Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Patrizia Annunziata is active.

Publication


Featured researches published by Patrizia Annunziata.


Molecular Therapy | 2011

Long-term amelioration of feline Mucopolysaccharidosis VI after AAV-mediated liver gene transfer.

Gabriella Cotugno; Patrizia Annunziata; Alessandra Tessitore; Thomas O'Malley; Anita Capalbo; Armida Faella; Rosa Bartolomeo; Patricia O'Donnell; Ping Wang; Fabio Russo; Meg M. Sleeper; Van W. Knox; Steven Fernandez; Leah Levanduski; John J. Hopwood; Elvira De Leonibus; Mark E. Haskins; Alberto Auricchio

Mucopolysaccharidosis VI (MPS VI) is caused by deficient arylsulfatase B (ARSB) activity resulting in lysosomal storage of glycosaminoglycans (GAGs). MPS VI is characterized by dysostosis multiplex, organomegaly, corneal clouding, and heart valve thickening. Gene transfer to a factory organ like liver may provide a lifetime source of secreted ARSB. We show that intravascular administration of adeno-associated viral vectors (AAV) 2/8-TBG-felineARSB in MPS VI cats resulted in ARSB expression up to 1 year, the last time point of the study. In newborn cats, normal circulating ARSB activity was achieved following delivery of high vector doses (6 × 10(13) genome copies (gc)/kg) whereas delivery of AAV2/8 vector doses as low as 2 × 10(12) gc/kg resulted in higher than normal serum ARSB levels in juvenile MPS VI cats. In MPS VI cats showing high serum ARSB levels, independent of the age at treatment, we observed: (i) clearance of GAG storage, (ii) improvement of long bone length, (iii) reduction of heart valve thickness, and (iv) improvement in spontaneous mobility. Thus, AAV2/ 8-mediated liver gene transfer represents a promising therapeutic strategy for MPS VI patients.


Human Gene Therapy | 2010

Different serum enzyme levels are required to rescue the various systemic features of the mucopolysaccharidoses.

Gabriella Cotugno; Alessandra Tessitore; Anita Capalbo; Patrizia Annunziata; Caterina Strisciuglio; Armida Faella; Michela Aurilio; Maurizio Di Tommaso; Fabio Russo; Antonio Mancini; Elvira De Leonibus; Luigi Aloj; Alberto Auricchio

Mucopolysaccharidoses (MPSs) are lysosomal storage disorders characterized by progressive accumulation of glycosaminoglycans (GAGs) in various tissues. Enzyme replacement therapy (ERT) for several MPSs is available to date. However, the efficacy of ERT is limited, in particular in compartments such as bone, cartilage, the brain, and the eyes. We selected a rodent model of an MPS, with no central nervous system storage, to study the impact, on systemic features of the disease, of various stable levels of exogenous enzymes produced by adeno-associated viral vector (AAV)-mediated liver gene transfer. Low levels (6% of normal) of circulating enzyme were enough to reduce storage and inflammation in the visceral organs and to ameliorate skull abnormalities; intermediate levels (11% of normal) were required to reduce urinary GAG excretion; and high levels (>or=50% of normal) rescued abnormalities of the long bones and motor activity. These data will be instrumental to design appropriate clinical protocols based on either enzyme or gene replacement therapy for MPS and to predict their impact on the pathological features of MPS.


Journal of Biological Chemistry | 2015

Enhancing Autophagy with Drugs or Lung-directed Gene Therapy Reverses the Pathological Effects of Respiratory Epithelial Cell Proteinopathy.

Tunda Hidvegi; Donna B. Stolz; John F. Alcorn; Samuel A. Yousem; Jieru Wang; Adriana S. Leme; A. McGarry Houghton; Pamela Hale; Michael Ewing; Houming Cai; Evelyn Akpadock Garchar; Nunzia Pastore; Patrizia Annunziata; Naftali Kaminski; Joseph M. Pilewski; Steven D. Shapiro; Stephen C. Pak; Gary A. Silverman; Nicola Brunetti-Pierri; David H. Perlmutter

Background: Several rare lung proteinopathies are characterized by lung fibrosis due to accumulation of misfolded proteins in epithelial cells. Results: The PiZ mouse models the pathological characteristics of these lung proteinopathies, and the pathology is ameliorated by autophagy enhancing therapies. Conclusion: Autophagy represents a key proteostasis mechanism for lung proteinopathies and a potential therapeutic target. Significance: The PiZ mouse is an attractive animal model of lung proteinopathies. Recent studies have shown that autophagy mitigates the pathological effects of proteinopathies in the liver, heart, and skeletal muscle but this has not been investigated for proteinopathies that affect the lung. This may be due at least in part to the lack of an animal model robust enough for spontaneous pathological effects from proteinopathies even though several rare proteinopathies, surfactant protein A and C deficiencies, cause severe pulmonary fibrosis. In this report we show that the PiZ mouse, transgenic for the common misfolded variant α1-antitrypsin Z, is a model of respiratory epithelial cell proteinopathy with spontaneous pulmonary fibrosis. Intracellular accumulation of misfolded α1-antitrypsin Z in respiratory epithelial cells of the PiZ model resulted in activation of autophagy, leukocyte infiltration, and spontaneous pulmonary fibrosis severe enough to elicit functional restrictive deficits. Treatment with autophagy enhancer drugs or lung-directed gene transfer of TFEB, a master transcriptional activator of the autophagolysosomal system, reversed these proteotoxic consequences. We conclude that this mouse is an excellent model of respiratory epithelial proteinopathy with spontaneous pulmonary fibrosis and that autophagy is an important endogenous proteostasis mechanism and an attractive target for therapy.


PLOS ONE | 2012

Impact of age at administration, lysosomal storage, and transgene regulatory elements on AAV2/8-mediated rat liver transduction

Gabriella Cotugno; Patrizia Annunziata; Maria Vittoria Barone; Marianthi Karali; Sandro Banfi; Alberto Auricchio

Liver-directed gene transfer is being investigated for the treatment of systemic or liver-specific diseases. Recombinant vectors based on adeno-associated virus serotype 8 (AAV2/8) efficiently transduce liver cells allowing long term transgene expression after a single administration in animal models and in patients. We evaluated the impact on AAV2/8-mediated rat liver transduction of the following variables: i) age at vector administration, ii) presence of lysosomal storage in liver cells, and iii) regulatory elements included in the transgene expression cassette. We found that systemic administration of AAV2/8 to newborn rats results in vector genome dilution and reduced transduction efficacy when compared to adult injected animals, presumably due to hepatocyte proliferation. Accumulation of glycosaminoglycans in lysosomes does not impact on levels and distribution of AAV2/8-mediated liver transduction. Transgene expression occurs in hepatocytes but not in Kupffer or liver endothelial cells when the liver-specific thyroxine-binding-globulin promoter is used. However, extra-hepatic transduction is observed in the spleen and kidney of animals injected at birth. The use of target sequences for the hematopoietic-specific microRNA miR142-3p does not improve liver transduction efficacy neither reduce immune responses to the lysosomal enzyme arylsulfatase B. The inclusion of a variant of the Woodchuck hepatitis virus post-transcriptional regulatory element (WPRE-m) decreases AAV2/8-mediated liver transduction levels. As AAV2/8-mediated liver gene transfer is entering in the clinical arena, these data will provide relevant information to the design of efficient AAV2/8-based therapeutic strategies.


Human Gene Therapy | 2011

Noninvasive Repetitive Imaging of Somatostatin Receptor 2 Gene Transfer with Positron Emission Tomography

Gabriella Cotugno; Michela Aurilio; Patrizia Annunziata; Anita Capalbo; Armida Faella; Valentina Rinaldi; Caterina Strisciuglio; Maurizio Di Tommaso; Luigi Aloj; Alberto Auricchio

Noninvasive in vivo imaging of gene expression is desirable to monitor gene transfer in both animal models and humans. Reporter transgenes with low endogenous expression levels are instrumental to this end. The human somatostatin receptor 2 (hSSTR2) has low expression levels in a variety of tissues, including muscle and liver. We tested the possibility of noninvasively and quantitatively monitoring hSSTR2 transgene expression, following adeno-associated viral (AAV) vector-mediated gene delivery to murine muscle and liver by positron emission tomography (PET) using (68)gallium-DOTA-Tyr(3)-Thr(8)-octreotate ((68)Ga-DOTATATE) as a highly specific SSTR2 ligand. Repetitive PET imaging showed hSSTR2 signal up to 6 months, which corresponds to the last time point of the analysis, after gene delivery in both transduced tissues. The levels of tracer accumulation measured in muscle and liver after gene delivery were significantly higher than in control tissues and correlated with the doses of AAV vector administered. As repetitive, quantitative, noninvasive imaging of AAV-mediated SSTR2 gene transfer to muscle and liver is feasible and efficient using PET, we propose this system to monitor the expression of therapeutic genes coexpressed with SSTR2.


Gene Therapy | 2014

SR-A and SREC-I binding peptides increase HDAd-mediated liver transduction.

Pasquale Piccolo; Patrizia Annunziata; Pratibha Mithbaokar; Nicola Brunetti-Pierri

Helper-dependent adenoviral (HDAd) vectors can mediate long-term, high-level transgene expression from transduced hepatocytes without inducing chronic toxicity. However, vector therapeutic index is narrow because of a toxic acute response with potentially lethal consequences elicited by high vector doses. Kupffer cells (KCs) and liver sinusoidal endothelial cells (LSECs) are major barriers to efficient hepatocyte transduction. We investigated two small peptides (PP1 and PP2) developed by phage display to block scavenger receptor type A (SR-A) and scavenger receptor expressed on endothelial cells type I (SREC-I), respectively, for enhancement of HDAd-mediated hepatocyte transduction efficiency. Pre-incubation of J774A.1 macrophages with either PP1 or PP2 prior to HDAd infection significantly reduced viral vector uptake. In vivo, fluorochrome-conjugated PP1 and PP2 injected intravenously into mice co-localized with both CD68 and CD31 on KCs and LSECs, respectively. Compared with saline pre-treated animals, intravenous injections of both peptides prior to the injection of an HDAd resulted in up to 3.7- and 2.9-fold increase of hepatic transgene expression with PP1 and PP2, respectively. In addition to greater hepatocyte transduction, compared with control saline injected mice, pre-treatment with either peptide resulted in no increased levels of serum interleukin-6, the major marker of adenoviral vector acute toxicity. In summary, we developed small peptides that significantly increase hepatocyte transduction efficacy and improve HDAd therapeutic index with potential for clinical applications.


Gene Therapy | 2016

Helper-dependent adenoviral vectors for liver-directed gene therapy of primary hyperoxaluria type 1

R Castello; R Borzone; S D'Aria; Patrizia Annunziata; Pasquale Piccolo; Nicola Brunetti-Pierri

Primary hyperoxaluria type 1 (PH1) is an inborn error of liver metabolism due to deficiency of the peroxisomal enzyme alanine:glyoxylate aminotransferase (AGT), which catalyzes conversion of glyoxylate into glycine. AGT deficiency results in overproduction of oxalate that ultimately leads to end-stage renal disease and death. Organ transplantation as either preemptive liver transplantation or combined liver/kidney transplantation is the only available therapy to prevent disease progression. Gene therapy is an attractive option to provide an alternative treatment for PH1. Toward this goal, we investigated helper-dependent adenoviral (HDAd) vectors for liver-directed gene therapy of PH1. Compared with saline controls, AGT-deficient mice injected with an HDAd encoding the AGT under the control of a liver-specific promoter showed a significant reduction of hyperoxaluria and less increase of urinary oxalate following challenge with ethylene glycol, a precursor of glyoxylate. These studies may thus pave the way to clinical application of HDAd for PH1 gene therapy.


Cell Reports | 2016

In Silico Modeling of Liver Metabolism in a Human Disease Reveals a Key Enzyme for Histidine and Histamine Homeostasis.

Roberto Pagliarini; Raffaele Castello; Francesco Napolitano; Roberta Borzone; Patrizia Annunziata; Giorgia Mandrile; Mario Marchi; Nicola Brunetti-Pierri; Diego di Bernardo

Summary Primary hyperoxaluria type I (PH1) is an autosomal-recessive inborn error of liver metabolism caused by alanine:glyoxylate aminotransferase (AGT) deficiency. In silico modeling of liver metabolism in PH1 recapitulated accumulation of known biomarkers as well as alteration of histidine and histamine levels, which we confirmed in vitro, in vivo, and in PH1 patients. AGT-deficient mice showed decreased vascular permeability, a readout of in vivo histamine activity. Histamine reduction is most likely caused by increased catabolism of the histamine precursor histidine, triggered by rerouting of alanine flux from AGT to the glutamic-pyruvate transaminase (GPT, also known as the alanine-transaminase ALT). Alanine administration reduces histamine levels in wild-type mice, while overexpression of GPT in PH1 mice increases plasma histidine, normalizes histamine levels, restores vascular permeability, and decreases urinary oxalate levels. Our work demonstrates that genome-scale metabolic models are clinically relevant and can link genotype to phenotype in metabolic disorders.


Hepatology | 2017

Down‐regulation of hepatocyte nuclear factor‐4α and defective zonation in livers expressing mutant Z α1‐antitrypsin

Pasquale Piccolo; Patrizia Annunziata; Leandro R. Soria; Sergio Attanasio; Anna Barbato; Raffaele Castello; Annamaria Carissimo; Luca Quagliata; Luigi Terracciano; Nicola Brunetti-Pierri

α1‐Antitrypsin (AAT) deficiency is one of the most common genetic disorders and the liver disease due to the Z mutant of AAT (ATZ) is a prototype of conformational disorder due to protein misfolding with consequent aberrant intermolecular protein aggregation. In the present study, we found that livers of PiZ transgenic mice expressing human ATZ have altered expression of a network of hepatocyte transcriptional factors, including hepatocyte nuclear factor‐4α, that is early down‐regulated and induces a transcriptional repression of ATZ expression. Reduced hepatocyte nuclear factor‐4α was associated with activation of β‐catenin, which regulates liver zonation. Livers of PiZ mice and human patients with AAT deficiency were both found to have a severe perturbation of liver zonation. Functionally, PiZ mice showed a severe defect of ureagenesis, as shown by increased baseline ammonia, and reduced urea production and survival after an ammonia challenge. Down‐regulation of hepatocyte nuclear factor‐4α expression and defective zonation in livers have not been recognized so far as features of the liver disease caused by ATZ and are likely involved in metabolic disturbances and in the increased risk of hepatocellular carcinoma in patients with AAT deficiency. Conclusion: The findings of this study are consistent with the concept that abnormal AAT protein conformation and intrahepatic accumulation have broad effects on metabolic liver functions. (Hepatology 2017;66:124–135).


American Journal of Medical Genetics Part A | 2010

Absence of microcephalin gene mutations in a large cohort of non-consanguineous patients with autosomal recessive primary microcephaly.

Iris Scala; Luigi Titomanlio; Ennio Del Giudice; Sandrine Passemard; Chiara Figliuolo; Patrizia Annunziata; Barbara Granese; Elena Scarpato; Alain Verloes; Generoso Andria

Absence of Microcephalin Gene Mutations in a Large Cohort of Non-Consanguineous Patients With Autosomal Recessive Primary Microcephaly Iris Scala, Luigi Titomanlio, Ennio Del Giudice, Sandrine Passemard, Chiara Figliuolo, Patrizia Annunziata, Barbara Granese, Elena Scarpato, Alain Verloes, and Generoso Andria* Department of Pediatrics, University Federico II, Naples, Italy Department of Clinical Genetics, Robert Debr e Hospital, AP-HP, Paris 7 University, Paris, France Service de Neurop ediatrie, Robert Debr e Hospital, AP-HP, Paris 7 University, Paris, France Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy

Collaboration


Dive into the Patrizia Annunziata's collaboration.

Top Co-Authors

Avatar

Nicola Brunetti-Pierri

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Alberto Auricchio

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Gabriella Cotugno

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Pasquale Piccolo

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Anita Capalbo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Generoso Andria

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Luigi Aloj

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Leandro R. Soria

National Scientific and Technical Research Council

View shared research outputs
Top Co-Authors

Avatar

Annamaria Carissimo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Caterina Strisciuglio

Seconda Università degli Studi di Napoli

View shared research outputs
Researchain Logo
Decentralizing Knowledge