Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 769
Filter
Add more filters

Publication year range
1.
Eur Cell Mater ; 36: 171-183, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30334244

ABSTRACT

Robust cellular bioenergetics is vital in the energy-demanding process of maintaining matrix homeostasis in the intervertebral disc. Age-related decline in disc cellular bioenergetics is hypothesised to contribute to the matrix homeostatic perturbation observed in intervertebral disc degeneration. The present study aimed to measure how ageing impacted disc cell mitochondria and bioenergetics. Age-related changes measured included matrix content and cellularity in disc tissue, as well as matrix synthesis, cell proliferation and senescence markers in cell cultures derived from annulus fibrosus (AF) and nucleus pulposus (NP) isolated from the discs of young (6-9 months) and older (36-50 months) New Zealand White rabbits. Cellular bioenergetic parameters were measured using a Seahorse XFe96 Analyzer, in addition to quantitating mitochondrial morphological changes and membrane potential. Ageing reduced mitochondrial number and membrane potential in both cell types. Also, it significantly reduced glycolytic capacity, mitochondrial reserve capacity, maximum aerobic capacity and non-glucose-dependent respiration in NP. Moreover, NP cells exhibited age-related decline in matrix synthesis and reduced cellularity in older tissues. Despite a lack of changes in mitochondrial respiration with age, AF cells showed an increase in glycolysis and altered matrix production. While previous studies report age-related matrix degenerative changes in disc cells, the present study revealed, for the first time, that ageing affected mitochondrial number and function, particularly in NP cells. Consequently, age-related bioenergetic changes may contribute to the functional alterations in aged NP cells that underlie disc degeneration.


Subject(s)
Aging/metabolism , Energy Metabolism , Intervertebral Disc/cytology , Intervertebral Disc/metabolism , Mitochondria/metabolism , Acids/metabolism , Animals , Annulus Fibrosus/metabolism , Biomarkers/metabolism , Cell Death , Cell Proliferation , Cellular Senescence , Extracellular Matrix/metabolism , Extracellular Space/metabolism , Female , Glycolysis , Membrane Potential, Mitochondrial , Nucleus Pulposus/metabolism , Oxidative Phosphorylation , Oxygen Consumption , Rabbits
2.
Biotechnol Lett ; 38(5): 787-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26892223

ABSTRACT

OBJECTIVES: The adhesion of colloidal probes of stainless steel, glass and cellulose to Pseudomonas fluorescens biofilms was examined using atomic force microscopy (AFM) to allow comparisons between surfaces to which biofilms might adhere. RESULTS: Biofilm was grown on a stainless steel substrate and covered most of the surface after 96 h. AFM approach and retraction curves were obtained when the biofilm was immersed in a tryptone/soy medium. On approach, all the colloidal probes experienced a long non-contact phase more than 100 nm in length, possibly due to the steric repulsion by extracellular polymers from the biofilm and hydrophobic effects. Retraction data showed that the adhesion varied from position to position on the biofilm. The mean value of adhesion of glass to the biofilm (48 ± 7 nN) was the greatest, followed by stainless steel (30 ± 7 nN) and cellulose (7.8 ± 0.4 nN). CONCLUSION: The method allows understanding of adhesion between the three materials and biofilm, and development of a better strategy to remove the biofilm from these surfaces relevant to different industrial applications.


Subject(s)
Bacterial Adhesion , Biofilms/growth & development , Cellulose , Glass , Pseudomonas fluorescens/physiology , Stainless Steel , Microscopy, Atomic Force
3.
Gene Ther ; 21(8): 715-22, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24849041

ABSTRACT

We previously demonstrated that intraperitoneal delivery of adeno-associated virus serotype 8 (AAV8) stably transduces the pancreas, including the ß cells in the pancreatic islets. We further demonstrated the ability to deliver and express target genes specifically in ß cells for at least 6 months using a murine insulin promoter in a double-stranded, self-complementary AAV vector. Recombinant interleukin (IL)-2 has been shown to induce CD4(+)CD25(+) regulatory T cells (Tregs) in several mouse models of autoimmune disease. Here we evaluated the effects of double-stranded adeno-associated virus serotype 8-mouse insulin promoter (dsAAV8-mIP)-mediated delivery of  2 to pancreatic ß cells in non-obese diabetic (NOD) mice. AAV8-mIP-mediated gene expression of IL-2 to pancreatic ß cells of 10-week-old NOD mice prevented the onset of hyperglycemia in NOD mice more in a dose-dependent manner with the lower dose of virus being more effective than a higher dose of AAV-mIP-IL-2 and IL-4. Moreover, the local ß-cell expression of IL-2 increased the number of CD4(+)CD25(+)FoxP3(+) cells in the pancreatic lymph node (PLN) and SPL in both NOD and C57BL/6 mice. Taken together, these results demonstrate that local, low expression of mIL-2 in islets prevents progress of diabetes through the regulation of Tregs.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Type 1/genetics , Forkhead Transcription Factors/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Diabetes Mellitus, Type 1/prevention & control , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Interleukin-2/genetics , Interleukin-4/metabolism , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
4.
Gene Ther ; 21(3): 337-42, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24451117

ABSTRACT

Adoptive immunotherapy of tumors with T cells specific for the cancer-testis antigen NY-ESO-1 has shown great promise in preclinical models and in early stage clinical trials. Tumor persistence or recurrence after NY-ESO-1-specific therapy occurs, however, and the mechanisms of recurrence remain poorly defined. In a murine xenograft model of NY-ESO-1(+) multiple myeloma, we observed tumor recurrence after adoptive transfer of CD8(+) T cells genetically redirected to the prototypic NY-ESO-1157-165 peptide presented by HLA-A*02:01. Analysis of the myeloma cells that had escaped from T-cell control revealed intact expression of NY-ESO-1 and B2M, but selective, complete loss of HLA-A*02:01 expression from the cell surface. Loss of heterozygosity (LOH) in the major histocompatibility complex (MHC) involving the HLA-A locus was identified in the tumor cells, and further analysis revealed selective loss of the allele encoding HLA-A*02:01. Although LOH involving the MHC has not been described in myeloma patients with persistent or recurrent disease after immune therapies such as allogeneic hematopoietic cell transplantation (HCT), it has been described in patients with acute myelogenous leukemia who relapsed after allogeneic HCT. These results suggest that MHC loss should be evaluated in patients with myeloma and other cancers who relapse after adoptive NY-ESO-1-specific T-cell therapy.


Subject(s)
HLA-A2 Antigen/genetics , Heterozygote , Immunotherapy , Multiple Myeloma/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Tumor Escape , Alleles , Animals , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/therapy
5.
Gene Ther ; 20(1): 1-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22241179

ABSTRACT

Gene transfer of key regulators of osteogenesis for mesenchymal stem cells represents a promising strategy to regenerate bone. It has been reported that LMP3, a transcription variant of LIM domain mineralization protein (LMP) lacking LIM domains, can induce osteogenesis in vitro and in vivo. As little is known about the effects of LMP3 gene therapy on periodontal ligament (PDL) cell osteogenic differentiation, this study sought to explore whether gene delivery of LMP3 can promote PDL cell mineralization and bone formation. Our results showed that adenoviral mediated gene transfer of LMP3 (AdLMP3) significantly upregulated ALP (Alkaline Phosphatase), BSP (Bone Sialoprotein) and BMP2 gene expression and increased in vitro matrix mineralization in human PDL. Although AdLMP3 gene delivery to PDL cells did not induce ectopic bone formation in vivo, we found that AdLMP3 augments new bone formation, which co-delivered with AdBMP7 gene transfer. Our study provides the evidence that there is a synergistic effect between LMP3 and BMP-7 in vivo, suggesting that LMP3 delivery may be used to augment BMP-mediated osteogenesis. LMP3 and BMP-7 combinatory gene therapy may also have specific applications for oral and periodontal regenerative medicine.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Periodontal Ligament/physiology , Regeneration/genetics , Adenoviridae/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Bone Matrix/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 7/metabolism , Calcification, Physiologic , Cell Differentiation , Cell Line , Genetic Vectors/genetics , Humans , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Periodontal Ligament/cytology , Transformation, Genetic , Up-Regulation
6.
Gene Ther ; 19(8): 791-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22089495

ABSTRACT

Type-I diabetes is a chronic disease mediated by autoimmune destruction of insulin-producing ß-cells. Although progress has been made towards improving diabetes-associated pathologies and the quality of life for those living with diabetes, no therapy has been effective at eliminating disease manifestations or reversing disease progression. Here, we examined whether double-stranded adeno-associated virus serotype 8 (dsAAV8)-mediated gene delivery to endogenous ß-cells of interleukin (IL)-4 in combination with ß-cell growth factors can reverse early-onset diabetes in NOD mice. Our results demonstrate that a single treatment with dsAAV8 vectors expressing IL-4 in combination with glucagon-like peptide-1 or hepatocyte growth factor/NK1 under the regulation of the insulin promoter enhanced ß-cell proliferation and survival in vivo, significantly delaying diabetes progression in NOD mice, and reversing disease in ∼10% of treated NOD mice. These results demonstrate the ability to reverse hyperglycemia in NOD mice with established diabetes by in vivo gene transfer to ß-cells of immunomodulatory factors and ß-cell growth factors.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Genetic Therapy , Glucagon-Like Peptide 1/genetics , Hepatocyte Growth Factor/genetics , Insulin-Secreting Cells/metabolism , Interleukin-4/genetics , Animals , Diabetes Mellitus, Type 1/genetics , Female , Gene Transfer Techniques , Genetic Vectors , Glucagon-Like Peptide 1/metabolism , Hepatocyte Growth Factor/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred NOD
7.
J Cell Physiol ; 227(2): 416-20, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21948071

ABSTRACT

Orthopedic gene therapy has been the topic of considerable research for two decades. The preclinical data are impressive and many orthopedic conditions are well suited to genetic therapies. But there have been few clinical trials and no FDA-approved product exists. This paper examines why this is so. The reasons are multifactorial. Clinical translation is expensive and difficult to fund by traditional academic routes. Because gene therapy is viewed as unsafe and risky, it does not attract major funding from the pharmaceutical industry. Start-up companies are burdened by the complex intellectual property environment and difficulties in dealing with the technology transfer offices of major universities. Successful translation requires close interactions between scientists, clinicians and experts in regulatory and compliance issues. It is difficult to create such a favorable translational environment. Other promising fields of biological therapy have contemplated similar frustrations approximately 20 years after their founding, so there seem to be more general constraints on translation that are difficult to define. Gene therapy has noted some major clinical successes in recent years, and a sense of optimism is returning to the field. We hope that orthopedic applications will benefit collaterally from this upswing and move expeditiously into advanced clinical trials.


Subject(s)
Genetic Therapy/methods , Musculoskeletal Diseases/genetics , Musculoskeletal Diseases/therapy , Orthopedics/methods , Clinical Trials as Topic , Humans , Musculoskeletal Diseases/pathology , Regenerative Medicine , Tissue Engineering
8.
Nat Cell Biol ; 2(8): 476-83, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10934467

ABSTRACT

Release of cytochrome c from mitochondria by apoptotic signals induces ATP/dATP-dependent formation of the oligomeric Apaf-1-caspase-9 apoptosome. Here we show that the documented anti-apoptotic effect of the principal heat-shock protein, Hsp70, is mediated through its direct association with the caspase-recruitment domain (CARD) of Apaf-1 and through inhibition of apoptosome formation. The interaction between Hsp70 and Apaf-1 prevents oligomerization of Apaf-1 and association of Apaf-1 with procaspase-9. On the basis of these results, we propose that resistance to apoptosis exhibited by stressed cells and some tumours, which constitutively express high levels of Hsp70, may be due in part to modulation of Apaf-1 function by Hsp70.


Subject(s)
Apoptosis , HSP70 Heat-Shock Proteins/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Apoptotic Protease-Activating Factor 1 , Blotting, Western , Caspase 9 , Caspases/chemistry , Caspases/metabolism , Cell Extracts , Cell Line , Cell-Free System , Cytochrome c Group/metabolism , Deoxyadenine Nucleotides/metabolism , Deoxyadenine Nucleotides/pharmacology , Enzyme Activation/drug effects , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Gene Expression , HSP70 Heat-Shock Proteins/genetics , Hot Temperature , Humans , Ligands , Macromolecular Substances , Precipitin Tests , Protein Binding/drug effects , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proteins/chemistry
9.
Int J Immunopathol Pharmacol ; 24(1 Suppl 2): 139-42, 2011.
Article in English | MEDLINE | ID: mdl-21669153

ABSTRACT

The debate is still ongoing about the long term effects of the mininvasive vertebral augmentation techniques and their usefulness in treating more complex cases where a bone inducing effect more than a merely bone substitution would be suitable, such as the vertebral fractures in young patients. We previously developed a clinically relevant gene therapy approach using modified dermal fibroblasts for inducing bone healing and bone formation in different animal models. The aim of this study is to show the feasibility of a minimally invasive percutaneous intrasomatic ex vivo gene therapy approach to treat thoracolumbar vertebral fractures and anterior column bone defects in a goat model.


Subject(s)
Fibroblasts , Genetic Therapy/methods , Skin/cytology , Spinal Fractures/therapy , Adenoviridae , Animals , Cell Separation , Female , Fracture Healing , Genetic Engineering , Genetic Vectors , Goats , Mandibular Injuries/diagnostic imaging , Mandibular Injuries/therapy , Radiography , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spinal Fractures/diagnostic imaging , Transduction, Genetic
11.
J Environ Manage ; 92(3): 457-71, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21074314

ABSTRACT

Lack of sanitation affects the lives of billions of people worldwide. It is now generally agreed that sustainable solutions to this complex problem require social and cultural factors to be addressed in addition to the habitual economic and technical aspects. Increasingly, sector professionals view the fragmented approaches to sanitation as a limiting factor. This refers to the fragmentation of the knowledge on the subject among often hermetic disciplines and to the distribution of political mandates on sanitation across many institutions, which independently tackle specific aspects of the issue. Holistic approaches have often been suggested as a solution. This paper presents the development of such a holistic approach, designed to assess sanitation development in rural and peri-urban settings. Tested in three Mexican communities, it relies on qualitative research tools to identify critical influences to sanitation development. This article presents generic results about micro and macro-factors affecting sanitation development in Mexican villages, and reflexively examines the research process as well as the strengths and limitations of the approach. The conceptual map developed for each case study successfully highlights the interconnectedness of all factors affecting sanitation development. Despite some weaknesses, these maps constitute a practical assessment tool for interdisciplinary teams deployed in integrated water and sanitation development programs and a valuable didactic tool for training activities.


Subject(s)
Rural Health , Sanitary Engineering , Urbanization , Mexico
12.
ESMO Open ; 6(3): 100101, 2021 06.
Article in English | MEDLINE | ID: mdl-33901870

ABSTRACT

BACKGROUND: Among patients with advanced renal cell carcinoma (RCC), those with sarcomatoid histology (sRCC) have the poorest prognosis. This analysis assessed the efficacy of avelumab plus axitinib versus sunitinib in patients with treatment-naive advanced sRCC. METHODS: The randomized, open-label, multicenter, phase III JAVELIN Renal 101 trial (NCT02684006) enrolled patients with treatment-naive advanced RCC. Patients were randomized 1 : 1 to receive either avelumab plus axitinib or sunitinib following standard doses and schedules. Assessments in this post hoc analysis of patients with sRCC included efficacy (including progression-free survival) and biomarker analyses. RESULTS: A total of 108 patients had sarcomatoid histology and were included in this post hoc analysis; 47 patients in the avelumab plus axitinib arm and 61 in the sunitinib arm. Patients in the avelumab plus axitinib arm had improved progression-free survival [stratified hazard ratio, 0.57 (95% confidence interval, 0.325-1.003)] and a higher objective response rate (46.8% versus 21.3%; complete response in 4.3% versus 0%) versus those in the sunitinib arm. Correlative gene expression analyses of patients with sRCC showed enrichment of gene pathway scores for cancer-associated fibroblasts and regulatory T cells, CD274 and CD8A expression, and tumors with The Cancer Genome Atlas m3 classification. CONCLUSIONS: In this subgroup analysis of JAVELIN Renal 101, patients with sRCC in the avelumab plus axitinib arm had improved efficacy outcomes versus those in the sunitinib arm. Correlative analyses provide insight into this subtype of RCC and suggest that avelumab plus axitinib may increase the chance of overcoming the aggressive features of sRCC.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Axitinib , Carcinoma, Renal Cell , Kidney Neoplasms , Sunitinib , Antineoplastic Combined Chemotherapy Protocols , Axitinib/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Sunitinib/therapeutic use
13.
Gene Ther ; 17(2): 171-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19865180

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an incretin hormone that performs a wide array of well-characterized antidiabetic actions, including stimulation of glucose-dependent insulin secretion, upregulation of insulin gene expression and improvements in beta-cell survival. GLP-1-receptor agonists have been developed for treatment of diabetes; however, the short biological half-lives of these peptide-based therapeutics requires that frequent injections be administered to maintain sufficient circulating levels. Thus, novel methods of delivering GLP-1 remain an important avenue of active research. It has recently been demonstrated that self-complimentary, double-stranded, adeno-associated virus serotype-8 (DsAAV8) can efficiently transduce pancreatic beta-cells in vivo, resulting in long-term transgene expression. In this study, we engineered a DsAAV8 vector containing a GLP-1 transgene driven by the mouse insulin-II promoter (MIP). Biological activity of the GLP-1 produced from this transgene was assessed using a luciferase-based bioassay. DsAAV8-MIP-GLP-1 was delivered via intraperitoneal injection and beta-cell damage induced by multiple low dose streptozotocin (STZ) administration. Glucose tolerance was assessed following intraperitoneal glucose injections and beta-cell proliferation measured by PCNA expression. Expression of GLP-1 in Min6 beta-cells resulted in glucose-dependent secretion of biologically active GLP-1. Intraperitoneal delivery of DsAAV8-MIP-GLP-1 to mice led to localized GLP-1 expression in beta-cells and protection against development of diabetes induced by multiple low-dose STZ administration. This protection was associated with significant increase in beta-cell proliferation. Results from this study indicate that expression and secretion of GLP-1 from beta-cells in vivo via DsAAV8 represents a novel therapeutic strategy for treatment of diabetes.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/prevention & control , Genetic Therapy/methods , Genetic Vectors , Glucagon-Like Peptide 1/genetics , Insulin-Secreting Cells/metabolism , Animals , Cell Proliferation , Glucagon-Like Peptide 1/metabolism , Injections, Intraperitoneal , Insulin/genetics , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic
14.
Gene Ther ; 17(12): 1476-83, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20720575

ABSTRACT

The IκB kinase (IKKα, ß and the regulatory subunit IKKγ) complex regulates nuclear factor of κB (NF-κB) transcriptional activity, which is upregulated in many chronic inflammatory diseases. NF-κB signaling promotes inflammation and limits muscle regeneration in Duchenne muscular dystrophy (DMD), resulting in fibrotic and fatty tissue replacement of muscle that exacerbates the wasting process in dystrophic muscles. Here, we examined whether dominant-negative forms of IKKα (IKKα-dn) and IKKß (IKKß-dn) delivered by adeno-associated viral (AAV) vectors to the gastrocnemius (GAS) and tibialis anterior (TA) muscles of 1, 2 and 11-month-old mdx mice, a murine DMD model, block NF-κB activation and increase muscle regeneration. At 1 month post-treatment, the levels of nuclear NF-κB in locally treated muscle were decreased by gene transfer with either AAV-CMV-IKKα-dn or AAV-CMV-IKKß-dn, but not by IKK wild-type controls (IKKα and ß) or phosphate-buffered saline (PBS). Although treatment with AAV-IKKα-dn or AAV-IKKß-dn vectors had no significant effect on muscle regeneration in young mdx mice treated at 1 and 2 months of age and collected 1 month later, treatment of old (11 months) mdx with AAV-CMV-IKKα-dn or AAV-CMV-IKKß-dn significantly increased levels of muscle regeneration. In addition, there was a significant decrease in myofiber necrosis in the AAV-IKKα-dn- and AAV-IKKß-dn-treated mdx muscle in both young and old mice. These results demonstrate that inhibition of IKKα or IKKß in dystrophic muscle reduces the adverse effects of NF-κB signaling, resulting in a therapeutic effect. Moreover, these results clearly demonstrate the therapeutic benefits of inhibiting NF-κB activation by AAV gene transfer in dystrophic muscle to promote regeneration, particularly in older mdx mice, and block necrosis.


Subject(s)
Dependovirus/genetics , Genetic Therapy , I-kappa B Kinase , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne , NF-kappa B , Animals , Cell Nucleus/enzymology , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/enzymology , Muscular Dystrophy, Duchenne/therapy , NF-kappa B/genetics , NF-kappa B/metabolism , Regeneration/physiology , Signal Transduction/genetics
15.
J Exp Med ; 173(1): 221-30, 1991 Jan 01.
Article in English | MEDLINE | ID: mdl-1898661

ABSTRACT

An HLA-A2+ cytotoxic T lymphocyte (CTL) line restricted by HLA-A2 in recognition of an influenza B virus nucleoprotein (BNP) peptide uses the CD8 coreceptor in the recognition of this viral peptide. Incubation of these CTL with BNP peptide in the absence of antigen-presenting cells downmodulates CD8 alpha and CD8 beta expression and reduces their ability to lyse target cells without inducing self-lysis. CD8 downmodulation was dependent on peptide concentration, time of exposure, and T cell receptor specificity. Another viral peptide from the influenza A virus matrix protein interacting with HLA-A2 had no effect on CD8 expression. Upon further investigation, an anti-HLA class I monoclonal antibody (mAb), anti-HLA class II mAb, and HLA alloantisera were found to downmodulate CD8 alpha and CD8 beta expression and induce CTL nonresponsiveness without causing degranulation. When CD8 alpha and CD8 beta expression was modulated by viral peptide or anti-HLA mAbs, other cell surface molecules were unchanged. Finally, incubation of peripheral blood lymphocytes with these anti-HLA mAbs induced no change in CD8 expression on resting cells but did downmodulate it on mitogen-activated cells. These results suggest that T cell recognition of the HLA-A2-BNP peptide complex on neighboring CTL may be the mechanism for CD8 downmodulation induced by the BNP viral peptide. This mechanism may be important in clonal anergy.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , HLA-A2 Antigen/physiology , T-Lymphocytes, Cytotoxic/immunology , Antibodies, Monoclonal , CD8 Antigens , Cell Line , Down-Regulation/immunology , Gene Expression Regulation , HLA-A2 Antigen/immunology , Humans , Receptors, Antigen, T-Cell/immunology , Time Factors , Viral Proteins/immunology
16.
J Exp Med ; 160(4): 1114-25, 1984 Oct 01.
Article in English | MEDLINE | ID: mdl-6481302

ABSTRACT

We have characterized the major glycolipid constituents of the mouse peritoneal macrophage, and have demonstrated that alterations in the amount and in the accessibility of specific glycolipid species to galactose oxidase/NaB3H4 labeling, an indicator of glycolipid surface exposure, occur in response to inflammation and as a consequence of activation to a tumoricidal state. The key findings are: (a) Asialo GM1, a major neutral glycolipid constituent of all macrophage populations examined, is accessible to galactose oxidase/NaB3H4 labeling on the surface of TG-elicited and BCG-activated macrophages but not on resident macrophages; (b) GM1 is the predominant ganglioside constituent of the mouse macrophage. Resident macrophages contain two distinct GM1 species, as determined by cholera toxin binding, while TG-elicited and BCG-activated macrophages contain an additional GM1 species. Differences in the relative amounts of these GM1 species, as well as in their accessibility to galactose oxidase/NaB3H4 labeling, exist among the macrophage populations. These observations suggest that both a chemical and spatial reorganization of surface glycolipids occurs in response to inflammation and tumoricidal activation.


Subject(s)
Antigens, CD , Glycolipids/metabolism , Inflammation/immunology , Lactosylceramides , Macrophage Activation , Macrophages/metabolism , Membrane Lipids/metabolism , Animals , Ascitic Fluid/immunology , Female , G(M1) Ganglioside/isolation & purification , Galactosylceramides/isolation & purification , Glycolipids/isolation & purification , Glycosphingolipids/isolation & purification , Leukemia, Experimental/immunology , Macrophages/classification , Macrophages/immunology , Mice , Mice, Inbred C57BL
17.
J Exp Med ; 181(2): 799-804, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7836932

ABSTRACT

The availability of antitumor cytotoxic T lymphocytes which can be generated from either peripheral blood lymphocytes after stimulation in vitro or tumor infiltrating lymphocytes (TIL) has made it possible to identify a number of melanoma antigens presented by major histocompatibility complex class I molecules. The present and previous studies indicated that TIL586 recognized an antigen expressed on most melanoma and normal melanocytes in the context of the HLA-A31 molecule. We report here the cloning of a cDNA that directs the expression of the shared melanoma antigen recognized by this TIL. The DNA sequence analysis revealed that the cDNA was almost identical to the gene encoding tyrosinase-related protein 1 or glycoprotein gp75 which was originally identified by serum antibodies in a patient with melanoma. The gene was found to be expressed only in melanoma, normal melanocyte cell lines, and retina, but not in other normal tissues tested. The gp75 antigen presented by HLA-A31 may therefore constitute a useful immune target for specific treatment of patients with melanoma, since both antibody- and T cell-mediated immune responses can be generated against this antigen.


Subject(s)
Antigens, Neoplasm/genetics , HLA-A Antigens/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Antigens, Neoplasm/immunology , Cell Line , Cloning, Molecular , DNA, Complementary , Humans , Melanoma/immunology , Tumor Cells, Cultured
18.
J Exp Med ; 182(2): 477-86, 1995 Aug 01.
Article in English | MEDLINE | ID: mdl-7629507

ABSTRACT

After the cloning of murine cytokine synthesis inhibitory factor, it was recognized that a homologous open reading frame was encoded within the Epstein-Barr virus (human herpes virus 4). This viral protein has now been termed viral interleukin 10 (vIL-10) to reflect its protein sequence homology to "cellular" IL-10 (cIL-10, either murine or human IL-10). It is now widely accepted that vIL-10 shares many functions with cIL-10, principally, the ability to enhance survival of newly infected B cells and to diminish the production of IFN-gamma and IL-2 during ongoing immune reactions. The immunomodulatory effect of locally secreted vIL-10 and murine IL-10 (mIL-10) was examined in tumor models using CL8-1 (a BL6 melanoma cell line transfected with the H-2Kb class I gene) in syngeneic animals. Although parental BL6 tumor cells grow in immunocompetent syngeneic hosts, CL8-1 are rejected. To achieve local secretion of vIL-10, we generated vIL-10 retroviral vectors. While nontransduced CL8-1 cells (1 x 10(4)) failed to grow when injected intradermally in C57BL/6 mice, CL8-1 cells (1 x 10(4)) transduced with vIL-10 formed palpable tumors and eventually killed 80% of injected animals. Suppression of tumor rejection was also noted when CL8-1 tumors with or without vIL-10 transfection were admixed with syngeneic vIL-10-transfected fibroblasts and inoculated. Since the in vitro proliferation of the tumor was not altered after transduction with the vIL-10 gene and injection of vIL-10-transduced CL8-1 does not affect the rejection of nontransduced CL8-1 inoculated at a distant site, local vIL-10 secretion appears to suppress the process of immune rejection of the target cells in a dose-dependent manner. Similar results were observed for the H-2b MCA105 sarcoma tumor model in allogeneic BALB/c mice (H-2d). Although all animals that received nontransfected MCA105 rapidly rejected these tumors, MCA105 sarcomas transfected with vIL-10 remained palpable for up to 37 d. The local immunosuppressive effect of gene-delivered vIL-10 could be neutralized by anti-human IL-10 monoclonal antibody or could be reversed by the systemic administration of IL-2 or IL-12. In marked contrast, mIL-10 transfection of CL8-1 significantly suppressed tumor growth and frequently led to the rejection of tumor. Similar results were obtained for the murine tumor cell lines MCA102.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Herpesvirus 4, Human/immunology , Interleukin-10 , Neoplasms, Experimental/immunology , Viral Proteins/immunology , Animals , Female , Graft Rejection , Immune Tolerance , Lymphocyte Subsets/immunology , Lymphocytes, Tumor-Infiltrating , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Recombinant Proteins , Species Specificity , Transfection
19.
J Exp Med ; 183(3): 1131-40, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8642255

ABSTRACT

Tumor infiltrating lymphocytes (TILs) derived from tumor-bearing patients recognize tumor-associated antigens presented by major histocompatibility complex (MHC) class I molecules. The infusion of TIL586 along with interleukin (IL) 2 into an autologous patient with metastatic melanoma resulted in the objective regression of tumor. A gene encoding a tumor antigen recognized by TIL586 was recently isolated and shown to encode gp75. Here we report that an antigenic peptide, MSLQRQFLR, recognized by TIL586 was not derived from the normal gp75 protein. Instead, this nonamer peptide resulted from translation of an alternative open reading frame of the same gene. Thus, the gp75 gene encodes two completely different polypeptides, gp75 as an antigen recognized by immunoglobulin G antibodies in sera from a patient with cancer, and a 24-amino acid product as a tumor rejection antigen recognized by T cells. This represents the first demonstration that a human tumor rejection antigen can be generated from a normal cellular gene using an open reading frame other than that used to encode the normal protein. These findings revealed a novel mechanism for generating tumor antigens, which may be useful as vaccines to induce tumor-specific cell-mediated immunity against cancer.


Subject(s)
Alternative Splicing , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Histocompatibility Antigens Class I/immunology , Interleukin-2/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/therapy , Membrane Glycoproteins , Open Reading Frames , Oxidoreductases , Protein Biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Cytotoxicity, Immunologic , DNA Primers , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Melanoma/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Polymerase Chain Reaction , Proteins/genetics , Recombinant Proteins/biosynthesis , Transfection , Tumor Cells, Cultured
20.
J Exp Med ; 180(1): 347-52, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-7516411

ABSTRACT

Four melanoma proteins, MART-1, gp100, tyrosinase, and tyrosinase-related protein-1 (gp75) were evaluated for recognition by HLA-A2-restricted melanoma-specific cytotoxic T lymphocytes (CTLs) derived from the tumor-infiltrating lymphocytes (TIL) of 10 different patients. 9 of 10 TIL recognized MART-1, 4 recognized gp100 (including 3 that also recognized MART-1), but none of the TIL recognized tyrosinase or gp75. Based on the known HLA-A2.1 peptide binding motifs, 23 peptides from MART-1 were synthesized in an attempt to identify the epitopes recognized by TIL. Three peptides were recognized by TIL when pulsed on T2 target cells. One of the 9-mer peptides, AAGIGILTV, was most effective in sensitizing the T2 cells for TIL lysis. This peptide was recognized by 9 of 10 HLA-A2-restricted melanoma-specific CTLs. Therefore, this peptide appears to be a very common immunogenic epitope for HLA-A2-restricted melanoma-specific TIL and may be useful for the development of immunotherapeutic strategies.


Subject(s)
Antigens, Neoplasm/analysis , HLA-A2 Antigen/physiology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Neoplasm Proteins/analysis , Amino Acid Sequence , Epitopes/analysis , Humans , Melanoma-Specific Antigens , Molecular Sequence Data , Neoplasm Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL