Network


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

Hotspot


Dive into the research topics where Faisal Al-Allaf is active.

Publication


Featured researches published by Faisal Al-Allaf.


Stem Cells | 2006

Characterization and Clinical Application of Human CD34+ Stem/Progenitor Cell Populations Mobilized into the Blood by Granulocyte Colony‐Stimulating Factor

Myrtle Y. Gordon; Nataša Levičar; Madhava Pai; Philippe Bachellier; Ioannis Dimarakis; Faisal Al-Allaf; Hanane M'Hamdi; Tamara Thalji; Jonathan Welsh; Stephen B. Marley; John Davies; Francesco Dazzi; Federica M. Marelli-Berg; Paul Tait; Raymond J. Playford; Long R. Jiao; Steen Jensen; Joanna Nicholls; Ahmet Ayav; Mahrokh Nohandani; Farzin Farzaneh; Joop Gaken; Rikke Dodge; Malcolm Alison; Jane F. Apperley; Robert I. Lechler; Nagy Habib

A phase I study was performed to determine the safety and tolerability of injecting autologous CD34+ cells into five patients with liver insufficiency. The study was based on the hypothesis that the CD34+ cell population in granulocyte colony‐stimulating factor (G‐CSF)‐mobilized blood contains a subpopulation of cells with the potential for regenerating damaged tissue. We separated a candidate CD34+ stem cell population from the majority of the CD34+ cells (99%) by adherence to tissue culture plastic. The adherent and nonadherent CD34+ cells were distinct in morphology, immunophenotype, and gene expression profile. Reverse transcription‐polymerase chain reaction‐based gene expression analysis indicated that the adherent CD34+ cells had the potential to express determinants consistent with liver, pancreas, heart, muscle, and nerve cell differentiation as well as hematopoiesis. Overall, the characteristics of the adherent CD34+ cells identify them as a separate putative stem/progenitor cell population. In culture, they produced a population of cells exhibiting diverse morphologies and expressing genes corresponding to multiple tissue types. Encouraged by this evidence that the CD34+ cell population contains cells with the potential to form hepatocyte‐like cells, we gave G‐CSF to five patients with liver insufficiency to mobilize their stem cells for collection by leukapheresis. Between 1 × 106 and 2 × 108 CD34+ cells were injected into the portal vein (three patients) or hepatic artery (two patients). No complications or specific side effects related to the procedure were observed. Three of the five patients showed improvement in serum bilirubin and four of five in serum albumin. These observations warrant further clinical trials.


Experimental Neurology | 2009

Umbilical cord blood mesenchymal stromal cells are neuroprotective and promote regeneration in a rat optic tract model

Isabel Zwart; Andrew J. Hill; Faisal Al-Allaf; Mili Shah; John Girdlestone; Athirah B.R. Sanusi; Huseyin Mehmet; Roberto Navarrete; Cristina Navarrete; Ling-Sun Jen

Exploitation of the ability of stem cells to protect damaged neuronal tissue may be a more viable strategy than cell replacement for repair of the central nervous system (CNS). In this study we assessed the capacity of human umbilical cord blood (hUCB)-derived mesenchymal stromal cells (MSCs) to protect and promote regeneration of axotomised neurons within the rat optic system. The optic tract of neonatal rats was transected at the level of the lateral geniculate nucleus, and MSCs were introduced into the lesion site. MSCs survived well up to 2 weeks after grafting, and did not migrate significantly or differentiate. In the presence of MSC grafts, host axonal processes were found to be present in the lesion site, and there was stimulation of an endogenous neural precursor population. Four weeks after grafting, retrograde tracer experiments demonstrated that grafted MSCs, as well as cells of a human fibroblast line, exerted a neuroprotective effect, rescuing a significant percentage of axotomised retinal ganglion cells (RGCs). Further experiments with retrograde and anterograde tracers strongly indicated that MSCs could also promote re-growth of axotomised RGCs to their target, the superior colliculus (SC). Further analysis showed that hUCB-derived MSCs secreted several immunomodulatory and neurotrophic factors in vitro, including TGFbeta1, CNTF, NT-3 and BDNF, which are likely to play a role in neuroprotection. Our data indicate that hUCB-derived MSCs may be an easily accessible, widely available source of cells that can contribute towards neural repair through rescue and regeneration of injured neurons.


Reproductive Biomedicine Online | 2008

Microchimeric fetal cells cluster at sites of tissue injury in lung decades after pregnancy

Keelin O'Donoghue; Hanan A. Sultan; Faisal Al-Allaf; Jonathan R. Anderson; Josephine Wyatt-Ashmead; Nicholas M. Fisk

Fetal cells trafficking into maternal blood during pregnancy engraft tissues and persist for decades in marrow and bone. While persistent fetal cells were initially implicated in autoimmune disease, animal studies suggest that microchimeric fetal cells play a broader role in response to tissue injury. This study investigated whether fetal cells participate in tissue repair after human pregnancy. Specimens were obtained from women undergoing surgery for suspected lung cancer. Y-fluorescence in-situ hybridization was performed on paraffin-embedded sections, with the investigator blinded to medical histories. Male cells were identified in lung/thymus tissue from all women with known male pregnancies, but not in those without sons. The frequency of male microchimeric cells was seven-fold greater in lung/thymus tissues than marrow and was two-fold greater than normal bone from the same women. Nested-polymerase chain reaction for sex determining region Y confirmed male DNA in tissues. Male cells in lung were clustered in tumour rather than surrounding healthy tissues. In conclusion, male presumed-fetal cells were identified in pathological post-reproductive tissues, where they were more likely to be located in diseased tissues at several-fold higher frequency than normal tissues. It is suggested that fetal cells are present at sites of tissue injury and may be stem cells, either recruited from marrow or having proliferated locally.


Gene Therapy | 2003

Long-term transgene expression by administration of a lentivirus-based vector to the fetal circulation of immuno-competent mice

Simon N. Waddington; Kyriacos Mitrophanous; Fm Ellard; Suzanne M. K. Buckley; M Nivsarkar; L Lawrence; Ht Cook; Faisal Al-Allaf; Brian Bigger; Susan Mary Kingsman; Charles Coutelle; M Themis

Inefficient gene transfer, inaccessibility of stem cell compartments, transient gene expression, and adverse immune and inflammatory reactions to vector and transgenic protein are major barriers to successful in vivo application of gene therapy for most genetic diseases. Prenatal gene therapy with integrating vectors may overcome these problems and prevent early irreparable organ damage. To this end, high-dose attenuated VSV-G pseudotyped equine infectious anaemia virus (EIAV) encoding β-galactosidase under the CMV promoter was injected into the fetal circulation of immuno-competent MF1 mice. We saw prolonged, extensive gene expression in the liver, heart, brain and muscle, and to a lesser extent in the kidney and lung of postnatal mice. Progressive clustered hepatocyte staining suggests clonal expansion of cells stably transduced. We thus provide proof of principle for efficient gene delivery and persistent transgene expression after prenatal application of the EIAV vector and its potential for permanent correction of genetic diseases.


Cell Cycle | 2009

Differentiation of human fetal mesenchymal stem cells into cells with an oligodendrocyte phenotype

Nigel L. Kennea; Simon N. Waddington; Jerry Ky Chan; Keelin O'Donoghue; D Yeung; Dl Taylor; Faisal Al-Allaf; G Pirianov; M Themis; Ad Edwards; Nicholas M. Fisk; Huseyin Mehmet

The potential of mesenchymal stem cells (MSC) to differentiate into neural lineages has raised the possibility of autologous cell transplantation as a therapy for neurodegenerative diseases. We have identified a population of circulating human fetal mesenchymal stem cells (hfMSC) that are highly proliferative and can readily differentiate into mesodermal lineages such as bone, cartilage, fat and muscle. Here, we demonstrate for the first time that primary hfMSC can differentiate into cells with an oligodendrocyte phenotype both in vitro and in vivo. By exposing hfMSC to neuronal conditioned medium or by introducing the pro-oligodendrocyte gene, Olig-2, hfMSC adopted an oligodendrocyte-like morphology, expressed oligodendrocyte markers and appeared to mature appropriately in culture. Importantly we also demonstrate the differentiation of a clonal population of hfMSC into both mesodermal (bone) and ectodermal (oligodendrocyte) lineages. In the developing murine brain transplanted hfMSC integrated into the parenchyma but oligodendrocyte differentiation of these naïve hfMSC was very low. However, the proportion of cells expressing oligodendrocyte markers increased significantly (from 0.2% to 4%) by pre-exposing the cells to differentiation medium in vitro prior to transplantation. Importantly, the process of in vivo differentiation occurred without cell fusion. These findings suggest that hfMSC may provide a potential source of oligodendrocytes for study and potential therapy.


International Archives of Medicine | 2010

LDLR-Gene therapy for familial hypercholesterolaemia: problems, progress, and perspectives.

Faisal Al-Allaf; Charles Coutelle; Simon N. Waddington; Anna L. David; Richard P. Harbottle; M Themis

Coronary artery diseases (CAD) inflict a heavy economical and social burden on most populations and contribute significantly to their morbidity and mortality rates. Low-density lipoprotein receptor (LDLR) associated familial hypercholesterolemia (FH) is the most frequent Mendelian disorder and is a major risk factor for the development of CAD. To date there is no cure for FH. The primary goal of clinical management is to control hypercholesterolaemia in order to decrease the risk of atherosclerosis and to prevent CAD. Permanent phenotypic correction with single administration of a gene therapeutic vector is a goal still needing to be achieved. The first ex vivo clinical trial of gene therapy in FH was conducted nearly 18 years ago. Patients who had inherited LDLR gene mutations were subjected to an aggressive surgical intervention involving partial hepatectomy to obtain the patients own hepatocytes for ex vivo gene transfer with a replication deficient LDLR-retroviral vector. After successful re-infusion of transduced cells through a catheter placed in the inferior mesenteric vein at the time of liver resection, only low-level expression of the transferred LDLR gene was observed in the five patients enrolled in the trial. In contrast, full reversal of hypercholesterolaemia was later demonstrated in in vivo preclinical studies using LDLR-adenovirus mediated gene transfer. However, the high efficiency of cell division independent gene transfer by adenovirus vectors is limited by their short-term persistence due to episomal maintenance and the cytotoxicity of these highly immunogenic viruses. Novel long-term persisting vectors derived from adeno-associated viruses and lentiviruses, are now available and investigations are underway to determine their safety and efficiency in preparation for clinical application for a variety of diseases. Several novel non-viral based therapies have also been developed recently to lower LDL-C serum levels in FH patients. This article reviews the progress made in the 18 years since the first clinical trial for gene therapy of FH, with emphasis on the development, design, performance and limitations of viral based gene transfer vectors used in studies to ameliorate the effects of LDLR deficiency.


Gene Therapy | 2004

Highly efficient EIAV-mediated in utero gene transfer and expression in the major muscle groups affected by Duchenne muscular dystrophy

Lisa G. Gregory; Simon N. Waddington; Maxine V. Holder; Kyriacos Mitrophanous; Suzanne M. K. Buckley; Karen Mosley; Brian Bigger; Fm Ellard; Le Walmsley; L Lawrence; Faisal Al-Allaf; Susan Mary Kingsman; Charles Coutelle; M Themis

Gene therapy for Duchenne muscular dystrophy has so far not been successful because of the difficulty in achieving efficient and permanent gene transfer to the large number of affected muscles and the development of immune reactions against vector and transgenic protein. In addition, the prenatal onset of disease complicates postnatal gene therapy. We have therefore proposed a fetal approach to overcome these barriers. We have applied β-galactosidase expressing equine infectious anaemia virus (EIAV) lentiviruses pseudotyped with VSV-G by single or combined injection via different routes to the MF1 mouse fetus on day 15 of gestation and describe substantial gene delivery to the musculature. Highly efficient gene transfer to skeletal muscles, including the diaphragm and intercostal muscles, as well as to cardiac myocytes was observed and gene expression persisted for at least 15 months after administration of this integrating vector. These findings support the concept of in utero gene delivery for therapeutic and long-term prevention/correction of muscular dystrophies and pave the way for a future application in the clinic.


Molecular Therapy | 2013

The Fetal Mouse Is a Sensitive Genotoxicity Model That Exposes Lentiviral-associated Mutagenesis Resulting in Liver Oncogenesis

Ali Nowrouzi; Wing Cheung; Tingting Li; Xuegong Zhang; Anne Arens; Anna Paruzynski; Simon N. Waddington; Emma Osejindu; Safia Reja; Christof von Kalle; Yoahe Wang; Faisal Al-Allaf; Lisa G. Gregory; M Themis; Maxine V. Holder; Niraja Dighe; Alaine T. Ruthe; Suzanne M. K. Buckley; Brian Bigger; Eugenio Montini; Adrian J. Thrasher; Robert Andrews; Terry P Roberts; Robert F. Newbold; Charles Coutelle; Manfred Schmidt

Genotoxicity models are extremely important to assess retroviral vector biosafety before gene therapy. We have developed an in utero model that demonstrates that hepatocellular carcinoma (HCC) development is restricted to mice receiving nonprimate (np) lentiviral vectors (LV) and does not occur when a primate (p) LV is used regardless of woodchuck post-translation regulatory element (WPRE) mutations to prevent truncated X gene expression. Analysis of 839 npLV and 244 pLV integrations in the liver genomes of vector-treated mice revealed clear differences between vector insertions in gene dense regions and highly expressed genes, suggestive of vector preference for insertion or clonal outgrowth. In npLV-associated clonal tumors, 56% of insertions occurred in oncogenes or genes associated with oncogenesis or tumor suppression and surprisingly, most genes examined (11/12) had reduced expression as compared with control livers and tumors. Two examples of vector-inserted genes were the Park 7 oncogene and Uvrag tumor suppressor gene. Both these genes and their known interactive partners had differential expression profiles. Interactive partners were assigned to networks specific to liver disease and HCC via ingenuity pathway analysis. The fetal mouse model not only exposes the genotoxic potential of vectors intended for gene therapy but can also reveal genes associated with liver oncogenesis.


13 | 2011

Designing lentiviral gene vectors

Oleg Tolmachov; Tanya Tolmachova; Faisal Al-Allaf

Gene therapy relies on the delivery of therapeutic genes into patients’ cells. The microdevices used to reach the cells and to transfer the gene payload are called gene vectors. Viral packaging machinery is often utilized to generate the particles transporting the cargo genes. Lentiviruses, a subgroup of retroviruses, are highly suitable for remodeling into gene transfer vectors because they offer the stability of transgene expression, the ability to reach the nuclei of the therapeutically important non-dividing cells and are known to have a low immunogenic profile. Well studied members of the lentiviruses include human immunodeficiency viruses 1 and 2 (HIV-1 and HIV-2), feline immunodeficiency virus (FIV) and equine infectious anemia virus (EIAV). It is important not to confuse “gene delivery vectors” and “gene cloning vectors”. While the former are microparticles delivering genes, the latter are replicating vehicles for the amplification of nucleic acid sequences. “Gene delivery vectors” and “gene cloning vectors” coincide when the naked DNA of replicating bacterial plasmids or replication competent viruses is used for gene delivery into cells. Viral gene delivery vectors are normally nonreplicating and should correctly be referred to as “viral vectors”, not “viruses”. Particles of viral vectors can be referred to as “virions” or “transducing particles”, because viral gene transfer is traditionally described as “transduction”. Replication deficient viral gene vector particles are similar to “defective interfering particles”, that is, faulty non-self-viable virions arising during natural viral infections and competing with non-defective virions, which were described in virology literature many years ago. Native lentiviral envelope proteins, which determine the cell range of viral infectivity (tropism) and mediate the fusion of viral and cellular membranes, are always composed from two non-covalently attached subunits, one of which (e.g. gp41 glycoprotein in HIV-1) is membrane-embedded and the other is an external subunit (e.g. gp120 glycoprotein in HIV-1). This arrangement makes lentiviruses notoriously unstable because of their tendency to shed the external subunit of the envelope protein. As the virion’s stability is a pre-requisite for the effective purification and concentration of viral vector preparations, in


Molecular Therapy | 2006

Erratum: "Oncogenesis following delivery of a nonprimate lentiviral gene therapy vector to fetal and neonatal mice" (Molecular Therapy (2005) vol. 12 (763-771) 10.1016/j.ymthe.2005.07.358)

M Themis; Simon N. Waddington; Manfred Schmidt; Christof von Kalle; Yoahe Wang; Faisal Al-Allaf; Lisa G. Gregory; M Nivsarkar; Maxine V. Holder; Suzanne M. K. Buckley; Niraja Dighe; Alaine T. Ruthe; A Mistry; Brian Bigger; Ahad A. Rahim; Tuan H. Nguyen; Didier Trono; Adrian J. Thrasher; Charles Coutelle

The authors regret that in Table 2 on page 768, one of the insertion sites of the SMART 2 provirus vector identified using LAM-PCR as present on chromosome 5 positioned 32374 bp upstream of Cyp3a11 was incorrectly assigned to Mouse (tumour) 2 T1. This insertion site should be assigned to an independent mouse not listed in Table 2. This animal had only a single provirus insertion found by Southern and LAM-PCR analyses and should be labeled as mouse 7.

Collaboration


Dive into the Faisal Al-Allaf's collaboration.

Top Co-Authors

Avatar

M Themis

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian Bigger

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa G. Gregory

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

M Nivsarkar

Imperial College London

View shared research outputs
Top Co-Authors

Avatar

A Mistry

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge