Network


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

Hotspot


Dive into the research topics where Agnes Jani is active.

Publication


Featured researches published by Agnes Jani.


Human Gene Therapy | 2002

Increased Survival and Function of SOD1 Mice After Glial Cell-Derived Neurotrophic Factor Gene Therapy

Gyula Acsadi; Roumen A. Anguelov; Huibin Yang; Gabor Toth; Ronald Thomas; Agnes Jani; Yuying Wang; Emilia Ianakova; Sulaiman Mohammad; Richard A. Lewis; Michael E. Shy

Amyotrophic lateral sclerosis (ALS) is caused by a progressive degeneration of motor neurons. The cause of sporadic ALS is not known, but 1-2% of all cases are familial and caused by mutations in the copper-zinc superoxide dismutase (SOD1) gene. Transgenic SOD1 mice serve as a transgenic mouse model for these cases. Glial cell-derived neurotrophic factor (GDNF) has a potent trophic effect on motor neurons. Clinical trials in which growth factors have been systemically administered to ALS patients have not been effective, owing in part to the short half-life of these factors and their low concentrations at target sites. Gene transfer of therapeutic factors to motor neurons and/or their target cells, such as muscle, may overcome these problems. Previously, we and others have shown that intramuscularly administered adenovirus vector (AVR) results in foreign gene expression not only in muscle cells, but also in relevant motor neurons in the spinal cord, because of retrograde axonal transport. In this study we utilized an AVR to introduce GDNF into muscles of neonatal SOD1 mice. We showed that AVR-mediated GDNF expression delayed the onset of disease by 7 +/- 8 days (mean +/- SD), prolonged survival by 17 +/- 10 days, and delayed the decline in motor functions (as determined on a rotating rod) by 7-14 days. These results demonstrate that gene delivery to muscle and motor neurons has the potential to treat devastating neurodegenerative diseases such as ALS.


Gene Therapy | 1997

Efficient muscle-specific transgene expression after adenovirus-mediated gene transfer in mice using a 1.35 kb muscle creatine kinase promoter/enhancer

Nancy Larochelle; Hanns Lochmüller; Ji-En Zhao; Agnes Jani; Patricia L. Hallauer; Kenneth E. M. Hastings; Bernard Massie; S. Prescott; Basil J. Petrof; George Karpati; Josephine Nalbantoglu

Replication-defective (E1+E3-deleted) human adenovirus vectors are a promising means of therapeutic gene delivery to skeletal muscle cells. Since the tropism of adenovirus is nonselective, muscle-specific expression of systemically administered vectors can only be achieved by the use of a tissue-specific promoter/enhancer that is small enough to fit the insert capacity of the vector. We have generated two replication-defective adenovirus recombinants (AV) in which the reporter gene (either firefly luciferase or E. coli β-galactosidase) was driven by a truncated (1.35 kb) muscle creatine kinase (MCK) promoter/enhancer or by the fast troponin I (TnI) promoter/enhancer. Highly efficient and muscle-specific transgene expression was demonstrated in immunodeficient mice after local injection of AV into muscles at an early age. In nonmuscle tissues (brain, liver, kidney, lung), the transgene expression was extremely low even though in these tissues in situ polymerase chain reaction showed as high an infectivity of the cells by the AV as in muscle. The relatively small size, the good efficiency and the muscle specificity of the MCK promoter would make it ideal to drive the 6.3 kb (truncated) dystrophin cDNA in first generation AV (with a limited (8 kb) insert capacity) designed for gene therapy of Duchenne muscular dystrophy.


Journal of Molecular Medicine | 1995

Adenovirus-mediated gene transfer into striated muscles.

Gyula Acsadi; Agnes Jani; Bernard Massie

Neuromuscular disorders include a variety of inherited and acquired diseases of a variable severity and curability. The inherited forms have a relatively high prevalence, and most lack adequate treatment. Recent advances in DNA diagnostics are likely to decrease the incidence of the familial forms if appropriate genetic counseling is employed [ 17]. However, the high frequency of sporadic cases, including de novo mutations, necessitates the search for cures to these diseases. Therapeutic strategies in various diseases include nonspecific measures to mitigate or eliminate cell dysfunction and prevent cell death, replacement of a missing or malfunctioning protein, introduction of functional nucleic acids (RNA or DNA) into cells to replace a mutated gene, and introduction of novel genetic constructs to alter cellular function. Advances in DNA technology have had a major impact in each of these therapeutic possibilities. Nucleic acid transfer into diseased cells appears by far the most promising therapeutic modality [45, 86, 112]. Diseases with a recessive course of inheritance are the primary targets for this approach (Table 1); hence the techniques for replacing malfunctional genes are more


Experimental Neurology | 1996

Toxicity of replication-defective adenoviral recombinants in dissociated cultures of nervous tissue

Heather D. Durham; Hanns Lochmüller; Agnes Jani; Gyula Acsadi; Bernard Massie; George Karpati

Replication-defective human type 5 adenoviral recombinants (AVR) are very efficient means of introducing foreign genes into neurons in vitro and in vivo; however, a significant reduction in the number of cells expressing reporter genes has been reported to occur over time. In vitro, this may be due to direct toxicity of the protein product of the transgene or adenoviral molecules. In vivo, in addition, an immune attack by the host could eliminate the transduced cells. To assess the direct toxicity of AVR or reporter gene products, a quantitative study of survival of transduced neurons over a period of 4 weeks was conducted in primary neural cultures. Cultures of dissociated murine spinal cord-dorsal root ganglia were exposed to AVR containing the Escherichia coli lacZ (E. coli lacZ) gene under control of either the very efficient cytomegalovirus enhancer/promoter or the fast muscle troponin I promoter, which is not active in these cells. Two factors contributed to loss of neuronal and nonneuronal cells: (i) direct toxicity of (E1 + E3)-deleted replication-incompetent AVR at high titers [> or = 5 x 10(8) viral particles/ml or multiplicity of infection (m.o.i.) 1000] and (ii) high levels of expression of the reporter gene product, beta-galactosidase, at titers that result in 55-75% transduction efficiency (5 x 10(7)-5 x 10(8) viral particles/ml or m.o.i. 100-1000). Despite the efficacy of adenoviral vectors in introducing foreign genes into primary, postmitotic cells, specific precautions must be taken in their use because of the narrow margin between concentrations of recombinants that transduce a sufficient percentage of cells and those that are cytotoxic.


Journal of Molecular Medicine | 1998

Interferons impair early transgene expression by adenovirus-mediated gene transfer in muscle cells

Gyula Acsadi; David O'Hagan; Hanns Lochmüller; Stephen Prescott; Nancy Larochelle; Josephine Nalbantoglu; Agnes Jani; George Karpati

Abstract Recombinant adenovirus (AVR) promises to be an efficient vector in gene therapy for neuromuscular diseases, but in preclinical experiments the expression of therapeutic genes is shorter lived in immunocompetent animals than in immunocompromised hosts. Interferons (IFN), which are known to have a role both in early antiviral activity and in late cytotoxic immunoreaction against the virus or transduced cells, may influence the efficiency of gene transfer. In this study we investigated the role of IFNs in determining the efficiency of gene transfer by AVR. AVRs expressing β-galactosidase (β-gal) from either a cytomegalovirus (CMV) or a troponin-I promoter were used. Muscle cells were infected by AVR after exposure to various IFNs. The αIFN treatment significantly reduced (up to fivefold) the CMV promoter-driven gene expression in muscle cells in vitro and in immature muscles in vivo, while the least effective inhibitor was βIFN. The decrease in gene expression by IFNs was more pronounced with the CMV-driven transgene than troponin-I promoter-driven one and was due to a decrease in transcript level. Intrinsic IFNs that are triggered by AVR administration can decrease the efficiency of gene transfer in muscle cells. Therefore the use of muscle specific promoters in AVR and/or IFN inhibitory agents will likely improve the prospects of effective gene therapy by AVR.


Journal of Virological Methods | 1997

Generation, validation, and large scale production of adenoviral recombinants with large size inserts such as a 6.3 kb human dystrophin cDNA

Agnes Jani; Hanns Lochmüller; Gyula Acsadi; Maude Simoneau; Johnny Huard; Alain Garnier; George Karpati; Bernard Massie

Human, serotype 5 (Ad 5), replication-defective recombinant adenoviruses (AdVs) expressing a 6.3 kb partial dystrophin cDNA (Becker) under the control of either the CMV early or the RSV LTR promoter/enhancer in combination with various polyadenylation sequences (polyA), were developed for gene transfer studies aimed at Duchenne muscular dystrophy. Based on previous experience, a strategy for generation, screening and validation of AdVs with relatively large size gene expression cassette inserts was established. Here we focus on some aspects of stability and safety of such AdVs as gene therapeutic tools based on relevant molecular biological methods. Furthermore, the quality of our best AdV-minidystrophin construct was validated following its large scale production and purification as well as its delivery in mdx mice. These results are of interest for establishing other AdVs, where the combined length of a tissue specific promoter, the gene of interest and the polyA sequences reach the upper limit of the packaging capacity of first generation AdVs.


Journal of Immunological Methods | 2001

Foreign antigenic peptides delivered to the tumor as targets of cytotoxic T cells

Wei Zen Wei; Stuart Ratner; Terry Y. Shibuya; George H. Yoo; Agnes Jani

Cytotoxic T cells (CTL) are readily activated by immunogenic peptides and they exert potent anti-tumor activity if the same peptides are displayed on class I major histocompatibility complex (MHC) of the tumor cells. A handful of tumor-associated antigens have been identified and many of them are weak antigens. As an alternative strategy, strongly antigenic foreign peptides are delivered to the tumor, marking them for CTL recognition. To establish the principle of this new strategy, in vitro and in vivo tumor destruction was tested with BALB/c CTL to L(d)-associated beta-galactosidase (beta-gal) peptide p876. In vitro, anti-p876 CTL destroyed tumor cells in a single-cell suspension or in 3-D tumor boluses when exogenous p876 was added. Exogenous IL-2 was required to sustain CTL activity for complete destruction of tumor boluses. In vivo, BALB/c mice were immunized with p876 and a CD4 activating Pan DR reactive epitope (PADRE). PADRE, which binds to several different MHC class II antigen and activates CD4 T cells, induced delayed-type hypersensitivity and stimulated T cell proliferation. Immunized mice were injected with tumor cells loaded with p876 and mixed with PADRE. Starting from the day after tumor injection, mice received five rounds of peptide injection at the tumor sites and all tumors were rejected. Injection with saline had no effect. Injection with PADRE had minor anti-tumor activity. Immunization and treatment with p876 alone was not protective. Therefore, by delivering CD4 and CD8 reactive foreign peptides to the tumor, peptide-specific T cells rejected the tumors as demonstrated by the in vitro and in vivo tests.


Human Gene Therapy | 1999

Modulation of cell-mediated immunity prolongs adenovirus-mediated transgene expression in sciatic nerve.

Agnes Jani; Daniela Menichella; Huiyuan Jiang; Taibi Chbihi; Gyula Acsadi; Michael E. Shy; John Kamholz

In a previous report, we demonstrated that a first-generation (E1- and E3-deleted) recombinant adenovirus can transduce expression of the E. coli lacZ gene into Schwann cells, both in vitro and in vivo, suggesting that this method might be useful for future therapy of peripheral neuropathy, including CMT1. Adenovirus-mediated gene transfer was limited, however, by demyelination and Wallerian degeneration at the site of virus injection, as well as by attenuation of viral transgene expression over time. In our current work we have optimized adenoviral vector-mediated transgene expression after intraneural injection into sciatic nerve. Using an improved injection protocol, peak expression of lacZ occurs between 10 and 14 days after injection of 2-week-old rats, decreases thereafter, and there is minimal associated tissue injury. In contrast, few lacZ-expressing Schwann cells are found in nerve of adult animals 10 days after injection, probably owing to immune clearance of virus-infected cells. Consistent with this notion, high levels of LacZ are found in sciatic nerve 30 days after injection of adult SCID mice, which have a genetic defect in both cellular and humoral immunity, of adult beta2-microglobulin-deficient mice (beta2M4-/-), which have a genetic defect in cellular immunity, or of adult mice treated with the immunosuppressing agent FK506. In addition, adenovirus-infected Schwann cells cocultured with axons in vitro, in the absence of a host immune response, ensheathe axons and express lacZ for at least 8 weeks. These data thus demonstrate that lacZ transgene expression of first-generation recombinant adenovirus in sciatic nerve in adult mice, as in other tissues, is limited mainly by the host cellular immune response to the virus, which can be overcome by attenuation of host cell-mediated immunity. Adenoviral vectors might thus be used to modulate Schwann cell gene expression in patients with peripheral neuropathy after appropriate immunosuppression.


Behavioral and Neural Biology | 1980

Differential contribution of fimbria and fornix fibers to behavior.

György Buzsáki; Gyula Acsadi; Agnes Jani

Rats with lesions severing either the subcallosal fornix (Fo) or medial half of the fimbria (Fi) were compared to sham-operated and intact animals in a series of behavioral tasks. Acquisition of Y-maze learning proceeded equally in all groups; the total number of rearing was, however, significantly lower in the lesioned subjects. Place-vs-cue test revealed that all rats utilized place hypothesis. In the cue guided spatial reversal task Fi and Fo rats were equally impaired. In the one trial passive avoidance problem Fo rats were inferior to both Fi and control animals. Open-field test failed to distinguish between the groups. Fo lesions destroyed, while Fi lesions did not affect rhythmic slow activity of the dorsal hippocampus. The results indicate differential involvement of the fibers of the fimbria—fornix complex in behavior.


Annals of the New York Academy of Sciences | 1999

Overcoming Cellular Immunity to Prolong Adenoviral-Mediated Gene Expression in Sciatic Nerve

Agnes Jani; Daniela Menichella; Huiyuan Jiang; Taibi Chbihi; Gyula Acsadi; John Kamholz; Michael E. Shy

ABSTRACT: In a previous report, we demonstrated that a first generation (E1‐ and E3‐deleted) recombinant adenovirus can transduce expression of the E. coli lacZ gene into Schwann cells, both in vitro and in vivo, suggesting that this method might be useful for future therapy of peripheral neuropathy, including CMT1. Adenoviral‐mediated gene transfer was limited, however, by demyelination and Wallerian degeneration at the site of virus injection, as well as by attenuation of viral gene expression over time. In our current work we have optimized adenoviral‐mediated gene expression after intraneural injection into sciatic nerve. Using an improved injection protocol, peak expression of lacZ occurs between 10 and 14 days after injection of 2‐week‐old animals, decreases thereafter, and there is minimal associated tissue injury. In contrast, very few adenoviral‐infected Schwann cells are found in nerves of adult animals 10 days after injection, probably due to immune clearance of viral‐infected cells. Consistent with this notion, high levels of lacZ are found in sciatic nerve 30 days after injection of adult SCID mice, which have a genetic defect in both cellular and humoral immunity, of adult β2 microglobulin‐deficient mice (β2 M −/−), which have a genetic defect in cellular immunity, or of adult mice treated with the immunosuppressing agent FK506. In addition, adenoviral‐infected Schwann cells co‐cultured with axons in vitro, in the absence of a host immune response, ensheath axons and express lacZ for at least 8 weeks. These data thus demonstrate that expression of first generation recombinant adenovirus in sciatic nerve in adult mice, as in other tissues, is limited mainly by the host cellular immune response to the virus, which can be overcome by attenuation of host cell‐mediated immunity. Adenoviral vectors might thus be used to modulate Schwann cell gene expression in patients with peripheral neuropathy after appropriate immunosuppression.

Collaboration


Dive into the Agnes Jani's collaboration.

Top Co-Authors

Avatar

Gyula Acsadi

University of Connecticut

View shared research outputs
Top Co-Authors

Avatar

Bernard Massie

National Research Council

View shared research outputs
Top Co-Authors

Avatar

George Karpati

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jon A. Wolff

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Richard A. Lewis

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge