Network


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

Hotspot


Dive into the research topics where Julien Baruteau is active.

Publication


Featured researches published by Julien Baruteau.


Journal of Inherited Metabolic Disease | 2014

Clinical and biological features at diagnosis in mitochondrial fatty acid beta-oxidation defects: a French pediatric study from 187 patients. Complementary data

Julien Baruteau; Philippe Sachs; Pierre Broué; Michèle Brivet; Hendy Abdoul; Christine Vianey-Saban; Hélène Ogier de Baulny

Dear Editor, We recently described a cohort of patients affected by fatty acid oxidation defects (FAOD) in the Journal of Inherited Metabolic Diseases (Baruteau et al 2012). To our knowledge this is the largest published group of fatty acid oxidation patients. In the weeks following the publication, we received several requests from the readers asking for the data to be analysed per disease set. In response to these requests we analysed the data accordingly (Table 1) also with three additional patients (making the overall n=190). The following important points in the individual disease presentations are highlighted:


Frontiers in Molecular Neuroscience | 2014

Perinatal systemic gene delivery using adeno-associated viral vectors

Rajvinder Karda; Suzanne M. K. Buckley; Citra Nurfarah Zaini Mattar; Joanne Ng; Giulia Massaro; M. Hughes; Manju A. Kurian; Julien Baruteau; Paul Gissen; Jerry Chan; Chiara Bacchelli; Simon N. Waddington; Ahad A. Rahim

Neurodegenerative monogenic diseases often affect tissues and organs beyond the nervous system. An effective treatment would require a systemic approach. The intravenous administration of novel therapies is ideal but is hampered by the inability of such drugs to cross the blood–brain barrier (BBB) and precludes efficacy in the central nervous system. A number of these early lethal intractable diseases also present devastating irreversible pathology at birth or soon after. Therefore, any therapy would ideally be administered during the perinatal period to prevent, stop, or ameliorate disease progression. The concept of perinatal gene therapy has moved a step further toward being a feasible approach to treating such disorders. This has primarily been driven by the recent discoveries that particular serotypes of adeno-associated virus (AAV) gene delivery vectors have the ability to cross the BBB following intravenous administration. Furthermore, safety has been demonstrated after perinatal administration mice and non-human primates. This review focuses on the progress made in using AAV to achieve systemic transduction and what this means for developing perinatal gene therapy for early lethal neurodegenerative diseases.


Journal of Inherited Metabolic Disease | 2017

Gene therapy for monogenic liver diseases: clinical successes, current challenges and future prospects

Julien Baruteau; Simon N. Waddington; Ian E. Alexander; Paul Gissen

Over the last decade, pioneering liver-directed gene therapy trials for haemophilia B have achieved sustained clinical improvement after a single systemic injection of adeno-associated virus (AAV) derived vectors encoding the human factor IX cDNA. These trials demonstrate the potential of AAV technology to provide long-lasting clinical benefit in the treatment of monogenic liver disorders. Indeed, with more than ten ongoing or planned clinical trials for haemophilia A and B and dozens of trials planned for other inherited genetic/metabolic liver diseases, clinical translation is expanding rapidly. Gene therapy is likely to become an option for routine care of a subset of severe inherited genetic/metabolic liver diseases in the relatively near term. In this review, we aim to summarise the milestones in the development of gene therapy, present the different vector tools and their clinical applications for liver-directed gene therapy. AAV-derived vectors are emerging as the leading candidates for clinical translation of gene delivery to the liver. Therefore, we focus on clinical applications of AAV vectors in providing the most recent update on clinical outcomes of completed and ongoing gene therapy trials and comment on the current challenges that the field is facing for large-scale clinical translation. There is clearly an urgent need for more efficient therapies in many severe monogenic liver disorders, which will require careful risk-benefit analysis for each indication, especially in paediatrics.


Gene Therapy | 2017

Delivering efficient liver-directed AAV-mediated gene therapy

Julien Baruteau; Simon N. Waddington; Ian E. Alexander; Paul Gissen

Adeno-associated virus vectors (AAV) have become the leading technology for liver-directed gene therapy. After the pioneering trials using AAV2 and AAV8 to treat haemophilia B, D’Avola et al. recently reported the first-in-human clinical trial of adeno-associated virus vector serotype 5 (AAV5) in acute intermittent porphyria (AIP). Treatment was reported as safe, but the main surrogate biomarkers of AIP, porphobilinogen (PBG) and delta-aminolevulinate (ALA) were unchanged. This lack of efficacy contrasts with results from the haemophilia B trial using AAV8 capsid by Nathwani et al., which showed a significant and long-lasting improvement of the clinical phenotype. Haemophilia B is an amenable target for successful gene therapy as raising expression of plasma factor IX (FIX) level above 1% can modify the phenotype from severe to moderate. Development of a variety of capsids for clinical application is useful to overcome pre-existing neutralising antibodies. The differences in cell-specific transduction by different AAV serotypes are primarily owing to specificities in cellular uptake or post cell-entry processing. Indeed AAV5 presents several theoretical advantages as an alternative capsid to AAV8 for liver-directed gene therapy: suitable liver tropism, less off-target biodistribution, low seroprevalence in humans and minimal cross-reactivity with other serotypes.


bioRxiv | 2018

Gene therapy in argininosuccinic aciduria

Julien Baruteau; Dany Perocheau; Joanna Hanley; Eridan Rocha Ferreira; Rajvinder Karda; Jo Ng; Natalie Suff; Ahad A. Rahim; M. Hughes; Blerida Banushi; Helen Prunty; Mariya Hristova; Deborah Ridout; Alex Virasami; Simon Heales; Steven J. Howe; Suzanne M. K. Buckley; Philippa B. Mills; Paul Gissen; Simon N. Waddington

Argininosuccinate lyase (ASL) belongs to the liver-based urea cycle detoxifying ammonia, and the citrulline-nitric oxide cycle synthesising nitric oxide (NO). ASL-deficient patients present argininosuccinic aciduria characterised by hyperammonaemia and a multi-organ disease with neurocognitive impairment. Current therapeutic guidelines aim to control ammonaemia without considering the systemic NO imbalance. Here, we observed a neuronal disease with oxidative/nitrosative stress in ASL-deficient mouse brains. A single systemic injection of gene therapy mediated by an adeno-associated viral vector serotype 8 (AAV8) in adult or neonatal mice demonstrated the long-term correction of the urea cycle and the citrulline-NO cycle in the brain, respectively. The neuronal disease persisted if ammonaemia only was normalised but was dramatically reduced after correction of both ammonaemia and neuronal ASL activity. This was correlated with behavioural improvement and a decrease of the cortical cell death rate. Thus, the cerebral disease in argininosuccinic aciduria involves neuronal oxidative/nitrosative stress not mediated by hyperammonaemia, which is reversed by AAV gene transfer targeting the brain and the liver, acting on two different metabolic pathways via a single vector delivered systemically. This approach provides new hope for hepatocerebral metabolic diseases.Argininosuccinate lyase (ASL) belongs to the liver-based urea cycle detoxifying ammonia, and the citrulline-nitric oxide cycle synthesising nitric oxide (NO). ASL-deficient patients present argininosuccinic aciduria characterised by hyperammonaemia and a multi-organ disease with neurocognitive impairment. Current therapeutic guidelines aim to control ammonaemia without considering the systemic NO imbalance. Here, we observed a neuronal disease with oxidative/nitrosative stress in ASL-deficient mouse brains. A single systemic injection of gene therapy mediated by an adeno-associated viral vector serotype 8 (AAV8) in adult or neonatal mice demonstrated the long-term correction of the urea cycle and the citrulline-NO cycle in the brain, respectively. The neuronal disease persisted if ammonaemia only was normalised but was dramatically reduced after correction of both ammonaemia and neuronal ASL activity. This was correlated with behavioural improvement and a decrease of the cortical cell death rate. Thus, the cerebral disease in argininosuccinic aciduria involves neuronal oxidative/nitrosative stress not mediated by hyperammonaemia, which is reversed by AAV gene transfer targeting the brain and the liver, acting on two different metabolic pathways via a single vector delivered systemically. This approach provides new hope for hepatocerebral metabolic diseases.


Nature Communications | 2018

Argininosuccinic aciduria fosters neuronal nitrosative stress reversed by Asl gene transfer

Julien Baruteau; Dany Perocheau; Joanna Hanley; Maëlle Lorvellec; Eridan Rocha-Ferreira; Rajvinder Karda; Joanne Ng; Natalie Suff; Juan Antinao Diaz; Ahad A. Rahim; M. Hughes; Blerida Banushi; Helen Prunty; Mariya Hristova; Deborah Ridout; Alex Virasami; Simon Heales; Stewen J. Howe; Suzanne M. K. Buckley; Philippa B. Mills; Paul Gissen; Simon N. Waddington

Argininosuccinate lyase (ASL) belongs to the hepatic urea cycle detoxifying ammonia, and the citrulline-nitric oxide (NO) cycle producing NO. ASL-deficient patients present argininosuccinic aciduria characterised by hyperammonaemia, multiorgan disease and neurocognitive impairment despite treatment aiming to normalise ammonaemia without considering NO imbalance. Here we show that cerebral disease in argininosuccinic aciduria involves neuronal oxidative/nitrosative stress independent of hyperammonaemia. Intravenous injection of AAV8 vector into adult or neonatal ASL-deficient mice demonstrates long-term correction of the hepatic urea cycle and the cerebral citrulline-NO cycle, respectively. Cerebral disease persists if ammonaemia only is normalised but is dramatically reduced after correction of both ammonaemia and neuronal ASL activity. This correlates with behavioural improvement and reduced cortical cell death. Thus, neuronal oxidative/nitrosative stress is a distinct pathophysiological mechanism from hyperammonaemia. Disease amelioration by simultaneous brain and liver gene transfer with one vector, to treat both metabolic pathways, provides new hope for hepatocerebral metabolic diseases.Patients with mutations in the ASL gene present with argininosuccinic aciduria characterised by hyperammonaemia and cognitive impairment. Here, the authors show that cerebral disease involves neuronal nitrosative/oxidative stress that is not induced by hyperammonaemia, and that it can be reversed using AAV-ASL directed to liver and brain in mice.


Molecular therapy. Nucleic acids | 2018

Foamy Virus Vectors Transduce Visceral Organs and Hippocampal Structures following In Vivo Delivery to Neonatal Mice

John R. Counsell; Rajvinder Karda; Juan Antinao Diaz; Louise Carey; Tatiana Wiktorowicz; Suzanne M. K. Buckley; Shima Ameri; Joanne Ng; Julien Baruteau; Filipa Almeida; Rohan de Silva; Roberto Simone; Eleonora Lugarà; Gabriele Lignani; Dirk Lindemann; Axel Rethwilm; Ahad A. Rahim; Simon N. Waddington; Steven J. Howe

Viral vectors are rapidly being developed for a range of applications in research and gene therapy. Prototype foamy virus (PFV) vectors have been described for gene therapy, although their use has mainly been restricted to ex vivo stem cell modification. Here we report direct in vivo transgene delivery with PFV vectors carrying reporter gene constructs. In our investigations, systemic PFV vector delivery to neonatal mice gave transgene expression in the heart, xiphisternum, liver, pancreas, and gut, whereas intracranial administration produced brain expression until animals were euthanized 49 days post-transduction. Immunostaining and confocal microscopy analysis of injected brains showed that transgene expression was highly localized to hippocampal architecture despite vector delivery being administered to the lateral ventricle. This was compared with intracranial biodistribution of lentiviral vectors and adeno-associated virus vectors, which gave a broad, non-specific spread through the neonatal mouse brain without regional localization, even when administered at lower copy numbers. Our work demonstrates that PFV can be used for neonatal gene delivery with an intracranial expression profile that localizes to hippocampal neurons, potentially because of the mitotic status of the targeted cells, which could be of use for research applications and gene therapy of neurological disorders.


Human Gene Therapy | 2018

Age-related seroprevalence of antibodies against AAV-LK03 in a UK population cohort

Dany Perocheau; Sharon Cunningham; Miss Juhee Lee; Juan Antinao Diaz; Simon N. Waddington; Kimberly Gilmour; Simon Eaglestone; Leszek Lisowski; Adrian J. Thrasher; Ian E. Alexander; Paul Gissen; Julien Baruteau

Recombinant adeno-associated virus (rAAV) vectors are a promising platform for in vivo gene therapy. The presence of neutralizing antibodies (Nab) against AAV capsids decreases cell transduction efficiency and is a common exclusion criterion for participation in clinical trials. Novel engineered capsids are being generated to improve gene delivery to the target cells and facilitate success of clinical trials; however, the prevalence of antibodies against such capsids remains largely unknown. We therefore assessed the seroprevalence of antibodies against a novel synthetic liver-tropic capsid AAV-LK03. We measured seroprevalence of immunoglobulin (Ig)G (i.e., neutralizing and nonneutralizing) antibodies and Nab to AAV-LK03 in a cohort of 323 UK patients (including 260 pediatric) and 52 juvenile rhesus macaques. We also performed comparative analysis of seroprevalence of Nab against wild-type AAV8 and AAV3B capsids. Overall IgG seroprevalence for AAV-LK03 was 39% in human samples. The titer increased with age. Prevalence of Nab was 23%, 35%, and 18% for AAV-LK03, AAV3B, and AAV8, respectively, with the lowest seroprevalence between 3 and 17 years of age for all serotypes. Presence of Nab against AAV-LK03 decreased from 36% in the youngest cohort (birth to 6 months) to 7% in older primary school-age children (9–11 years) and then progressively increased to 54% in late adulthood. Cross-reactivity between serotypes was >60%. Nab seroprevalence in macaques was 62%, 85%, and 40% for AAV-LK03, AAV3B, and AAV8, respectively. When planning for AAV gene therapy clinical trials, knowing the seropositivity of the target population is critical. In the population studied, AAV seroprevalence for AAV serotypes tested was low. However, high cross-reactivity between AAV serotypes remains a barrier for re-injection. Shifts in Nab seroprevalence during the first decade need to be confirmed by longitudinal studies. This possibility suggests that pediatric patients could respond differently to AAV therapy according to age. If late childhood is an ideal age window, intervention at an early age when maternal Nab levels are high may be challenging. Nab-positive children excluded from trials could be rescreened for eligibility at regular intervals because this status may change.


American Journal of Pathology | 2018

Ascending Vaginal Infection Using Bioluminescent Bacteria Evokes Intrauterine Inflammation, Preterm Birth, and Neonatal Brain Injury in Pregnant Mice

Natalie Suff; Rajvinder Karda; Juan Antinao Diaz; Joanne Ng; Julien Baruteau; Dany Perocheau; Mark Tangney; Peter W. Taylor; Donald Peebles; Suzanne M. K. Buckley; Simon N. Waddington

Preterm birth is a serious global health problem and the leading cause of infant death before 5 years of age. At least 40% of cases are associated with infection. The most common way for pathogens to access the uterine cavity is by ascending from the vagina. Bioluminescent pathogens have revolutionized the understanding of infectious diseases. We hypothesized that bioluminescent Escherichia coli can be used to track and monitor ascending vaginal infections. Two bioluminescent strains were studied: E. coli K12 MG1655-lux, a nonpathogenic laboratory strain, and E. coli K1 A192PP-lux2, a pathogenic strain capable of causing neonatal meningitis and sepsis in neonatal rats. On embryonic day 16, mice received intravaginal E. coli K12, E. coli K1, or phosphate-buffered saline followed by whole-body bioluminescent imaging. In both cases, intravaginal delivery of E. coli K12 or E. coli K1 led to bacterial ascension into the uterine cavity, but only E. coli K1 induced preterm parturition. Intravaginal administration of E. coli K1 significantly reduced the proportion of pups born alive compared with E. coli K12 and phosphate-buffered saline controls. However, in both groups of viable pups born after bacterial inoculation, there was evidence of comparable brain inflammation by postnatal day 6. This study ascribes specific mechanisms by which exposure to intrauterine bacteria leads to premature delivery and neurologic inflammation in neonates.


Molecular Therapy | 2015

392. Evaluating Promoter Regulated Dopaminergic Neuron Targeting with AAV9 Delivered To the Neonatal Mouse

Joanne Ng; Rajvinder Karda; Giulia Massaro; E. Meyer; S. Barrel; Julien Baruteau; M. Hughes; Dp Perocheau; Smk Buckley; Manju A. Kurian; Ahad A. Rahim; Simon N. Waddington

Here we observe evidence that targeting mid-brain DA neurons is influenced by both delivery method and promoter following administration to the neonatal mouse brain.IV delivery using rTH and hSyn promoters showed the highest DA neuron transduction whilst IC delivery and GUSB promoter was least effective. Interestingly IC hSyn did not transduce TH positive neurons despite demonstrating the most extensive intracranial transduction. Targeting the neonatal mid-brain DA neurons remains a significant challenge and these data suggest that both promoter of choice and delivery method are important factors to consider.

Collaboration


Dive into the Julien Baruteau's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rajvinder Karda

University College London

View shared research outputs
Top Co-Authors

Avatar

Joanne Ng

University College London

View shared research outputs
Top Co-Authors

Avatar

Paul Gissen

University College London

View shared research outputs
Top Co-Authors

Avatar

Ahad A. Rahim

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dany Perocheau

University College London

View shared research outputs
Top Co-Authors

Avatar

M. Hughes

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Natalie Suff

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge