Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Sci Adv ; 6(19): eaaz0571, 2020 05.
Article in English | MEDLINE | ID: mdl-32494707

ABSTRACT

The prototypical genetic autoimmune disease is immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome, a severe pediatric disease with limited treatment options. IPEX syndrome is caused by mutations in the forkhead box protein 3 (FOXP3) gene, which plays a critical role in immune regulation. As a monogenic disease, IPEX is an ideal candidate for a therapeutic approach in which autologous hematopoietic stem and progenitor (HSPC) cells or T cells are gene edited ex vivo and reinfused. Here, we describe a CRISPR-based gene correction permitting regulated expression of FOXP3 protein. We demonstrate that gene editing preserves HSPC differentiation potential, and that edited regulatory and effector T cells maintain their in vitro phenotype and function. Additionally, we show that this strategy is suitable for IPEX patient cells with diverse mutations. These results demonstrate the feasibility of gene correction, which will be instrumental for the development of therapeutic approaches for other genetic autoimmune diseases.


Subject(s)
Gene Editing , Genetic Diseases, X-Linked , Child , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/therapy , Humans , Mutation , Phenotype , T-Lymphocytes, Regulatory
2.
Sci Rep ; 7(1): 15820, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29150659

ABSTRACT

FOXP3 is the transcription factor ruling regulatory T cell function and maintenance of peripheral immune tolerance, and mutations in its coding gene causes IPEX autoimmune syndrome. FOXP3 is also a cell-cycle inhibitor and onco-suppressor in different cell types. In this work, we investigate the effect of ectopic FOXP3 expression on HSC differentiation and we challenged this approach as a possible HSC-based gene therapy for IPEX. FOXP3-expressing HSC showed reduced proliferation ability and increased maintenance of primitive markers in vitro in both liquid and OP9-ΔL1 co-cultures. When transplanted into immunodeficient mice, FOXP3-expressing HSC showed significantly enhanced engraftment ability. This was due to a pronounced increase in the frequency of repopulating cells, as assessed by extreme limiting dilution assay. Likely underlying the increased repopulating ability, FOXP3 expressing HSC showed significantly enhanced expression of genes controlling stemness features. However, peripheral T cells developed in the FOXP3-humanized mice were quantitatively reduced and hyporesponsive to cytokine and polyclonal stimulation. Our findings reveal unpredicted effects of FOXP3 in the biology of HSC and may provide new tools to manipulate primitive features in HSC for clinical applications. Moreover, they formally prove the need of preserving endogenous FOXP3 regulation for an HSC-based gene therapy approach for IPEX syndrome.


Subject(s)
Cell Differentiation , Forkhead Transcription Factors/metabolism , Hematopoietic Stem Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Cells, Cultured , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Humans , Mice
4.
Mol Genet Metab ; 115(1): 48-52, 2015 May.
Article in English | MEDLINE | ID: mdl-25796965

ABSTRACT

Metachromatic Leukodystrophy (MLD; MIM# 250100) is a rare inherited lysosomal storage disorder caused by the deficiency of Arylsulfatase A (ARSA). The enzymatic defect results in the accumulation of the ARSA substrate that is particularly relevant in myelin forming cells and leads to progressive dysmyelination and dysfunction of the central and peripheral nervous system. Sulfatide accumulation has also been reported in various visceral organs, although little is known about the potential clinical consequences of such accumulation. Different forms of MLD-associated gallbladder disease have been described, and there is one reported case of an MLD patient presenting with functional consequences of sulfatide accumulation in the kidney. Here we describe a wide cohort of MLD patients in whom a tendency to sub-clinical metabolic acidosis was observed. Furthermore in some of them we report episodes of metabolic acidosis of different grades of severity developed in acute clinical conditions of various origin. Importantly, we finally show how a careful acid-base balance monitoring and prompt correction of imbalances might prevent severe consequences of acidosis.


Subject(s)
Acidosis/complications , Leukodystrophy, Metachromatic/complications , Leukodystrophy, Metachromatic/metabolism , Monitoring, Physiologic , Acid-Base Equilibrium , Acid-Base Imbalance , Acidosis/blood , Acidosis/prevention & control , Acidosis/urine , Child , Child, Preschool , Cohort Studies , Follow-Up Studies , Genotype , Humans , Infant , Retrospective Studies , Time Factors
5.
Leukemia ; 29(2): 396-405, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24897508

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) from human leukocyte antigen (HLA) haploidentical family donors is a promising therapeutic option for high-risk hematologic malignancies. Here we explored in 121 patients, mostly with advanced stage diseases, a sirolimus-based, calcineurin-inhibitor-free prophylaxis of graft-versus-host disease (GvHD) to allow the infusion of unmanipulated peripheral blood stem cell (PBSC) grafts from partially HLA-matched family donors (TrRaMM study, Eudract 2007-5477-54). Conditioning regimen was based on treosulfan and fludarabine, and GvHD prophylaxis on antithymocyte globulin Fresenius (ATG-F), rituximab and oral administration of sirolimus and mycophenolate. Neutrophil and platelet engraftment occurred in median at 17 and 19 days after HSCT, respectively, and full donor chimerism was documented in patients' bone marrow since the first post-transplant evaluation. T-cell immune reconstitution was rapid, and high frequencies of circulating functional T-regulatory cells (Treg) were documented during sirolimus prophylaxis. Incidence of acute GvHD grade II-IV was 35%, and occurrence and severity correlated negatively with Treg frequency. Chronic GvHD incidence was 47%. At 3 years after HSCT, transpant-related mortality was 31%, relapse incidence 48% and overall survival 25%. In conclusion, GvHD prophylaxis with sirolimus-mycophenolate-ATG-F-rituximab promotes a rapid immune reconstitution skewed toward Tregs, allowing the infusion of unmanipulated haploidentical PBSC grafts.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , HLA Antigens/immunology , Peripheral Blood Stem Cell Transplantation , Sirolimus/therapeutic use , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Adolescent , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antilymphocyte Serum/therapeutic use , Blood Platelets/cytology , Busulfan/analogs & derivatives , Busulfan/therapeutic use , Child , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Neutrophils/cytology , Prospective Studies , Rituximab , T-Lymphocytes/immunology , Tissue Donors , Transplantation Conditioning , Treatment Outcome , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use , Young Adult
6.
Am J Transplant ; 13(8): 1963-75, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23834659

ABSTRACT

The immune system is comprised of several CD4(+) T regulatory (Treg) cell types, of which two, the Foxp3(+) Treg and T regulatory type 1 (Tr1) cells, have frequently been associated with transplant tolerance. However, whether and how these two Treg-cell types synergize to promote allograft tolerance remains unknown. We previously developed a mouse model of allogeneic transplantation in which a specific immunomodulatory treatment leads to transplant tolerance through both Foxp3(+) Treg and Tr1 cells. Here, we show that Foxp3(+) Treg cells exert their regulatory function within the allograft and initiate engraftment locally and in a non-antigen (Ag) specific manner. Whereas CD4(+) CD25(-) T cells, which contain Tr1 cells, act from the spleen and are key to the maintenance of long-term tolerance. Importantly, the role of Foxp3(+) Treg and Tr1 cells is not redundant once they are simultaneously expanded/induced in the same host. Moreover, our data show that long-term tolerance induced by Foxp3(+) Treg-cell transfer is sustained by splenic Tr1 cells and functionally moves from the allograft to the spleen.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , CD4 Antigens/immunology , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Graft Survival , Islets of Langerhans/metabolism , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
7.
Tissue Antigens ; 79(5): 326-32, 2012 May.
Article in English | MEDLINE | ID: mdl-22489942

ABSTRACT

Polymorphisms in the 3' untranslated region (3'UTR) of HLA-G, an important player in immunological tolerance, could be involved in post-transcriptional expression control, and their association with different clinical immune-related conditions including autoimmunity and transplantation is of mounting interest. Most studies have focused on a 14 base pair (bp) insertion/deletion (ins/del), while additional single-nucleotide polymorphisms (SNPs) in the HLA-G 3'UTR have been described but not extensively investigated for their clinical relevance. Here we have comparatively studied the association between 3'UTR haplotypes of HLA-G, or the 14 bp ins/del, with clinical outcome of HLA-identical sibling hematopoietic stem cell transplantation (HSCT) in 147 Middle Eastern beta-thalassemia patients. Sequence based typing of 3'UTR HLA-G polymorphisms in the patients and in 102 healthy Italian blood donors showed strong linkage disequilibrium between the 14 bp ins/del and five 3'UTR SNPs, which together could be arranged into eight distinct haplotypes based on expectation-maximization studies, with four predominant haplotypes (UTRs1-4). After HSCT, we found a moderate though not significant association between the presence of UTR-2 in double dose and protection from acute graft versus host disease (hazard ratio (HR) 0.45, 95% confidence intervals (CI): 0.14-1.45; P = 0.18), an effect that was also seen when the corresponding 14 bp ins/ins genotype was considered alone (HR 0.42, 95% CI: 0.16-1.06; P = 0.07). No association was found with rejection or survival. Taken together, our data show that there is no apparent added value of considering entire 3'UTR HLA-G haplotypes for risk prediction after allogeneic HSCT for beta-thalassemia.


Subject(s)
3' Untranslated Regions/genetics , Graft vs Host Disease/genetics , HLA-G Antigens/genetics , Hematopoietic Stem Cell Transplantation , beta-Thalassemia/genetics , 3' Untranslated Regions/immunology , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Female , Genotype , Graft vs Host Disease/diagnosis , Graft vs Host Disease/immunology , Haplotypes/genetics , Haplotypes/immunology , Humans , Immune Tolerance , Italy , Linkage Disequilibrium , Male , Mutagenesis, Insertional , Polymorphism, Genetic , Sequence Deletion , Siblings , Transplantation, Homologous , Treatment Outcome , beta-Thalassemia/immunology , beta-Thalassemia/therapy
8.
J Autoimmun ; 38(1): 49-58, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22264504

ABSTRACT

Immune dysregulation, Polyendocrinopathy, Enteropathy X-linked (IPEX) syndrome is a unique example of primary immunodeficiency characterized by autoimmune manifestations due to defective regulatory T (Treg) cells, in the presence of FOXP3 mutations. However, autoimmune symptoms phenotypically resembling IPEX often occur in the absence of detectable FOXP3 mutations. The cause of this "IPEX-like" syndrome presently remains unclear. To investigate whether a defect in Treg cells sustains the immunological dysregulation in IPEX-like patients, we measured the amount of peripheral Treg cells within the CD3(+) T cells by analysing demethylation of the Treg cell-Specific-Demethylated-Region (TSDR) in the FOXP3 locus and demethylation of the T cell-Specific-Demethylated-Region (TLSDR) in the CD3 locus, highly specific markers for stable Treg cells and overall T cells, respectively. TSDR demethylation analysis, alone or normalized for the total T cells, showed that the amount of peripheral Treg cells in a cohort of IPEX-like patients was significantly reduced, as compared to both healthy subjects and unrelated disease controls. This reduction could not be displayed by flow cytometric analysis, showing highly variable percentages of FOXP3(+) and CD25(+)FOXP3(+) T cells. These data provide evidence that a quantitative defect of Treg cells could be considered a common biological hallmark of IPEX-like syndrome. Since Treg cell suppressive function was not impaired, we propose that this reduction per se could sustain autoimmunity.


Subject(s)
DNA Methylation , Forkhead Transcription Factors/genetics , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adolescent , Adult , CD3 Complex/immunology , CD3 Complex/metabolism , Child , Child, Preschool , Cohort Studies , Female , Flow Cytometry , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Infant , Male , Syndrome , Young Adult
9.
Gene Ther ; 19(12): 1150-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22189416

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency caused by the defective expression of the WAS protein (WASP) in hematopoietic cells. It has been shown that dendritic cells (DCs) are functionally impaired in WAS patients and was(-/-) mice. We have previously demonstrated the efficacy and safety of a murine model of WAS gene therapy (GT), using stem cells transduced with a lentiviral vector (LV). The aim of this study was to investigate whether GT can correct DC defects in was(-/-) mice. As DCs expressing WASP were detected in the secondary lymphoid organs of the treated mice, we tested the in vitro and in vivo function of bone marrow-derived DCs (BMDCs). The BMDCs showed efficient in vitro uptake of latex beads and Salmonella typhimurium. When BMDCs from the treated mice (GT BMDCs) and the was(-/-) mice were injected into wild-type hosts, we found a higher number of cells that had migrated to the draining lymph nodes compared with mice injected with was(-/-) BMDCs. Finally, we found that ovalbumin (OVA)-pulsed GT BMDCs or vaccination of GT mice with anti-DEC205 OVA fusion protein can efficiently induce antigen-specific T-cell activation in vivo. These findings show that WAS GT significantly improves DC function, thus adding new evidence of the preclinical efficacy of LV-mediated WAS GT.


Subject(s)
Dendritic Cells/immunology , Genetic Therapy , Lentivirus/genetics , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/therapy , Animals , Bone Marrow Cells/immunology , Cell Movement , Dendritic Cells/metabolism , Humans , Lymphoid Tissue/metabolism , Mice , Models, Genetic , Phagocytosis , Transduction, Genetic , Wiskott-Aldrich Syndrome Protein/metabolism
10.
Bone Marrow Transplant ; 45(6): 1047-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19881553

ABSTRACT

Mixed chimerism (MC) and secondary graft failure are frequent events following SCT for thalassemia. There is limited information regarding the outcome of donor lymphocyte infusion (DLI) to prevent rejection, mainly from case reports describing only successful cases. We describe a series of seven children affected by beta-thalassemia treated with escalating doses of DLI for level 2-3 MC (donor<90%) following myeloablative SCT from a matched family donor. The infusions were safe and no acute or chronic GVHD were documented; five patients experienced neutropenia and thrombocytopenia resolving spontaneously. DLI was successful in converting to full donor chimerism two patients stratified in the low-risk class (Pesaro class II). Conversely, for five high-risk patients, DLI was not effective in preventing secondary graft failure. This limited series suggests that escalating doses of DLI are safe in thalassemia patients post myeloablative therapy but efficacy may be jeopardized by rapidly growing anti-donor alloimmunity in high-risk patients. We suggest giving escalating doses of donor T cells to attempt a graft-versus-thalassemia as soon as level 2-3 MC is detected.


Subject(s)
Graft Rejection/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Lymphocyte Transfusion/methods , beta-Thalassemia/therapy , Adolescent , Child , Child, Preschool , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Lymphocyte Count , Male , Myeloablative Agonists/therapeutic use , Transplantation Chimera , Treatment Outcome
12.
Clin Genet ; 74(4): 349-57, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18786133

ABSTRACT

Metachromatic leukodystrophy (MLD) is a rare lysosomal storage disorder resulting from the inherited deficiency of the arylsulfatase A (ARSA) enzyme. Currently, no valid therapeutic options are available for affected patients. A thorough knowledge of disease progression in its diverse clinical variants, together with the identification of reliable prognostic factors, could be instrumental in accurate patient selection for new upcoming therapeutic opportunities, such as enzyme replacement and gene therapy. The described correlation between genotype and clinical presentation proved helpful in predicting patient's prognosis, only in the minority of MLD patients harboring common mutations. Molecular characterization of a cohort of 26 MLD patients allowed us to identify 18 mutations, excluding the common 0 and R alleles, 10 of which are rare and 8 are novel. By categorizing the rare mutations, we were able to confirm a correlation between ARSA gene mutations, age at onset and patterns of disease progression, not only in those patients bearing common mutations, but also in those carrying rare mutant alleles. Moreover, in the case of absent or delayed molecular diagnosis, or of newly identified mutations, the involvement of peripheral nervous system from disease onset proved to be a sensitive prognostic marker predicting a severe progression.


Subject(s)
Genotype , Leukodystrophy, Metachromatic/diagnosis , Leukodystrophy, Metachromatic/genetics , Mutation/genetics , Alleles , Brain/pathology , Cerebroside-Sulfatase/genetics , Cohort Studies , DNA Mutational Analysis , Family , Female , Humans , Leukodystrophy, Metachromatic/enzymology , Male , Phenotype
13.
Bone Marrow Transplant ; 42(6): 379-84, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18574444

ABSTRACT

Allogeneic BMT represents the only chance of cure for beta-thalassemia. Occasionally, two affected individuals from the same family share a matched healthy sibling. Moreover, a high incidence of transplant rejection is still observed in Pesaro class III patients, requiring a second BMT procedure. In these settings, one option is to perform a second BM harvest from the same donor. Although BM harvest is a safe procedure in children, ethical issues concerning this invasive practice still arise. Here, we describe our series of seven pediatric, healthy donors, who donated BM more than once in favor of their beta-thalassemic HLA-identical siblings between June 2005 and January 2008. Three donors donated BM twice to two affected siblings and four donors donated twice for the same sibling following graft rejection of the first BMT. All donors tolerated the procedures well and no relevant side effects occurred. There was no significant difference between the two harvests concerning cell yield and time to engraftment. Our experience shows that for pediatric donors, a second BM donation is safe and feasible and good cellularity can be obtained. We suggest that a second harvest of a pediatric donor can be performed when a strong indication for BMT exists.


Subject(s)
Bioethical Issues , Bone Marrow Transplantation/ethics , Bone Marrow , Donor Selection/ethics , Living Donors/ethics , Safety , beta-Thalassemia/therapy , Adolescent , Child , Child, Preschool , Donor Selection/methods , Female , HLA Antigens , Humans , Male , Retrospective Studies , Siblings , Transplantation, Homologous
14.
Bone Marrow Transplant ; 42(6): 397-404, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18574445

ABSTRACT

There is a substantial incidence of graft failure in patients with thalassemia after myeloablative conditioning regimens especially in class 3 patients in whom its incidence could be as high as 8-38.5%. Most patients with graft failure have recurrence of thalassemic marrow. Historically, results of second transplants for thalassemia were poor because of a high rejection rate and/or increased TRM. Sixteen patients with thalassemia recurrence following rejection of the first graft and with a median age of 9 years (range, 4-20) were given second transplants using BM (n=7) or PBSC (n=9) after preparation with a new treatment protocol. All but two patients received stem cells from the same donor. The median interval between two transplants was 28 months (range, 8-204). The sustained engraftment rate was high (94%) with only one patient having primary graft failure. The probability of overall survival, event-free survival, TRM and graft failure were 79, 79, 16 and 6%, respectively. There were three transplant-related deaths. Thirteen patients are alive with Lansky/Karnofsky score of 100. This intensified treatment protocol was well tolerated with no significant increase in toxicity. The excellent results obtained with this new preparative regimen allow us to recommend it for second transplantation for patients with thalassemia recurrence.


Subject(s)
Graft Rejection , Graft Survival , Thalassemia/therapy , Transplantation Conditioning , Adolescent , Adult , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Male , Prospective Studies , Recurrence , Siblings , Survival Rate , Thalassemia/mortality , Time Factors
15.
Gene Ther ; 14(22): 1555-63, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17984995

ABSTRACT

This is the second part of a review summarizing progress and prospects in gene therapy clinical research. Twenty key diseases/strategies are succinctly described and commented on by leaders in the field. This part includes clinical trials for skin diseases, neurological disorders, HIV/AIDS, ornithine transcarbamylase deficiency, alpha(1)-antitrypsin deficiency, haemophilia and cancer.


Subject(s)
Genetic Therapy/trends , Clinical Trials as Topic , Gene Transfer Techniques/adverse effects , Gene Transfer Techniques/trends , Genetic Therapy/methods , Genetic Vectors , Humans , Neoplasms/therapy , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/trends
16.
J Gene Med ; 8(8): 962-71, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16732552

ABSTRACT

BACKGROUND: Demyelination in globoid cell leukodystrophy (GLD) is due to a deficiency of galactocerebrosidase (GALC) activity. Up to now, in vivo brain viral gene transfer of GALC showed modest impact on disease development in Twitcher mice, an animal model for GLD. Lentiviral vectors, which are highly efficient to transfer the expression of therapeutic genes in neurons and glial cells, have not been evaluated for direct cerebral therapy in GLD mice. METHODS: Lentiviral vectors containing the untagged cDNA or the hemagglutinin (HA)-tagged cDNA for the full-length mouse GALC sequence were generated and validated in vitro. In vivo therapeutic efficacy of these vectors was evaluated by histology, biochemistry and electrophysiology after transduction of ependymal or subependymal layers in young Twitcher pups. RESULTS: Both GALC lentiviral vectors transduced neurons, oligodendrocytes and astrocytes with efficiencies above 75% and conferred high levels of enzyme activity. GALC accumulated in lysosomes of transduced cells and was also secreted to the extracellular medium. Conditioned GALC medium was able to correct the enzyme deficiency when added to non-transduced Twitcher glial cultures. Mice that received intraventricular injections of GALC vector showed accumulation of GALC in ependymal cells but no diffusion of the enzyme from the ependymal ventricular tree into the cerebral parenchyma. Significant expression of GALC-HA was detected in neuroglioblasts when GALC-HA lentiviral vectors were injected in the subventricular zone of Twitcher mice. Life span and motor conduction in both groups of treated Twitcher mice were not significantly ameliorated. CONCLUSIONS: Lentiviral vectors showed to be efficient for reconstitution of the GALC expression in Twitcher neural cells. GALC was able to accumulate in lysosomes as well as to enter the secretory pathway of lysosomal enzymes, two fundamental aspects for gene therapy of lysosomal storage diseases. Our in vivo results, while showing the capacity of lentiviral vectors to transfer expression of therapeutic GALC in the Twitcher brain, did not limit progression of disease in Twitchers and highlight the need to evaluate other routes of administration.


Subject(s)
Brain/metabolism , Galactosylceramidase/genetics , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Lentivirus/genetics , Action Potentials/physiology , Animals , Animals, Newborn , Astrocytes/metabolism , Biological Assay , Brain/cytology , Brain/physiology , Cells, Cultured , Culture Media, Conditioned/pharmacology , DNA, Complementary , Disease Models, Animal , Galactosylceramidase/analysis , Genetics , HeLa Cells , Hemagglutinins/chemistry , Homozygote , Humans , Immunohistochemistry , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/pathology , Leukodystrophy, Globoid Cell/therapy , Lysosomes/enzymology , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/metabolism , Oligodendroglia/metabolism
17.
Cytotherapy ; 7(2): 158-65, 2005.
Article in English | MEDLINE | ID: mdl-16040395

ABSTRACT

Effective resetting of the immune system cannot be achieved by non-specific immunosuppression. Instead, novel strategies aim at harnessing the body's natural tolerance mechanisms to rectify an Ag-specific response without disturbing other immune functions. Fine-tuning of the balance between Ag-specific effector and regulatory T (Tr) cells is a promising strategy that requires detailed understanding of the differentiation and expansion pathways of the relevant Tr cell subsets. Here we review recent developments regarding the control of alloreactivity by induction and expansion of Tr cells. T-cell activation in the presence of tolerogenic APC and cytokines leads to the induction of Tr cells, which can mediate tolerance through cytokine-dependent and/or contact-dependent mechanisms. Better understanding of the mechanisms of immune regulation mediated by Tr cells may enable fine-tuning of specific immune responses and pave the way for novel therapeutic approaches.


Subject(s)
Immune Tolerance , Self Tolerance , T-Lymphocytes/transplantation , Animals , Antigen-Presenting Cells/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Humans , Immune Tolerance/immunology , Self Tolerance/immunology , Stem Cell Transplantation , T-Lymphocytes/immunology , Transplantation, Homologous
18.
Curr Top Microbiol Immunol ; 293: 303-26, 2005.
Article in English | MEDLINE | ID: mdl-15981486

ABSTRACT

T regulatory (Tr) cells have an essential role in the induction and maintenance of tolerance to both and foreign self-antigens. Many types of T cells with regulatory activity have been described in mice and humans, and those within the CD4+ subset have been extensively characterized. CD4+ Type-1 regulatory T (Tr1) cells produce high levels of IL-10 and mediate IL-10-dependent suppression, whereas the effects of naturally occurring CD4+CD25+ Tr cells appear to be cell-contact-dependent. Tr1 cells arise in the periphery upon encountering antigen in a tolerogenic environment. In contrast, it appears that CD4+CD25+ Tr cells can either arise directly in the thymus or be induced by antigen in the periphery. We have been interested in defining the phenotype and function of different subsets of CD4+ Tr cells present in human peripheral blood, with the ultimate aim of designing therapeutic strategies to harness their immunoregulatory effects. This review will discuss the similarities and differences between human Tr1 and naturally occurring CD4+CD25+ Tr cells, as well as evidence that indicates that they have nonoverlapping, but synergistic roles in immune homeostasis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , Animals , Humans , Immunophenotyping , T-Lymphocyte Subsets/immunology
19.
J Neurosci Res ; 81(4): 597-604, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15948181

ABSTRACT

We have used magnetic resonance imaging (MRI) and motor evoked potentials (MEPs) for monitoring disease progression within the CNS of the Twitcher mouse, the murine model for globoid cell leukodystrophy (GLD). GLD is a lysosomal storage disorder, resulting from galactocerebrosidase deficiency, causing central and peripheral myelin impairment, leading to death, usually during early infancy. Neuroradiological, electrophysiological, and pathological parameters of myelin maturation were evaluated in Twitcher mice between postnatal days 20 and 45. Healthy controls showed a gradual-appearance MRI T2-weighted hypointensity of the corpus callosum (CC) starting at about P30 and ending at about P37, whereas MRI of age-matched Twitcher mice showed a complete loss of the CC-related MRI signal. MEPs allowed the functional assessment of myelin maturation within corticospinal motor pathways and showed a progressive deterioration of MEPs in Twitcher mice with increased central conduction time (CCT; 5.12 +/- 0.49 msec at P27 to 6.45 +/- 1.96 msec at P32), whereas physiological CCT shortening was found in healthy controls (3.01 +/- 0.81 msec at P27 to 2.5 +/- 0.27 msec at P32). These findings were not paralleled by traditional histological stainings. Optical observation of Bielchowsky and Luxol fast blue-PAS stainings showed mild axonal/myelin deterioration of the Twitcher brain within this time frame. Our results demonstrate that serial MRI and MEP readings are sensitive evaluation tools for in vivo monitoring of dysmyelination in Twitcher mice and underscore their potential use for longitudinal evaluation of the therapeutic impact of gene and cell therapies on these animals.


Subject(s)
Evoked Potentials, Motor , Leukodystrophy, Globoid Cell/pathology , Leukodystrophy, Globoid Cell/physiopathology , Magnetic Resonance Imaging , Myelin Sheath/pathology , Animals , Axons/pathology , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Sciatic Nerve/physiology
20.
Transplant Proc ; 37(1): 65-6, 2005.
Article in English | MEDLINE | ID: mdl-15808548

ABSTRACT

When engrafted with donor stem cells and lymphoid cells, patients develop transplantation tolerance to donor antigens. We analyzed the mechanism of tolerance induction in immunoincompetent recipients whose immunity has been reconstituted by transplantation of mismatched stem cells. Seven infants or human fetuses received fetal liver transplants as a treatment for severe combined immunodeficiency disease. After reconstitution of immunity by lymphocytes developed from donor stem cells, T-cell clones were produced and analyzed. Because donors and recipients were HLA mismatched, it was easy to demonstrate the donor origin of the T-cell clones. These clones were shown to have developed tolerance to histocompatibility antigens of the stem cell donor via a process of clonal deletion (probably as a result of contact with donor-derived macrophages and dendritic cells). They were also tolerant to histocompatibility antigens of the host but through a different mechanism: many clones recognized these antigens but had no detrimental effect on the target cells exhibiting host antigens, either in vitro or in vivo. Clonal anergy was therefore the cause of this tolerance to host determinants, resulting in a lack of graft-versus-host disease and of autoimmunity. The contact between developing T cells of donor origin and host epithelial cells within the host thymus may explain this colonal anergy. It should be noted that all patients had high serum levels of interleukin-10, which might have contributed to the persistent engraftment and tolerance.


Subject(s)
Fetal Tissue Transplantation/immunology , Isoantigens/immunology , Transplantation Tolerance/immunology , Humans , Infant , Severe Combined Immunodeficiency/embryology , Severe Combined Immunodeficiency/surgery , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transplantation, Homologous/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...