogenesis has been associated with loss-of-function mutations in three genes, namely KRIT1 (CCM1), CCM2 and PDCD10 (CCM3), which are implicated in defense mechanisms against oxidative stress and inflammation. Herein, we screened 21 Italian CCM cases using clinical exome sequencing and found six cases (~29%) with pathogenic variants in CCM genes, including a large 145−256 kb genomic deletion spanning the KRIT1 gene and flanking regions, and the KRIT1 c.1664C>T variant, which we demonstrated to activate a donor splice site in exon 16. The segregation of this cryptic splicing mutation was studied in a large Italian family (five affected and seven unaffected cases), and showed a largely heterogeneous clinical presentation, suggesting the implication of genetic modifiers. Moreover, by analyzing ad hoc gene panels, including a virtual panel of 23 cerebrovascular disease-related genes (Cerebro panel), we found two variants in NOTCH3 and PTEN genes, which could contribute to the abnormal oxidative stress and inflammatory responses to date implicated in CCM disease pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
19. Polymorphisms in genes related to oxidative stress and inflammation: Emerging links with the pathogenesis and severity of Cerebral Cavernous Malformation disease.
- Author
-
Perrelli A and Retta SF
- Subjects
- Genome-Wide Association Study, Humans, Inflammation genetics, KRIT1 Protein genetics, Microtubule-Associated Proteins genetics, Mutation, Oxidative Stress genetics, Polymorphism, Genetic, Hemangioma, Cavernous, Central Nervous System genetics
- Abstract
Cerebral Cavernous Malformation (CCM) is a cerebrovascular disease of genetic origin affecting 0.5% of the population and characterized by abnormally enlarged and leaky capillaries that predispose to seizures, neurological deficits, and intracerebral hemorrhage (ICH). CCM occurs sporadically or is inherited as dominant condition with incomplete penetrance and highly variable expressivity. Three disease genes have been identified: KRIT1 (CCM1), CCM2 and CCM3. Previous results demonstrated that loss-of-function mutations of CCM genes cause pleiotropic effects, including defective autophagy, altered reactive oxygen species (ROS) homeostasis, and enhanced sensitivity to oxidative stress and inflammatory events, suggesting a novel unifying pathogenetic mechanism, and raising the possibility that CCM disease onset and severity are influenced by the presence of susceptibility and modifier genes. Consistently, genome-wide association studies (GWAS) in large and homogeneous cohorts of patients sharing the familial form of CCM disease and identical mutations in CCM genes have led to the discovery of distinct genetic modifiers of major disease severity phenotypes, such as development of numerous and large CCM lesions, and susceptibility to ICH. This review deals with the identification of genetic modifiers with a significant impact on inter-individual variability in CCM disease onset and severity, including highly polymorphic genes involved in oxidative stress, inflammatory and immune responses, such as cytochrome P450 monooxygenases (CYP), matrix metalloproteinases (MMP), and Toll-like receptors (TLR), pointing to their emerging prognostic value, and opening up new perspectives for risk stratification and personalized medicine strategies., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
20. Towards precision nanomedicine for cerebrovascular diseases with emphasis on Cerebral Cavernous Malformation (CCM).
- Author
-
Perrelli A, Fatehbasharzad P, Benedetti V, Ferraris C, Fontanella M, De Luca E, Moglianetti M, Battaglia L, and Retta SF
- Subjects
- Humans, Inflammation, Mutation, Nanomedicine, Cerebrovascular Disorders diagnosis, Cerebrovascular Disorders genetics, Cerebrovascular Disorders therapy, Hemangioma, Cavernous, Central Nervous System diagnosis, Hemangioma, Cavernous, Central Nervous System genetics, Hemangioma, Cavernous, Central Nervous System therapy
- Abstract
Introduction : Cerebrovascular diseases encompass various disorders of the brain vasculature, such as ischemic/hemorrhagic strokes, aneurysms, and vascular malformations, also affecting the central nervous system leading to a large variety of transient or permanent neurological disorders. They represent major causes of mortality and long-term disability worldwide, and some of them can be inherited, including Cerebral Cavernous Malformation (CCM), an autosomal dominant cerebrovascular disease linked to mutations in CCM1/KRIT1, CCM2, or CCM3/PDCD10 genes. Areas covered : Besides marked clinical and etiological heterogeneity, some commonalities are emerging among distinct cerebrovascular diseases, including key pathogenetic roles of oxidative stress and inflammation, which are increasingly recognized as major disease hallmarks and therapeutic targets. This review provides a comprehensive overview of the different clinical features and common pathogenetic determinants of cerebrovascular diseases, highlighting major challenges, including the pressing need for new diagnostic and therapeutic strategies, and focusing on emerging innovative features and promising benefits of nanomedicine strategies for early detection and targeted treatment of such diseases. Expert opinion : Specifically, we describe and discuss the multiple physico-chemical features and unique biological advantages of nanosystems, including nanodiagnostics, nanotherapeutics, and nanotheranostics, that may help improving diagnosis and treatment of cerebrovascular diseases and neurological comorbidities, with an emphasis on CCM disease.
- Published
- 2021
- Full Text
- View/download PDF
21. An international call for a new grading system for cerebral and cerebellar cavernomas.
- Author
-
Fontanella MM, Bacigaluppi S, Doglietto F, Zanin L, Agosti E, Panciani P, Belotti F, Saraceno G, Spena G, Draghi R, Fiorindi A, Cornali C, Biroli A, Kivelev J, Chiesa M, Retta SF, Gasparotti R, Kato Y, Hernesniemi J, and Rigamonti D
- Subjects
- Basal Ganglia, Humans, Middle Aged, Prospective Studies, Radiography, Retrospective Studies, Hemangioma, Cavernous, Central Nervous System diagnostic imaging, Hemangioma, Cavernous, Central Nervous System surgery
- Abstract
Surgical indications for cerebral cavernous malformations remain significantly center- and surgeon-dependent. Available grading systems are potentially limited, as they do not include epileptological and radiological data. A novel grading system is proposed for supratentorial and cerebellar cavernomas: it considers neuroradiological features (bleeding, increase in size), neurological status (focal deficits and seizures), location of the lesion and age of the patient. The score ranges from -1 to 10; furthermore, surgery should be considered when a score of 4 or higher is present. Based on neuroradiological characteristics, 0 points are assigned if the CCM is stable in size at different neuroradiological controls, 1 point if there is an increase in volume during follow-up, 2 points if intra- or extra-lesional bleeding <1 cm is present and 3 points if the CCM produced a hematoma >1 cm. Regarding focal neurological deficits, 0 points are assigned if absent and 2 points if present. For seizures, 0 points are assigned if absent, 1 point if present, but controlled by medications, and 2 points if drug resistant. We considered the site of the CCM, and in case of deep-seated lesions in a critical area (basal ganglia, thalamus) 1 point (-1) is subtracted, while for subcortical or deep cerebellar lesions 0 points are assigned, for CCMs in a cortical critical area 1 point is assigned and in case of lesions in cortical not in critical area or superficial cerebellar area, 2 points are assigned. As far as age is concerned, 0 points are assigned for patients older than 50 years and 1 point for patients younger than 50. In conclusion, a novel grading for surgical decision making in cerebral cavernomas, based on the experience of selected neurosurgeons, basic scientists, and patients, is suggested with the aim of further improving and standardizing the treatment of CCMs. The aim of this paper was also to call for both retrospective and prospective multicenter studies with the aim of testing the efficacy of the grading system in different centers.
- Published
- 2021
- Full Text
- View/download PDF
22. Protein kinase Cα regulates the nucleocytoplasmic shuttling of KRIT1.
- Author
-
De Luca E, Perrelli A, Swamy H, Nitti M, Passalacqua M, Furfaro AL, Salzano AM, Scaloni A, Glading AJ, and Retta SF
- Subjects
- HeLa Cells, Humans, Phosphorylation, Tetradecanoylphorbol Acetate, Active Transport, Cell Nucleus, KRIT1 Protein metabolism, Protein Kinase C-alpha genetics
- Abstract
KRIT1 is a scaffolding protein that regulates multiple molecular mechanisms, including cell-cell and cell-matrix adhesion, and redox homeostasis and signaling. However, rather little is known about how KRIT1 is itself regulated. KRIT1 is found in both the cytoplasm and the nucleus, yet the upstream signaling proteins and mechanisms that regulate KRIT1 nucleocytoplasmic shuttling are not well understood. Here, we identify a key role for protein kinase C (PKC) in this process. In particular, we found that PKC activation promotes the redox-dependent cytoplasmic localization of KRIT1, whereas inhibition of PKC or treatment with the antioxidant N-acetylcysteine leads to KRIT1 nuclear accumulation. Moreover, we demonstrated that the N-terminal region of KRIT1 is crucial for the ability of PKC to regulate KRIT1 nucleocytoplasmic shuttling, and may be a target for PKC-dependent regulatory phosphorylation events. Finally, we found that silencing of PKCα, but not PKCδ, inhibits phorbol 12-myristate 13-acetate (PMA)-induced cytoplasmic enrichment of KRIT1, suggesting a major role for PKCα in regulating KRIT1 nucleocytoplasmic shuttling. Overall, our findings identify PKCα as a novel regulator of KRIT1 subcellular compartmentalization, thus shedding new light on the physiopathological functions of this protein., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2021. Published by The Company of Biologists Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
23. Vitamin D Deficiency and the Risk of Cerebrovascular Disease.
- Author
-
Kim HA, Perrelli A, Ragni A, Retta F, De Silva TM, Sobey CG, and Retta SF
- Abstract
Vitamin D deficiency has been clearly linked to major chronic diseases associated with oxidative stress, inflammation, and aging, including cardiovascular and neurodegenerative diseases, diabetes, and cancer. In particular, the cardiovascular system appears to be highly sensitive to vitamin D deficiency, as this may result in endothelial dysfunction and vascular defects via multiple mechanisms. Accordingly, recent research developments have led to the proposal that pharmacological interventions targeting either vitamin D deficiency or its key downstream effects, including defective autophagy and abnormal pro-oxidant and pro-inflammatory responses, may be able to limit the onset and severity of major cerebrovascular diseases, such as stroke and cerebrovascular malformations. Here we review the available evidence supporting the role of vitamin D in preventing or limiting the development of these cerebrovascular diseases, which are leading causes of disability and death all over the world.
- Published
- 2020
- Full Text
- View/download PDF
24. KRIT1 loss-mediated upregulation of NOX1 in stromal cells promotes paracrine pro-angiogenic responses.
- Author
-
Finetti F, Schiavo I, Ercoli J, Zotta A, Boda E, Retta SF, and Trabalzini L
- Subjects
- Animals, Antigens, CD metabolism, Cadherins metabolism, Cell Movement drug effects, Cell Proliferation drug effects, Culture Media, Conditioned pharmacology, Cyclooxygenase 2 metabolism, Dinoprostone biosynthesis, Fibroblasts drug effects, Fibroblasts metabolism, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells metabolism, Humans, Matrix Metalloproteinase 2 metabolism, Mice, Knockout, Stromal Cells drug effects, Stromal Cells metabolism, Vascular Endothelial Growth Factor A metabolism, KRIT1 Protein metabolism, NADPH Oxidase 1 metabolism, Neovascularization, Physiologic drug effects, Paracrine Communication drug effects, Up-Regulation drug effects
- 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., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
25. Dicarbonyl Stress and S-Glutathionylation in Cerebrovascular Diseases: A Focus on Cerebral Cavernous Malformations.
- Author
-
Antognelli C, Perrelli A, Armeni T, Nicola Talesa V, and Retta SF
- Abstract
Dicarbonyl stress is a dysfunctional state consisting in the abnormal accumulation of reactive α-oxaldehydes leading to increased protein modification. In cells, post-translational changes can also occur through S-glutathionylation, a highly conserved oxidative post-translational modification consisting of the formation of a mixed disulfide between glutathione and a protein cysteine residue. This review recapitulates the main findings supporting a role for dicarbonyl stress and S-glutathionylation in the pathogenesis of cerebrovascular diseases, with specific emphasis on cerebral cavernous malformations (CCM), a vascular disease of proven genetic origin that may give rise to various clinical signs and symptoms at any age, including recurrent headaches, seizures, focal neurological deficits, and intracerebral hemorrhage. A possible interplay between dicarbonyl stress and S-glutathionylation in CCM is also discussed.
- Published
- 2020
- Full Text
- View/download PDF
26. Intracellular Antioxidant Activity of Biocompatible Citrate-Capped Palladium Nanozymes.
- Author
-
Moglianetti M, Pedone D, Udayan G, Retta SF, Debellis D, Marotta R, Turco A, Rella S, Malitesta C, Bonacucina G, De Luca E, and Pompa PP
- Abstract
A method for the aqueous synthesis of stable and biocompatible citrate-coated palladium nanoparticles (PdNPs) in the size range comparable to natural enzymes (4-8 nm) has been developed. The toxicological profile of PdNPs was assessed by different assays on several cell lines demonstrating their safety in vitro also at high particle concentrations. To elucidate their cellular fate upon uptake, the localization of PdNPs was analyzed by Transmission Electron Microscopy (TEM). Moreover, crucial information about their intracellular stability and oxidation state was obtained by Sputtering-Enabled Intracellular X-ray Photoelectron Spectroscopy (SEI-XPS). TEM/XPS results showed significant stability of PdNPs in the cellular environment, an important feature for their biocompatibility and potential for biomedical applications. On the catalytic side, these PdNPs exhibited strong and broad antioxidant activities, being able to mimic the three main antioxidant cellular enzymes, i.e., peroxidase, catalase, and superoxide dismutase. Remarkably, using an experimental model of a human oxidative stress-related disease, we demonstrated the effectiveness of PdNPs as antioxidant nanozymes within the cellular environment, showing that they are able to completely re-establish the physiological Reactive Oxygen Species (ROS) levels in highly compromised intracellular redox conditions.
- Published
- 2020
- Full Text
- View/download PDF
27. Study of CCM Microvascular Endothelial Phenotype by an In Vitro Tubule Differentiation Model.
- Author
-
Delle Monache S and Retta SF
- Subjects
- Cell Culture Techniques, Cells, Cultured, Hemangioma, Cavernous, Central Nervous System etiology, Humans, KRIT1 Protein genetics, KRIT1 Protein metabolism, Endothelial Cells metabolism, Endothelium, Vascular metabolism, Hemangioma, Cavernous, Central Nervous System metabolism, Microvessels metabolism, Neovascularization, Pathologic metabolism, Phenotype
- Abstract
Cerebral cavernous malformation (CCM) proteins play critical roles for endothelial cell functions, including cytoskeletal remodeling, cell-cell interactions, cell polarity, tube formation, and angiogenesis. It has been shown that the mutation of even one of the CCM genes involved in CCMs can determine an alteration in the angiogenesis process, but the precise mechanism is yet to be clarified.Here using a model of cerebral microvascular endothelial cells (hBMEC) transiently silenced by CCM1, we tried to mimic the physiological conditions that occur in the presence of CCM1 gene know-down evaluating their ability to form tube structures through an in vitro angiogenesis assay.
- Published
- 2020
- Full Text
- View/download PDF
28. Detection of p62/SQSTM1 Aggregates in Cellular Models of CCM Disease by Immunofluorescence.
- Author
-
Marchi S, Retta SF, and Pinton P
- Subjects
- Autophagy genetics, Biomarkers, Cells, Cultured, Disease Susceptibility, Endothelial Cells metabolism, Fluorescent Antibody Technique, Hemangioma, Cavernous, Central Nervous System etiology, Hemangioma, Cavernous, Central Nervous System pathology, Humans, Microtubule-Associated Proteins metabolism, Mutation, Hemangioma, Cavernous, Central Nervous System metabolism, Protein Aggregates, Protein Aggregation, Pathological metabolism, Sequestosome-1 Protein metabolism
- Abstract
Cerebral cavernous malformations (CCM) is a familial or sporadic rare disorder that is characterized by capillary vascular lesions with a mulberry-like appearance on MRI scans. Three distinct genes have been associated to CCM disease, known as CCM1/KRIT1, CCM2/MGC4607, and CCM3/PDCD10. Loss-of-functions mutations on these genes lead to deregulation in multiple signaling pathways, thereby resulting in disturbed vessel organization and function. Insufficient autophagy has been observed upon downregulation of all three CCM genes, both in cells and human patient tissues, revealed as aberrant accumulation of the autophagy receptor p62/SQSTM1. The autophagic process is conceived as an adaptive response to stress and is essential for the maintenance of cellular homeostasis. The aim of this review is to briefly summarize the current knowledge on the role of autophagy in CCM disease and to furnish a detailed protocol for detecting and measuring p62/SQSTM1 cytoplasmic aggregates through immunofluorescence technique.
- Published
- 2020
- Full Text
- View/download PDF
29. Generation of CCM Phenotype by a Human Microvascular Endothelial Model.
- Author
-
Delle Monache S and Retta SF
- Subjects
- Biomarkers, Cell Culture Techniques, Disease Susceptibility, Endothelial Cells metabolism, Gene Expression, Gene Knockdown Techniques, Hemangioma, Cavernous, Central Nervous System etiology, Hemangioma, Cavernous, Central Nervous System metabolism, Humans, KRIT1 Protein genetics, KRIT1 Protein metabolism, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, RNA, Small Interfering genetics, Transfection, Endothelium, Vascular metabolism, Hemangioma, Cavernous, Central Nervous System diagnosis, Phenotype
- Abstract
Cerebral cavernous malformations (CCMs) is a disorder of endothelial cells predominantly localized in the brain. Although a complete inactivation of each CCM protein has been found in the affected endothelium of diseased patients and a necessary and additional role of microenvironment has been demonstrated to determine in vivo the occurrence of vascular lesions, a microvascular endothelial model based on knockdown of a CCM gene represents today a convenient method to study some of critical signaling events regulating pathogenesis of CCM. For these reasons, in our laboratory we developed a microvascular cerebral endothelial model of Krit1 deficiency performing silencing experiments of CCM1 gene (Krit1) with siRNA procedure.
- Published
- 2020
- Full Text
- View/download PDF
30. Next Generation Sequencing (NGS) Strategies for Genetic Testing of Cerebral Cavernous Malformation (CCM) Disease.
- Author
-
Benedetti V, Pellegrino E, Brusco A, Piva R, and Retta SF
- Subjects
- Alleles, Computational Biology methods, DNA Copy Number Variations, Disease Management, Genomics, Humans, Microtubule-Associated Proteins genetics, Mutation, Pedigree, Phenotype, Exome Sequencing, Genetic Association Studies methods, Genetic Predisposition to Disease, Genetic Testing methods, Hemangioma, Cavernous, Central Nervous System diagnosis, Hemangioma, Cavernous, Central Nervous System genetics, High-Throughput Nucleotide Sequencing methods
- Abstract
The application of next generation sequencing (NGS) technique has a great impact on complex disease studies. Indeed, genetic heterogeneity, phenotypic variability, and disease rarity are all factors that make the traditional diagnostic approach to genetic disorders, whereby a specific gene is selected for sequencing based on the clinical phenotype, very challenging and obsolete.Exome sequencing, which sequences the protein-coding region of the genome, has been rapidly applied to variant discovery in research settings. Recent coverage and accuracy improvements have accelerated the development of clinical exome sequencing (CES) platforms targeting disease-related genes and enabling variant identification in patients with suspected genetic diseases. Nowadays, CES is rapidly becoming the diagnostic test of choice in patients with suspected Mendelian diseases, especially for those with heterogeneous etiology and clinical presentation. Reporting large CES series can improve guidelines on best practices for test utilization, and a better variant interpretation through clinically oriented data sharing.Herein, we suggest a feasible CES procedure for the genetic testing of Cerebral Cavernous Malformation (CCM) disease, including proband identification, library preparation, data analysis, and variant interpretation.
- Published
- 2020
- Full Text
- View/download PDF
31. Fluorescence Analysis of Reactive Oxygen Species (ROS) in Cellular Models of Cerebral Cavernous Malformation Disease.
- Author
-
Perrelli A and Retta SF
- Subjects
- Animals, Biomarkers, Cell Line, Hemangioma, Cavernous, Central Nervous System etiology, Hemangioma, Cavernous, Central Nervous System pathology, Humans, Mice, Microscopy, Fluorescence, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Mitochondria metabolism, Oxidation-Reduction, Oxidative Stress genetics, Superoxides metabolism, Fluorescent Antibody Technique methods, Hemangioma, Cavernous, Central Nervous System metabolism, Reactive Oxygen Species metabolism
- Abstract
Cerebral cavernous malformation (CCM) is a vascular disease of proven genetic origin, which may arise sporadically or can be inherited as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. CCM disease exhibits a range of different phenotypes, including wide interindividual differences in lesion number, size, and susceptibility to intracerebral hemorrhage (ICH). Mutations of the KRIT1 gene account for over 50% of familial cases. Previously, we demonstrated that KRIT1 loss-of-function is associated with altered homeostasis of intracellular reactive oxygen species (ROS) and abnormal activation of redox-sensitive transcription factors, which collectively result in pro-oxidative, pro-inflammatory, and pro-angiogenic effects, suggesting a novel pathogenic mechanism for CCM disease. Consistently, these original discoveries have been confirmed and extended by subsequent findings showing mechanistic relationships between pleiotropic redox-dependent effects of KRIT1 loss-of-function and enhanced cell sensitivity to oxidative stress, which may eventually lead to cellular dysfunctions and CCM disease pathogenesis. In this chapter, we describe few basic methods used for qualitative and quantitative analysis of intracellular ROS in cellular models of CCM disease.
- Published
- 2020
- Full Text
- View/download PDF
32. Production of KRIT1-knockout and KRIT1-knockin Mouse Embryonic Fibroblasts as Cellular Models of CCM Disease.
- Author
-
Goitre L, Fornelli C, Zotta A, Perrelli A, and Retta SF
- Subjects
- Animals, Disease Models, Animal, Gene Order, Gene Targeting, Genetic Loci, Genetic Vectors genetics, Hemangioma, Cavernous, Central Nervous System diagnosis, Homologous Recombination, Homozygote, Mice, Mice, Knockout, Mice, Transgenic, Transduction, Genetic, Fibroblasts metabolism, Hemangioma, Cavernous, Central Nervous System genetics, Hemangioma, Cavernous, Central Nervous System metabolism, KRIT1 Protein genetics
- Abstract
The development of distinct cellular and animal models has allowed the identification and characterization of molecular mechanisms underlying the pathogenesis of cerebral cavernous malformation (CCM) disease. This is a major cerebrovascular disorder of proven genetic origin, affecting 0.5% of the population. Three disease genes have been identified: CCM1/KRIT1, CCM2, and CCM3. These genes encode for proteins implicated in the regulation of major cellular structures and mechanisms, such as cell-cell and cell-matrix adhesion, actin cytoskeleton dynamics, and endothelial-to-mesenchymal transition, suggesting that they may act as pleiotropic regulators of cellular homeostasis. Indeed, accumulated evidence in cellular and animal models demonstrates that emerged pleiotropic functions of CCM proteins are mainly due to their ability to modulate redox-sensitive pathways and mechanisms involved in adaptive responses to oxidative stress and inflammation, thus contributing to the preservation of cellular homeostasis and stress defenses. In particular, we demonstrated that KRIT1 loss-of-function affects master regulators of cellular redox homeostasis and responses to oxidative stress, including major redox-sensitive transcriptional factors and antioxidant proteins, and autophagy, suggesting that altered redox signaling and oxidative stress contribute to CCM pathogenesis, and opening novel preventive and therapeutic perspectives.In this chapter, we describe materials and methods for isolation of mouse embryonic fibroblast (MEF) cells from homozygous KRIT1-knockout mouse embryos, and their transduction with a lentiviral vector encoding KRIT1 to generate cellular models of CCM disease that contributed significantly to the identification of pathogenetic mechanisms.
- Published
- 2020
- Full Text
- View/download PDF
33. Spectrophotometric Method for Determining Glyoxalase 1 Activity in Cerebral Cavernous Malformation (CCM) Disease.
- Author
-
Antognelli C, Talesa VN, and Retta SF
- Subjects
- Animals, Enzyme Activation, Fibroblasts enzymology, Hemangioma, Cavernous, Central Nervous System etiology, Humans, KRIT1 Protein genetics, KRIT1 Protein metabolism, Lactoylglutathione Lyase chemistry, Mice, Oxidative Stress, Hemangioma, Cavernous, Central Nervous System enzymology, Lactoylglutathione Lyase metabolism, Spectrophotometry methods
- Abstract
Glyoxalase 1 (Glo1) is a glutathione (GSH)-dependent enzyme that catalyzes the isomerization of the hemithioacetal formed non-enzymatically from methylglyoxal (MG) and GSH to S-D-lactoylglutathione (SLG). The activity of Glo1 is measured spectrophotometrically by following the increase of absorbance at 240 nm and 25 °C, attributable to the formation of SLG. The hemithioacetal is preformed by incubation of 2 mM MG and 1 mM GSH in 0.1 M sodium phosphate buffer (PBS) pH 7.2, at 25 °C for 10 min. The cell extract is then added, and the A
240 is monitored over 5-min incubation against correction for blank. Glo1 activity is given in units per mg of protein where one unit activity is defined as 1 μmole of SLG produced per min under assay conditions. Here, we describe measurement of Glo1 activity in established cellular models of cerebral cavernous malformation (CCM) disease, including KRIT1-knockout mouse embryonic fibroblast (MEF) and KRIT1-silenced human brain microvascular endothelial (hBMEC) cells.- Published
- 2020
- Full Text
- View/download PDF
34. From Genes and Mechanisms to Molecular-Targeted Therapies: The Long Climb to the Cure of Cerebral Cavernous Malformation (CCM) Disease.
- Author
-
Retta SF, Perrelli A, Trabalzini L, and Finetti F
- Subjects
- Alleles, Animals, Biomarkers, Disease Management, Disease Models, Animal, Hemangioma, Cavernous, Central Nervous System diagnosis, Hemangioma, Cavernous, Central Nervous System metabolism, Humans, Magnetic Resonance Imaging, Microtubule-Associated Proteins chemistry, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Mutation, Signal Transduction drug effects, Structure-Activity Relationship, Genetic Association Studies methods, Genetic Predisposition to Disease, Hemangioma, Cavernous, Central Nervous System genetics, Hemangioma, Cavernous, Central Nervous System therapy, Molecular Targeted Therapy
- Abstract
Cerebral cavernous malformation (CCM) is a rare cerebrovascular disorder of genetic origin consisting of closely clustered, abnormally dilated and leaky capillaries (CCM lesions), which occur predominantly in the central nervous system. CCM lesions can be single or multiple and may result in severe clinical symptoms, including focal neurological deficits, seizures, and intracerebral hemorrhage. Early human genetic studies demonstrated that CCM disease is linked to three chromosomal loci and can be inherited as autosomal dominant condition with incomplete penetrance and highly variable expressivity, eventually leading to the identification of three disease genes, CCM1/KRIT1, CCM2, and CCM3/PDCD10, which encode for structurally unrelated intracellular proteins that lack catalytic domains. Biochemical, molecular, and cellular studies then showed that these proteins are involved in endothelial cell-cell junction and blood-brain barrier stability maintenance through the regulation of major cellular structures and mechanisms, including endothelial cell-cell and cell-matrix adhesion, actin cytoskeleton dynamics, autophagy, and endothelial-to-mesenchymal transition, suggesting that they act as pleiotropic regulators of cellular homeostasis, and opening novel therapeutic perspectives. Indeed, accumulated evidence in cellular and animal models has eventually revealed that the emerged pleiotropic functions of CCM proteins are mainly due to their ability to modulate redox-sensitive pathways and mechanisms involved in adaptive responses to oxidative stress and inflammation, thus contributing to the preservation of cellular homeostasis and stress defenses.In this introductory review, we present a general overview of 20 years of amazing progress in the identification of genetic culprits and molecular mechanisms underlying CCM disease pathogenesis, and the development of targeted therapeutic strategies.
- Published
- 2020
- Full Text
- View/download PDF
35. KRIT1 Deficiency Promotes Aortic Endothelial Dysfunction.
- Author
-
Vieceli Dalla Sega F, Mastrocola R, Aquila G, Fortini F, Fornelli C, Zotta A, Cento AS, Perrelli A, Boda E, Pannuti A, Marchi S, Pinton P, Ferrari R, Rizzo P, and Retta SF
- Subjects
- Animals, Aorta pathology, Apoptosis, Atherosclerosis metabolism, Endothelium, Vascular pathology, Human Umbilical Vein Endothelial Cells metabolism, Humans, Intercellular Adhesion Molecule-1 metabolism, KRIT1 Protein deficiency, KRIT1 Protein metabolism, Lipid Metabolism, Mice, Mice, Inbred C57BL, Oxidative Stress, Receptor, Notch1 metabolism, Tumor Necrosis Factor-alpha metabolism, Vascular Cell Adhesion Molecule-1 metabolism, Aorta metabolism, Atherosclerosis genetics, Endothelium, Vascular metabolism, KRIT1 Protein genetics, Loss of Function Mutation
- Abstract
Loss-of-function mutations of the gene encoding Krev interaction trapped protein 1 (KRIT1) are associated with the pathogenesis of Cerebral Cavernous Malformation (CCM), a major cerebrovascular disease characterized by abnormally enlarged and leaky capillaries and affecting 0.5% of the human population. However, growing evidence demonstrates that KRIT1 is implicated in the modulation of major redox-sensitive signaling pathways and mechanisms involved in adaptive responses to oxidative stress and inflammation, suggesting that its loss-of-function mutations may have pathological effects not limited to CCM disease. The aim of this study was to address whether KRIT1 loss-of-function predisposes to the development of pathological conditions associated with enhanced endothelial cell susceptibility to oxidative stress and inflammation, such as arterial endothelial dysfunction (ED) and atherosclerosis. Silencing of KRIT1 in human aortic endothelial cells (HAECs), coronary artery endothelial cells (HCAECs), and umbilical vein endothelial cells (HUVECs) resulted in increased expression of endothelial proinflammatory adhesion molecules vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) and in enhanced susceptibility to tumor necrosis factor alpha (TNF-α)-induced apoptosis. These effects were associated with a downregulation of Notch1 activation that could be rescued by antioxidant treatment, suggesting that they are consequent to altered intracellular redox homeostasis induced by KRIT1 loss-of-function. Furthermore, analysis of the aorta of heterozygous KRIT1
+/- mice fed a high-fructose diet to induce systemic oxidative stress and inflammation demonstrated a 1.6-fold increased expression of VCAM-1 and an approximately 2-fold enhanced fat accumulation (7.5% vs 3.6%) in atherosclerosis-prone regions, including the aortic arch and aortic root, as compared to corresponding wild-type littermates. In conclusion, we found that KRIT1 deficiency promotes ED, suggesting that, besides CCM, KRIT1 may be implicated in genetic susceptibility to the development of atherosclerotic lesions.- Published
- 2019
- Full Text
- View/download PDF
36. KRIT1 Loss-Of-Function Associated with Cerebral Cavernous Malformation Disease Leads to Enhanced S -Glutathionylation of Distinct Structural and Regulatory Proteins.
- Author
-
Cianfruglia L, Perrelli A, Fornelli C, Magini A, Gorbi S, Salzano AM, Antognelli C, Retta F, Benedetti V, Cassoni P, Emiliani C, Principato G, Scaloni A, Armeni T, and Retta SF
- Abstract
Loss-of-function mutations in the KRIT1 gene are associated with the pathogenesis of cerebral cavernous malformations (CCMs), a major cerebrovascular disease still awaiting therapies. Accumulating evidence demonstrates that KRIT1 plays an important role in major redox-sensitive mechanisms, including transcriptional pathways and autophagy, which play major roles in cellular homeostasis and defense against oxidative stress, raising the possibility that KRIT1 loss has pleiotropic effects on multiple redox-sensitive systems. Using previously established cellular models, we found that KRIT1 loss-of-function affects the glutathione (GSH) redox system, causing a significant decrease in total GSH levels and increase in oxidized glutathione disulfide (GSSG), with a consequent deficit in the GSH/GSSG redox ratio and GSH-mediated antioxidant capacity. Redox proteomic analyses showed that these effects are associated with increased S -glutathionylation of distinct proteins involved in adaptive responses to oxidative stress, including redox-sensitive chaperonins, metabolic enzymes, and cytoskeletal proteins, suggesting a novel molecular signature of KRIT1 loss-of-function. Besides providing further insights into the emerging pleiotropic functions of KRIT1, these findings point definitively to KRIT1 as a major player in redox biology, shedding new light on the mechanistic relationship between KRIT1 loss-of-function and enhanced cell sensitivity to oxidative stress, which may eventually lead to cellular dysfunctions and CCM disease pathogenesis.
- Published
- 2019
- Full Text
- View/download PDF
37. Multifunctional Platinum@BSA-Rapamycin Nanocarriers for the Combinatorial Therapy of Cerebral Cavernous Malformation.
- Author
-
De Luca E, Pedone D, Moglianetti M, Pulcini D, Perrelli A, Retta SF, and Pompa PP
- Abstract
Platinum nanoparticles (PtNPs) are antioxidant enzyme-mimetic nanomaterials with significant potential for the treatment of complex diseases related to oxidative stress. Among such diseases, Cerebral Cavernous Malformation (CCM) is a major cerebrovascular disorder of genetic origin, which affects at least 0.5% of the general population. Accumulated evidence indicates that loss-of-function mutations of the three known CCM genes predispose endothelial cells to oxidative stress-mediated dysfunctions by affecting distinct redox-sensitive signaling pathways and mechanisms, including pro-oxidant and antioxidant pathways and autophagy. A multitargeted combinatorial therapy might thereby represent a promising strategy for the effective treatment of this disease. Herein, we developed a multifunctional nanocarrier by combining the radical scavenging activity of PtNPs with the autophagy-stimulating activity of rapamycin (Rapa). Our results show that the combinatorial targeting of redox signaling and autophagy dysfunctions is effective in rescuing major molecular and cellular hallmarks of CCM disease, suggesting its potential for the treatment of this and other oxidative stress-related diseases., Competing Interests: The authors declare no competing financial interest.
- Published
- 2018
- Full Text
- View/download PDF
38. Yeast-Derived Recombinant Avenanthramides Inhibit Proliferation, Migration and Epithelial Mesenchymal Transition of Colon Cancer Cells.
- Author
-
Finetti F, Moglia A, Schiavo I, Donnini S, Berta GN, Di Scipio F, Perrelli A, Fornelli C, Trabalzini L, and Retta SF
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma pathology, Antineoplastic Agents isolation & purification, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cyclin-Dependent Kinase Inhibitor p27 metabolism, HT29 Cells, Humans, Neoplasm Invasiveness, Signal Transduction drug effects, Tumor Suppressor Protein p53 metabolism, ortho-Aminobenzoates isolation & purification, Adenocarcinoma drug therapy, Antineoplastic Agents pharmacology, Cell Movement drug effects, Cell Proliferation drug effects, Colorectal Neoplasms drug therapy, Epithelial-Mesenchymal Transition drug effects, Saccharomyces cerevisiae metabolism, ortho-Aminobenzoates pharmacology
- Abstract
Avenanthramides (Avns), polyphenols found exclusively in oats, are emerging as promising therapeutic candidates for the treatment of several human diseases, including colon cancer. By engineering a Saccharomyces cerevisiae strain, we previously produced two novel phenolic compounds, N -( E )- p -coumaroyl-3-hydroxyanthranilic acid (Yeast avenanthramide I, YAvnI) and N -( E )-caffeoyl-3-hydroxyanthranilic acid (Yeast avenanthramide II, YAvnII), which are endowed with a structural similarity to bioactive oat avenanthramides and stronger antioxidant properties. In this study, we evaluated the ability of these yeast-derived recombinant avenanthramides to inhibit major hallmarks of colon cancer cells, including sustained proliferation, migration and epithelial-mesenchymal transition (EMT). Using the human colon adenocarcinoma cell line HT29, we compared the impact of YAvns and natural Avns, including Avn-A and Avn-C, on colon cancer cells by performing MTT, clonogenic, adhesion, migration, and anchorage-independent growth assays, and analyzing the expression of EMT markers. We found that both YAvns and Avns were able to inhibit colon cancer cell growth by increasing the expression of p21, p27 and p53 proteins. However, YAvns resulted more effective than natural compounds in inhibiting cancer cell migration and reverting major molecular features of the EMT process, including the down-regulation of E-cadherin mRNA and protein levels.
- Published
- 2018
- Full Text
- View/download PDF
39. Biological Activities, Health Benefits, and Therapeutic Properties of Avenanthramides: From Skin Protection to Prevention and Treatment of Cerebrovascular Diseases.
- Author
-
Perrelli A, Goitre L, Salzano AM, Moglia A, Scaloni A, and Retta SF
- Subjects
- Animals, Anti-Inflammatory Agents pharmacology, Antioxidants pharmacology, Humans, Protective Agents chemistry, Protective Agents pharmacology, ortho-Aminobenzoates chemistry, ortho-Aminobenzoates pharmacology, Cerebrovascular Disorders drug therapy, Cerebrovascular Disorders prevention & control, Protective Agents therapeutic use, Skin drug effects, ortho-Aminobenzoates therapeutic use
- Abstract
Oat ( Avena sativa ) is a cereal known since antiquity as a useful grain with abundant nutritional and health benefits. It contains distinct molecular components with high antioxidant activity, such as tocopherols, tocotrienols, and flavanoids. In addition, it is a unique source of avenanthramides, phenolic amides containing anthranilic acid and hydroxycinnamic acid moieties, and endowed with major beneficial health properties because of their antioxidant, anti-inflammatory, and antiproliferative effects. In this review, we report on the biological activities of avenanthramides and their derivatives, including analogs produced in recombinant yeast, with a major focus on the therapeutic potential of these secondary metabolites in the treatment of aging-related human diseases. Moreover, we also present recent advances pointing to avenanthramides as interesting therapeutic candidates for the treatment of cerebral cavernous malformation (CCM) disease, a major cerebrovascular disorder affecting up to 0.5% of the human population. Finally, we highlight the potential of foodomics and redox proteomics approaches in outlining distinctive molecular pathways and redox protein modifications associated with avenanthramide bioactivities in promoting human health and contrasting the onset and progression of various pathologies. The paper is dedicated to the memory of Adelia Frison.
- Published
- 2018
- Full Text
- View/download PDF
40. KRIT1 loss-of-function induces a chronic Nrf2-mediated adaptive homeostasis that sensitizes cells to oxidative stress: Implication for Cerebral Cavernous Malformation disease.
- Author
-
Antognelli C, Trapani E, Delle Monache S, Perrelli A, Daga M, Pizzimenti S, Barrera G, Cassoni P, Angelucci A, Trabalzini L, Talesa VN, Goitre L, and Retta SF
- Subjects
- Animals, Apoptosis, Autophagy genetics, Cells, Cultured, Central Nervous System Neoplasms metabolism, HSP27 Heat-Shock Proteins metabolism, HSP70 Heat-Shock Proteins metabolism, Hemangioma, Cavernous, Central Nervous System metabolism, Homeostasis, Humans, KRIT1 Protein metabolism, Lactoylglutathione Lyase metabolism, Mice, Mice, Knockout, NF-E2-Related Factor 2 metabolism, Oxidation-Reduction, Protein Processing, Post-Translational, Pyruvaldehyde metabolism, Brain pathology, Central Nervous System Neoplasms genetics, Endothelial Cells physiology, Hemangioma, Cavernous, Central Nervous System genetics, KRIT1 Protein genetics, Mutation genetics, Oxidative Stress genetics
- Abstract
KRIT1 (CCM1) is a disease gene responsible for Cerebral Cavernous Malformations (CCM), a major cerebrovascular disease of proven genetic origin affecting 0.3-0.5% of the population. Previously, we demonstrated that KRIT1 loss-of-function is associated with altered redox homeostasis and abnormal activation of the redox-sensitive transcription factor c-Jun, which collectively result in pro-oxidative, pro-inflammatory and pro-angiogenic effects, suggesting a novel pathogenic mechanism for CCM disease and raising the possibility that KRIT1 loss-of-function exerts pleiotropic effects on multiple redox-sensitive mechanisms. To address this possibility, we investigated major redox-sensitive pathways and enzymatic systems that play critical roles in fundamental cytoprotective mechanisms of adaptive responses to oxidative stress, including the master Nrf2 antioxidant defense pathway and its downstream target Glyoxalase 1 (Glo1), a pivotal stress-responsive defense enzyme involved in cellular protection against glycative and oxidative stress through the metabolism of methylglyoxal (MG). This is a potent post-translational protein modifier that may either contribute to increased oxidative molecular damage and cellular susceptibility to apoptosis, or enhance the activity of major apoptosis-protective proteins, including heat shock proteins (Hsps), promoting cell survival. Experimental outcomes showed that KRIT1 loss-of-function induces a redox-sensitive sustained upregulation of Nrf2 and Glo1, and a drop in intracellular levels of MG-modified Hsp70 and Hsp27 proteins, leading to a chronic adaptive redox homeostasis that counteracts intrinsic oxidative stress but increases susceptibility to oxidative DNA damage and apoptosis, sensitizing cells to further oxidative challenges. While supporting and extending the pleiotropic functions of KRIT1, these findings shed new light on the mechanistic relationship between KRIT1 loss-of-function and enhanced cell predisposition to oxidative damage, thus providing valuable new insights into CCM pathogenesis and novel options for the development of preventive and therapeutic strategies., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
41. Data in support of sustained upregulation of adaptive redox homeostasis mechanisms caused by KRIT1 loss-of-function.
- Author
-
Antognelli C, Trapani E, Delle Monache S, Perrelli A, Fornelli C, Retta F, Cassoni P, Talesa VN, and Retta SF
- Abstract
This article contains additional data related to the original research article entitled "KRIT1 loss-of-function induces a chronic Nrf2-mediated adaptive homeostasis that sensitizes cells to oxidative stress: implication for Cerebral Cavernous Malformation disease" (Antognelli et al., 2017) [1]. Data were obtained by si-RNA-mediated gene silencing, qRT-PCR, immunoblotting, and immunohistochemistry studies, and enzymatic activity and apoptosis assays. Overall, they support, complement and extend original findings demonstrating that KRIT1 loss-of-function induces a redox-sensitive and JNK-dependent sustained upregulation of the master Nrf2 antioxidant defense pathway and its downstream target Glyoxalase 1 (Glo1), and a drop in intracellular levels of AP-modified Hsp70 and Hsp27 proteins, leading to a chronic adaptive redox homeostasis that sensitizes cells to oxidative DNA damage and apoptosis. In particular, immunoblotting analyses of Nrf2, Glo1, AP-modified Hsp70 and Hsp27 proteins, HO-1, phospho-c-Jun, phospho-ERK5, and KLF4 expression levels were performed both in KRIT1-knockout MEF cells and in KRIT1-silenced human brain microvascular endothelial cells (hBMEC) treated with the antioxidant Tiron, and compared with control cells. Moreover, immunohistochemistry analysis of Nrf2, Glo1, phospho-JNK, and KLF4 was performed on histological samples of human CCM lesions. Finally, the role of Glo1 in the downregulation of AP-modified Hsp70 and Hsp27 proteins, and the increase in apoptosis susceptibility associated with KRIT1 loss-of-function was addressed by si-RNA-mediated Glo1 gene silencing in KRIT1-knockout MEF cells.
- Published
- 2017
- Full Text
- View/download PDF
42. Up-regulation of NADPH oxidase-mediated redox signaling contributes to the loss of barrier function in KRIT1 deficient endothelium.
- Author
-
Goitre L, DiStefano PV, Moglia A, Nobiletti N, Baldini E, Trabalzini L, Keubel J, Trapani E, Shuvaev VV, Muzykantov VR, Sarelius IH, Retta SF, and Glading AJ
- Subjects
- Animals, Antioxidants metabolism, Capillary Permeability drug effects, Capillary Permeability genetics, Gene Expression Regulation, KRIT1 Protein genetics, KRIT1 Protein metabolism, Mice, Mice, Knockout, NADPH Oxidase 4 genetics, NADPH Oxidase 4 metabolism, NF-kappa B metabolism, Reactive Oxygen Species metabolism, Tumor Necrosis Factor-alpha metabolism, Endothelium metabolism, KRIT1 Protein deficiency, NADPH Oxidases metabolism, Oxidation-Reduction, Signal Transduction
- Abstract
The intracellular scaffold KRIT1/CCM1 is an established regulator of vascular barrier function. Loss of KRIT1 leads to decreased microvessel barrier function and to the development of the vascular disorder Cerebral Cavernous Malformation (CCM). However, how loss of KRIT1 causes the subsequent deficit in barrier function remains undefined. Previous studies have shown that loss of KRIT1 increases the production of reactive oxygen species (ROS) and exacerbates vascular permeability triggered by several inflammatory stimuli, but not TNF-α. We now show that endothelial ROS production directly contributes to the loss of barrier function in KRIT1 deficient animals and cells, as targeted antioxidant enzymes reversed the increase in permeability in KRIT1 heterozygous mice as shown by intravital microscopy. Rescue of the redox state restored responsiveness to TNF-α in KRIT1 deficient arterioles, but not venules. In vitro, KRIT1 depletion increased endothelial ROS production via NADPH oxidase signaling, up-regulated Nox4 expression, and promoted NF-κB dependent promoter activity. Recombinant yeast avenanthramide I, an antioxidant and inhibitor of NF-κB signaling, rescued barrier function in KRIT1 deficient cells. However, KRIT1 depletion blunted ROS production in response to TNF-α. Together, our data indicate that ROS signaling is critical for the loss of barrier function following genetic deletion of KRIT1.
- Published
- 2017
- Full Text
- View/download PDF
43. Oxidative stress and inflammation in cerebral cavernous malformation disease pathogenesis: Two sides of the same coin.
- Author
-
Retta SF and Glading AJ
- Subjects
- Humans, Neovascularization, Pathologic genetics, Neovascularization, Pathologic physiopathology, Hemangioma, Cavernous, Central Nervous System physiopathology, Inflammation, Oxidative Stress
- Abstract
Cerebral Cavernous Malformation (CCM) is a vascular disease of proven genetic origin, which may arise sporadically or is inherited as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. CCM lesions exhibit a range of different phenotypes, including wide inter-individual differences in lesion number, size, and susceptibility to intracerebral hemorrhage (ICH). Lesions may remain asymptomatic or result in pathological conditions of various type and severity at any age, with symptoms ranging from recurrent headaches to severe neurological deficits, seizures, and stroke. To date there are no direct therapeutic approaches for CCM disease besides the surgical removal of accessible lesions. Novel pharmacological strategies are particularly needed to limit disease progression and severity and prevent de novo formation of CCM lesions in susceptible individuals. Useful insights into innovative approaches for CCM disease prevention and treatment are emerging from a growing understanding of the biological functions of the three known CCM proteins, CCM1/KRIT1, CCM2 and CCM3/PDCD10. In particular, accumulating evidence indicates that these proteins play major roles in distinct signaling pathways, including those involved in cellular responses to oxidative stress, inflammation and angiogenesis, pointing to pathophysiological mechanisms whereby the function of CCM proteins may be relevant in preventing vascular dysfunctions triggered by these events. Indeed, emerging findings demonstrate that the pleiotropic roles of CCM proteins reflect their critical capacity to modulate the fine-tuned crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses, providing a novel mechanistic scenario that reconciles both the multiple signaling pathways linked to CCM proteins and the distinct therapeutic approaches proposed so far. In addition, recent studies in CCM patient cohorts suggest that genetic susceptibility factors related to differences in vascular sensitivity to oxidative stress and inflammation contribute to inter-individual differences in CCM disease susceptibility and severity. This review discusses recent progress into the understanding of the molecular basis and mechanisms of CCM disease pathogenesis, with specific emphasis on the potential contribution of altered cell responses to oxidative stress and inflammatory events occurring locally in the microvascular environment, and consequent implications for the development of novel, safe, and effective preventive and therapeutic strategies., (Copyright © 2016 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
44. Cytochrome P450 and matrix metalloproteinase genetic modifiers of disease severity in Cerebral Cavernous Malformation type 1.
- Author
-
Choquet H, Trapani E, Goitre L, Trabalzini L, Akers A, Fontanella M, Hart BL, Morrison LA, Pawlikowska L, Kim H, and Retta SF
- Subjects
- Adult, Aged, Brain diagnostic imaging, Brain pathology, Female, Genotype, Hemangioma, Cavernous, Central Nervous System diagnostic imaging, Hemangioma, Cavernous, Central Nervous System pathology, Heterozygote, Humans, KRIT1 Protein, Magnetic Resonance Imaging, Male, Microtubule-Associated Proteins genetics, Middle Aged, Multigene Family genetics, Mutation, Phenotype, Polymorphism, Single Nucleotide, Proto-Oncogene Proteins genetics, Severity of Illness Index, Brain metabolism, Cytochrome P-450 Enzyme System genetics, Hemangioma, Cavernous, Central Nervous System genetics, Matrix Metalloproteinases genetics, Oxidative Stress genetics
- Abstract
Background: Familial Cerebral Cavernous Malformation type 1 (CCM1) is an autosomal dominant disease caused by mutations in the Krev Interaction Trapped 1 (KRIT1/CCM1) gene, and characterized by multiple brain lesions. CCM lesions manifest across a range of different phenotypes, including wide differences in lesion number, size and susceptibility to intracerebral hemorrhage (ICH). Oxidative stress plays an important role in cerebrovascular disease pathogenesis, raising the possibility that inter-individual variability in genes related to oxidative stress may contribute to the phenotypic differences observed in CCM1 disease. Here, we investigated whether candidate oxidative stress-related cytochrome P450 (CYP) and matrix metalloproteinase (MMP) genetic markers grouped by superfamilies, families or genes, or analyzed individually influence the severity of CCM1 disease., Methods: Clinical assessment and cerebral susceptibility-weighted magnetic resonance imaging (SWI) were performed to determine total and large (≥5mm in diameter) lesion counts as well as ICH in 188 Hispanic CCM1 patients harboring the founder KRIT1/CCM1 'common Hispanic mutation' (CCM1-CHM). Samples were genotyped on the Affymetrix Axiom Genome-Wide LAT1 Human Array. We analyzed 1,122 genetic markers (both single nucleotide polymorphisms (SNPs) and insertion/deletions) grouped by CYP and MMP superfamily, family or gene for association with total or large lesion count and ICH adjusted for age at enrollment and gender. Genetic markers bearing the associations were then analyzed individually., Results: The CYP superfamily showed a trend toward association with total lesion count (P=0.057) and large lesion count (P=0.088) in contrast to the MMP superfamily. The CYP4 and CYP8 families were associated with either large lesion count or total lesion count (P=0.014), and two other families (CYP46 and the MMP Stromelysins) were associated with ICH (P=0.011 and 0.007, respectively). CYP4F12 rs11085971, CYP8A1 rs5628, CYP46A1 rs10151332, and MMP3 rs117153070 single SNPs, mainly bearing the above-mentioned associations, were also individually associated with CCM1 disease severity., Conclusions: Overall, our candidate oxidative stress-related genetic markers set approach outlined CYP and MMP families and identified suggestive SNPs that may impact the severity of CCM1 disease, including the development of numerous and large CCM lesions and ICH. These novel genetic risk factors of prognostic value could serve as early objective predictors of disease outcome and might ultimately provide better options for disease prevention and treatment., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. Platinum nanozymes recover cellular ROS homeostasis in an oxidative stress-mediated disease model.
- Author
-
Moglianetti M, De Luca E, Pedone D, Marotta R, Catelani T, Sartori B, Amenitsch H, Retta SF, and Pompa PP
- Subjects
- Animals, Caco-2 Cells, Endosomes metabolism, HeLa Cells, Hemangioma, Cavernous, Central Nervous System metabolism, Hemangioma, Cavernous, Central Nervous System pathology, Humans, Lysosomes metabolism, MCF-7 Cells, Mice, Oxidative Stress, Antioxidants chemistry, Antioxidants pharmacokinetics, Antioxidants pharmacology, Hemangioma, Cavernous, Central Nervous System drug therapy, Models, Biological, Nanoparticles chemistry, Platinum chemistry, Platinum pharmacokinetics, Platinum pharmacology, Reactive Oxygen Species metabolism
- Abstract
In recent years, the use of nanomaterials as biomimetic enzymes has attracted great interest. In this work, we show the potential of biocompatible platinum nanoparticles (Pt NPs) as antioxidant nanozymes, which combine abundant cellular internalization and efficient scavenging activity of cellular reactive oxygen species (ROS), thus simultaneously integrating the functions of nanocarriers and antioxidant drugs. Careful toxicity assessment and intracellular tracking of Pt NPs proved their cytocompatibility and high cellular uptake, with compartmentalization within the endo/lysosomal vesicles. We have demonstrated that Pt NPs possess strong and broad antioxidant properties, acting as superoxide dismutase, catalase, and peroxidase enzymes, with similar or even superior performance than natural enzymes, along with higher adaptability to the changes in environmental conditions. We then exploited their potent activity as radical scavenging materials in a cellular model of an oxidative stress-related disorder, namely human Cerebral Cavernous Malformation (CCM) disease, which is associated with a significant increase in intracellular ROS levels. Noteworthily, we found that Pt nanozymes can efficiently reduce ROS levels, completely restoring the cellular physiological homeostasis.
- Published
- 2016
- Full Text
- View/download PDF
46. Beyond multiple mechanisms and a unique drug: Defective autophagy as pivotal player in cerebral cavernous malformation pathogenesis and implications for targeted therapies.
- Author
-
Marchi S, Trapani E, Corricelli M, Goitre L, Pinton P, and Retta SF
- Abstract
Cerebral Cavernous Malformation (CCM) is a major cerebrovascular disease of proven genetic origin affecting 0.3-0.5% of the general population. It is characterized by abnormally enlarged and leaky capillaries, which predispose to seizures, focal neurological deficits and intracerebral hemorrhage. Causative loss-of-function mutations have been identified in 3 genes, KRIT1 (CCM1), CCM2 and PDCD10 (CCM3). While providing new options for the development of pharmacological therapies, recent advances in knowledge of the functions of these genes have clearly indicated that they exert pleiotropic effects on several biological pathways. Recently, we found that defective autophagy is a common feature of loss-of-function mutations of the 3 known CCM genes, and underlies major phenotypic signatures of CCM disease, including endothelial-to-mesenchymal transition and enhanced ROS production, suggesting a unifying pathogenetic mechanism and reconciling the distinct therapeutic approaches proposed so far. In this invited review, we discuss autophagy as a possible unifying mechanism in CCM disease pathogenesis, and new perspectives and avenues of research for disease prevention and treatment, including novel potential drug repurposing and combination strategies, and identification of genetic risk factors as basis for development of personalized medicine approaches.
- Published
- 2016
- Full Text
- View/download PDF
47. Cellular processes underlying cerebral cavernous malformations: Autophagy as another point of view.
- Author
-
Marchi S, Retta SF, and Pinton P
- Subjects
- Humans, Models, Biological, Autophagy, Hemangioma, Cavernous, Central Nervous System pathology
- Abstract
A growing amount of evidence indicates that autophagy plays a pivotal role in a plethora of human pathological conditions. We have recently broadened the list of the so-called autophagy-related diseases, describing the involvement of defective autophagy in the pathogenesis of cerebral cavernous malformations.
- Published
- 2016
- Full Text
- View/download PDF
48. Defective autophagy is a key feature of cerebral cavernous malformations.
- Author
-
Marchi S, Corricelli M, Trapani E, Bravi L, Pittaro A, Delle Monache S, Ferroni L, Patergnani S, Missiroli S, Goitre L, Trabalzini L, Rimessi A, Giorgi C, Zavan B, Cassoni P, Dejana E, Retta SF, and Pinton P
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Endothelial Cells physiology, Gene Knockdown Techniques, Humans, KRIT1 Protein, Mice, Mice, Knockout, Microtubule-Associated Proteins genetics, Proto-Oncogene Proteins genetics, Autophagy, Hemangioma, Cavernous, Central Nervous System genetics, Hemangioma, Cavernous, Central Nervous System pathology, Microtubule-Associated Proteins metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Cerebral cavernous malformation (CCM) is a major cerebrovascular disease affecting approximately 0.3-0.5% of the population and is characterized by enlarged and leaky capillaries that predispose to seizures, focal neurological deficits, and fatal intracerebral hemorrhages. Cerebral cavernous malformation is a genetic disease that may arise sporadically or be inherited as an autosomal dominant condition with incomplete penetrance and variable expressivity. Causative loss-of-function mutations have been identified in three genes, KRIT1 (CCM1), CCM2 (MGC4607), and PDCD10 (CCM3), which occur in both sporadic and familial forms. Autophagy is a bulk degradation process that maintains intracellular homeostasis and that plays essential quality control functions within the cell. Indeed, several studies have identified the association between dysregulated autophagy and different human diseases. Here, we show that the ablation of the KRIT1 gene strongly suppresses autophagy, leading to the aberrant accumulation of the autophagy adaptor p62/SQSTM1, defective quality control systems, and increased intracellular stress. KRIT1 loss-of-function activates the mTOR-ULK1 pathway, which is a master regulator of autophagy, and treatment with mTOR inhibitors rescues some of the mole-cular and cellular phenotypes associated with CCM. Insufficient autophagy is also evident in CCM2-silenced human endothelial cells and in both cells and tissues from an endothelial-specific CCM3-knockout mouse model, as well as in human CCM lesions. Furthermore, defective autophagy is highly correlated to endothelial-to-mesenchymal transition, a crucial event that contributes to CCM progression. Taken together, our data point to a key role for defective autophagy in CCM disease pathogenesis, thus providing a novel framework for the development of new pharmacological strategies to prevent or reverse adverse clinical outcomes of CCM lesions., (© 2015 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2015
- Full Text
- View/download PDF
49. Professional athletes and cerebral cavernomas: an obstacle to overcome.
- Author
-
Fontanella MM, Panciani PP, Spena G, Roca E, Migliorati K, Ambrosi C, Sturiale CL, and Retta SF
- Subjects
- Adult, Central Nervous System Neoplasms surgery, Hemangioma, Cavernous, Central Nervous System surgery, Humans, Magnetic Resonance Imaging, Male, Seizures etiology, Athletes, Central Nervous System Neoplasms diagnostic imaging, Hemangioma, Cavernous, Central Nervous System diagnostic imaging
- Published
- 2015
50. Cerebral cavernous malformation (CCM) disease: from monogenic forms to genetic susceptibility factors.
- Author
-
Trapani E and Retta SF
- Subjects
- Humans, Central Nervous System Neoplasms genetics, Genetic Predisposition to Disease genetics, Hemangioma, Cavernous, Central Nervous System genetics
- Abstract
Cerebral cavernous malformation (CCM) is a vascular disease of proven genetic origin, which may arise sporadically or can be inherited as autosomal dominant condition with incomplete penetrance and highly variable expressivity. CCM lesions manifest across a range of different phenotypes, including wide interindividual differences in lesion number, size and susceptibility to intracerebral hemorrhage (ICH), and may remain asymptomatic during the host's lifetime or result in pathological conditions of various type and severity at any age, with symptoms ranging from relatively minor (but still disabling) headaches through to very severe neurological deficits, seizures, and stroke. Currently, surgical removal of accessible lesions is the only direct therapeutic approach for CCM disease. However, whereas little information is available on the natural history of risk for patients to develop serious complications, such as ICH, prognostic biomarkers remain to be identified in order to ensure timely and optimal clinical decision making. In recent years, it has become clear that the three known CCM genes play an important role in controlling signalling pathways involved in cell responses to oxidative stress, pointing to a novel pathogenic mechanism whereby the function of CCM genes may be relevant in preventing vascular dysfunctions triggered by oxidative stress events. In turn, these novel findings have raised the possibility that genetic susceptibility factors related to differences in sensitivity to oxidative stress, including genetic polymorphisms, may contribute to interindividual differences in CCM disease susceptibility and severity. This review discusses recent progress toward the understanding of molecular mechanisms of pathogenesis and the identification of genetic susceptibility factors that could influence onset, progression and clinical severity of CCM disease, as well as consequent implications for the development of novel, safe and effective therapeutic strategies.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.