Network


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

Hotspot


Dive into the research topics where Michael J. Gait is active.

Publication


Featured researches published by Michael J. Gait.


The EMBO Journal | 1990

HIV-1 tat protein stimulates transcription by binding to a U-rich bulge in the stem of the TAR RNA structure.

Colin Dingwall; Ingemar Ernberg; Michael J. Gait; Sheila M. Green; Shaun Heaphy; Jonathan Karn; Anthony D. Lowe; Mohinder Singh; Michael A. Skinner

The HIV‐1 trans‐activator protein, tat, is an RNA binding protein with a high affinity for a U‐rich bulge near the tip of the stem in the RNA stem‐loop structure encoded by the trans‐activation responsive region (TAR). A Scatchard analysis of tat binding has shown that the purified protein forms a one‐to‐one complex with HIV‐1 TAR RNA with a dissociation constant of Kd = 12 nM. Deletion of the uridine residues in the bulge or substitution with guanine residues produced RNAs with a 6‐ to 8‐fold lower affinity than wild‐type TAR. Introduction of a point mutation expected to destabilize base pairing in nearby residues of the TAR stem‐loop structure reduced tat binding 10‐fold. In contrast, mutations that alter the sequence of the six nucleotide long loop at the tip of TAR RNA structure, and mutations which alter the sequence of the stem whilst preserving Watson‐Crick base pairing, do not affect tat binding significantly. There is a direct correlation between the ability of tat to bind to TAR RNA and to activate HIV transcription. Viral LTRs carrying TAR sequences encoding any of the mutations known to produce transcripts which bind tat weakly, are not stimulated efficiently by tat in vivo.


Cellular and Molecular Life Sciences | 2010

Cell penetrating peptides: overview and applications to the delivery of oligonucleotides

F. Said Hassane; Amer F. Saleh; Rachida Abes; Michael J. Gait; Bernard Lebleu

Crossing biological barriers represents a major limitation for clinical applications of biomolecules such as nucleic acids, peptides or proteins. Cell penetrating peptides (CPP), also named protein transduction domains, comprise short and usually basic amino acids-rich peptides originating from proteins able to cross biological barriers, such as the viral Tat protein, or are rationally designed. They have emerged as a new class of non-viral vectors allowing the delivery of various biomolecules across biological barriers from low molecular weight drugs to nanosized particles. Encouraging data with CPP-conjugated oligonucleotides have been obtained both in vitro and in vivo in animal models of diseases such as Duchenne muscular dystrophy. Whether CPP-cargo conjugates enter cells by direct translocation across the plasma membrane or by endocytosis remains controversial. In many instances, however, endosomal escape appears as a major limitation of this new delivery strategy.


Advanced Drug Delivery Reviews | 2008

Cell penetrating peptide conjugates of steric block oligonucleotides

Bernard Lebleu; Hong M. Moulton; Rachida Abes; Gabriela D. Ivanova; Saïd Abes; David A. Stein; Patrick L. Iversen; Andrey A. Arzumanov; Michael J. Gait

Abstract Charge neutral steric block oligonucleotide analogues, such as peptide nucleic acids (PNA) or phosphorodiamidate morpholino oligomers (PMO), have promising biological and pharmacological properties for antisense applications, such as for example in mRNA splicing redirection. However, cellular uptake of free oligomers is poor and the utility of conjugates of PNA or PMO to cell penetrating peptides (CPP), such as Tat or Penetratin, is limited by endosomal sequestration. Two new families of arginine-rich CPPs named (R-Ahx-R)4 AhxB and R6Pen allow efficient nuclear delivery of splice correcting PNA and PMO at micromolar concentrations in the absence of endosomolytic agents. The in vivo efficacy of (R-Ahx-R)4 AhxB PMO conjugates has been demonstrated in mouse models of Duchenne muscular dystrophy and in various viral infections.


Cellular and Molecular Life Sciences | 2003

Peptide-mediated cellular delivery of antisense oligonucleotides and their analogues

Michael J. Gait

Abstract: Improving the delivery of synthetic oligonucleotides and their analogues into cells is an important goal in the full development of antisense technology for control of gene expression in cell culture and in vivo. This review describes the harnessing of certain peptides, either as noncovalent complexes or as covalent conjugates, to enhance the delivery of antisense oligonucleotides into cells and/or to affect their cell localization. Phosphodiester and phosphorothioate oligonucleotides are included as well as peptide nucleic acids (PNAs), analogues of oligonucleotides where the negatively charged phosphate backbone is replaced by a neutral amide linkage. This review contains a critical evaluation of claims for certain peptide-oligonucleotide conjugates to translocate into cultured cells by a non-energy-dependent nonendosomal route. In addition, the available evidence for the utility of stable versus nonstable linkages between peptide and oligonucleotide or PNA is discussed.


Nucleic Acids Research | 2008

Improved cell-penetrating peptide–PNA conjugates for splicing redirection in HeLa cells and exon skipping in mdx mouse muscle

Gabriela D. Ivanova; Andrey A. Arzumanov; Rachida Abes; HaiFang Yin; Matthew J.A. Wood; Bernard Lebleu; Michael J. Gait

Steric blocking peptide nucleic acid (PNA) oligonucleotides have been used increasingly for redirecting RNA splicing particularly in therapeutic applications such as Duchenne muscular dystrophy (DMD). Covalent attachment of a cell-penetrating peptide helps to improve cell delivery of PNA. We have used a HeLa pLuc705 cell splicing redirection assay to develop a series of PNA internalization peptides (Pip) conjugated to an 18-mer PNA705 model oligonucleotide with higher activity compared to a PNA705 conjugate with a leading cell-penetrating peptide being developed for therapeutic use, (R-Ahx-R)(4). We show that Pip-PNA705 conjugates are internalized in HeLa cells by an energy-dependent mechanism and that the predominant pathway of cell uptake of biologically active conjugate seems to be via clathrin-dependent endocytosis. In a mouse model of DMD, serum-stabilized Pip2a or Pip2b peptides conjugated to a 20-mer PNA (PNADMD) targeting the exon 23 mutation in the dystrophin gene showed strong exon-skipping activity in differentiated mdx mouse myotubes in culture in the absence of an added transfection agent at concentrations where naked PNADMD was inactive. Injection of Pip2a-PNADMD or Pip2b-PNADMD into the tibealis anterior muscles of mdx mice resulted in approximately 3-fold higher numbers of dystrophin-positive fibres compared to naked PNADMD or (R-Ahx-R)(4)-PNADMD.


Trends in Biochemical Sciences | 1993

RNA recognition by the human immuno-deficiency virus Tat and Rev proteins

Michael J. Gait; Jonathan Karn

The human immunodeficiency virus (HIV-1) regulatory proteins, Tat and Rev, are important potential targets for the development of new drug therapies against HIV infection. Both proteins are highly specific RNA-binding proteins that recognize cis-acting regulatory elements in the viral mRNAs. These interactions are fascinating paradigms of a new principle of RNA recognition in which the protein makes contact with functional groups displayed in a distorted major groove of an RNA duplex.


Molecular therapy. Nucleic acids | 2012

Pip6-PMO, A New Generation of Peptide-oligonucleotide Conjugates With Improved Cardiac Exon Skipping Activity for DMD Treatment

Corinne Betts; Amer F. Saleh; Andrey A. Arzumanov; Suzan M. Hammond; Caroline Godfrey; Thibault Coursindel; Michael J. Gait; Matthew J.A. Wood

Antisense oligonucleotides (AOs) are currently the most promising therapeutic intervention for Duchenne muscular dystrophy (DMD). AOs modulate dystrophin pre-mRNA splicing, thereby specifically restoring the dystrophin reading frame and generating a truncated but semifunctional dystrophin protein. Challenges in the development of this approach are the relatively poor systemic AO delivery and inefficient dystrophin correction in affected non-skeletal muscle tissues, including the heart. We have previously reported impressive heart activity including high-splicing efficiency and dystrophin restoration following a single administration of an arginine-rich cell-penetrating peptide (CPPs) conjugated to a phosphorodiamidate morpholino oligonucleotide (PMO): Pip5e-PMO. However, the mechanisms underlying this activity are poorly understood. Here, we report studies involving single dose administration (12.5 mg/kg) of derivatives of Pip5e-PMO, consecutively assigned as Pip6-PMOs. These peptide-PMOs comprise alterations to the central hydrophobic core of the Pip5e peptide and illustrate that certain changes to the peptide sequence improves its activity; however, partial deletions within the hydrophobic core abolish its efficiency. Our data indicate that the hydrophobic core of the Pip sequences is critical for PMO delivery to the heart and that specific modifications to this region can enhance activity further. The results have implications for therapeutic PMO development for DMD.


Biochemical Society Transactions | 2007

Cell-penetrating-peptide-based delivery of oligonucleotides: an overview

Rachida Abes; Andrey A. Arzumanov; Hong M. Moulton; Saïd Abes; Gabriela D. Ivanova; P.L. Iversen; Michael J. Gait; Bernard Lebleu

Cationic CPPs (cell-penetrating peptides) have been used largely for intracellular delivery of low-molecular-mass drugs, biomolecules and particles. Most cationic CPPs bind to cell-associated glycosaminoglycans and are internalized by endocytosis, although the detailed mechanisms involved remain controversial. Sequestration and degradation in endocytic vesicles severely limits the efficiency of cytoplasmic and/or nuclear delivery of CPP-conjugated material. Re-routing the splicing machinery by using steric-block ON (oligonucleotide) analogues, such as PNAs (peptide nucleic acids) or PMOs (phosphorodiamidate morpholino oligomers), has consequently been inefficient when ONs are conjugated with standard CPPs such as Tat (transactivator of transcription), R(9) (nona-arginine), K(8) (octalysine) or penetratin in the absence of endosomolytic agents. New arginine-rich CPPs such as (R-Ahx-R)(4) (6-aminohexanoic acid-spaced oligo-arginine) or R(6) (hexa-arginine)-penetratin conjugated to PMO or PNA resulted in efficient splicing correction at non-cytotoxic doses in the absence of chloroquine. SAR (structure-activity relationship) analyses are underway to optimize these peptide delivery vectors and to understand their mechanisms of cellular internalization.


Molecular Medicine | 2001

ARP, a peptide derived from the stress-associated acetylcholinesterase variant has hematopoietic growth promoting activities

Dan Grisaru; Varda Deutsch; Michael Y. Shapira; Marjorie Pick; Meira Sternfeld; Naomi Melamed-Book; Daniela Kaufer; Nilly Galyam; Michael J. Gait; David Owen; Joseph B. Lessing; Hermona Soreq

BackgroundPsychological stress induces rapid and long-lasting changes in blood cell composition, implying the existence of stress-induced factors that modulate hematopoiesis. Here we report the involvement of the stress-associated “readthrough” acetylcholinesterase (AChE-R) variant, and its 26 amino acid C-terminal domain (ARP) in hematopoietic stress responses.Materials and MethodsWe studied the effects of stress, cortisol, antisense oligonucleotides to AChE, and synthetic ARP on peripheral blood cell composition and clonogenic progenitor status in mice under normal and stress conditions, and on purified CD341 cells of human origin. We employed in situ hybridization and immunocytochemical staining to monitor gene expression, and 5-bromo-2-deoxyuridine (BrdU), primary liquid cultures, and clonogenic progenitor assays to correlate AChE-R and ARP with proliferation and differentiation of hematopoietic progenitors.ResultsWe identified two putative glucocorticoid response elements in the human ACHE gene encoding AChE. In human CD341 hematopoietic progenitor cells, cortisol elevated AChE-R mRNA levels and promoted hematopoietic expansion. In mice, a small peptide crossreacting with anti-ARP antiserum appeared in serum following forced swim stress. Ex vivo, ARP was more effective than cortisol and equally as effective as stem cell factor in promoting expansion and differentiation of early hematopoietic progenitor cells into myeloid and megakaryocyte lineages.ConclusionsOur findings attribute a role to AChE-R and ARP in hematopoietic homeostasis following stress, and suggest the use of ARP in clinical settings where ex vivo expansion of progenitor cells is required.


Molecular Therapy | 2011

Pip5 Transduction Peptides Direct High Efficiency Oligonucleotide-mediated Dystrophin Exon Skipping in Heart and Phenotypic Correction in mdx Mice

HaiFang Yin; Amer F. Saleh; Corinne Betts; Patrizia Camelliti; Yiqi Seow; Shirin Ashraf; Andrey A. Arzumanov; Suzan M. Hammond; Thomas Merritt; Michael J. Gait; Matthew J.A. Wood

Induced splice modulation of pre-mRNAs shows promise to correct aberrant disease transcripts and restore functional protein and thus has therapeutic potential. Duchenne muscular dystrophy (DMD) results from mutations that disrupt the DMD gene open reading frame causing an absence of dystrophin protein. Antisense oligonucleotide (AO)-mediated exon skipping has been shown to restore functional dystrophin in mdx mice and DMD patients treated intramuscularly in two recent phase 1 clinical trials. Critical to the therapeutic success of AO-based treatment will be the ability to deliver AOs systemically to all affected tissues including the heart. Here, we report identification of a series of transduction peptides (Pip5) as AO conjugates for enhanced systemic and particularly cardiac delivery. One of the lead peptide-AO conjugates, Pip5e-AO, showed highly efficient exon skipping and dystrophin production in mdx mice with complete correction of the aberrant DMD transcript in heart, leading to >50% of the normal level of dystrophin in heart. Mechanistic studies indicated that the enhanced activity of Pip5e-phosphorodiamidate morpholino (PMO) is partly explained by more efficient nuclear delivery. Pip5 series derivatives therefore have significant potential for advancing the development of exon skipping therapies for DMD and may have application for enhanced cardiac delivery of other biotherapeutics.

Collaboration


Dive into the Michael J. Gait's collaboration.

Top Co-Authors

Avatar

Andrey A. Arzumanov

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Dmitry A. Stetsenko

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amer F. Saleh

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Jonathan Karn

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Bernard Lebleu

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabriela D. Ivanova

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Mohinder Singh

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

John J. Turner

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge