Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Biol Rep ; 47(7): 5145-5154, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32562174

ABSTRACT

Proper bony tissue regeneration requires mechanical stabilization, an osteogenic biological activity and appropriate scaffolds. The latter two elements can be combined in a hydrogel format for effective delivery, so it can readily adapt to the architecture of the defect. We evaluated a Good Manufacturing Practice-compliant formulation composed of bone marrow-derived mesenchymal stromal cells in combination with bone particles (Ø = 0.25 to 1 µm) and fibrin, which can be readily translated into the clinical setting for the treatment of bone defects, as an alternative to bone tissue autografts. Remarkably, cells survived with unaltered phenotype (CD73+, CD90+, CD105+, CD31-, CD45-) and retained their osteogenic capacity up to 48 h after being combined with hydrogel and bone particles, thus demonstrating the stability of their identity and potency. Moreover, in a subchronic toxicity in vivo study, no toxicity was observed upon subcutaneous administration in athymic mice and signs of osteogenesis and vascularization were detected 2 months after administration. The preclinical data gathered in the present work, in compliance with current quality and regulatory requirements, demonstrated the feasibility of formulating an osteogenic cell-based tissue engineering product with a defined profile including identity, purity and potency (in vitro and in vivo), and the stability of these attributes, which complements the preclinical package required prior to move towards its use of prior to its clinical use.


Subject(s)
Hydrogels/standards , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Tissue Scaffolds/standards , Animals , Bone Transplantation/methods , Bone Transplantation/standards , Cells, Cultured , Clinical Trials as Topic , Female , Humans , Hydrogels/adverse effects , Mice , Neovascularization, Physiologic , Osteoclasts/cytology , Tissue Engineering/standards , Tissue Scaffolds/adverse effects
2.
Front Immunol ; 11: 271, 2020.
Article in English | MEDLINE | ID: mdl-32161589

ABSTRACT

Immunosuppressed patients are susceptible to virus reactivation or de novo infection. Adoptive immunotherapy, based on virus-specific T lymphocytes (VST), can prevent or treat viral diseases. However, donor availability, HLA-compatibility restrictions, high costs, and time required for the production of personalized medicines constitute considerable limitations to this treatment. Ex vivo rapid and large-scale expansion of VST, compliant with current good manufacturing practice (cGMP) standards, with an associated cell donor registry would overcome these limitations. This study aimed to characterize a VST product obtained through an expansion protocol transferable to cGMP standards. Antigenic stimulus consisted of cytomegalovirus (CMV) pp65 peptide pool-pulsed autologous dendritic cells (DCs) derived from monocytes. G-Rex technology, cytokines IL-2, IL-7, and IL-15, and anti-CD3 and anti-CD28 antibodies were used for culture. At day 14 of cell culture, the final product was characterized regarding T cell subsets, specificity, and functionality. The final product, comprised mainly CD4+ and CD8+ T lymphocytes (49.2 ± 24.7 and 42.3 ± 25.2, respectively). The culture conditions made it possible to achieve at least a 98.89-fold increase in pp65-specific CD3+ IFN-γ+ cells. These cells were specific, as pp65-specific cytotoxicity was demonstrated. Additionally, in complete HLA mismatch and without the presence of pp65, alloreactivity resulted in <5% cell lysis. In conclusion, a cGMP scalable process for the generation of a large number of doses of CMV-specific cytotoxic T cells was successfully performed.


Subject(s)
Cytomegalovirus/metabolism , Immunotherapy, Adoptive/methods , T-Lymphocytes, Cytotoxic/immunology , Antigens, Differentiation, T-Lymphocyte , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Culture Techniques , Cytokines/metabolism , Cytomegalovirus Infections , Dendritic Cells , Healthy Volunteers , Humans , Interferon-gamma , Leukocytes, Mononuclear , Monocytes , Viral Matrix Proteins
3.
Cytotherapy ; 21(1): 32-40, 2019 01.
Article in English | MEDLINE | ID: mdl-30447901

ABSTRACT

BACKGROUND AIMS: Multipotent mesenchymal stromal cell (MSC)-based medicines are extensively investigated for use in regenerative medicine and immunotherapy applications. The International Society for Cell and Gene Therapy (ISCT) proposed a panel of cell surface molecules for MSC identification that includes human leukocyte antigen (HLA)-DR as a negative marker. However, its expression is largely unpredictable despite production under tightly controlled conditions and compliance with current Good Manufacturing Practices. Herein, we report the frequency of HLA-DR expression in 81 batches of clinical grade bone marrow (BM)-derived MSCs and investigated its impact on cell attributes and culture environment. METHODS: The levels of 15 cytokines (interleukin [IL]-1ß, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-21, IL-22, IL-23, IL-25, IL-31, IL-33, interferon-γ, soluble CD40 ligand and tumor necrosis factor-α) were determined in sera supplements and supernatants of BM-MSC cultures. Identity, multipotentiality and immunopotency assays were performed on high (>20% of cells) and low (≤20% of cells) HLA-DR+ cultures. RESULTS: A correlation was found between HLA-DR expression and levels of IL-17F and IL-33. Expression of HLA-DR did neither affect MSC identity, in vitro tri-lineage differentiation potential (into osteogenic, chondrogenic and adipogenic lineages), nor their ability to inhibit the proliferation of stimulated lymphocytes. DISCUSSION: Out of 81 batches of BM-MSCs for autologous use analyzed, only three batches would have passed the ISCT criteria (<2%), whereas 60.5% of batches were compliant with low HLA-DR values (≤20%). Although a cause-effect relationship cannot be drawn, we have provided a better understanding of signaling events and cellular responses in expansion culture conditions relating with HLA-DR expression.


Subject(s)
HLA-DR Antigens/immunology , Interleukin-17/blood , Interleukin-33/blood , Mesenchymal Stem Cells/immunology , Primary Cell Culture/methods , Adipogenesis , Biomarkers/metabolism , Bone Marrow/immunology , Cell Differentiation/physiology , Cells, Cultured , Chondrogenesis , Humans , Lymphocyte Activation , Mesenchymal Stem Cell Transplantation , Osteogenesis
4.
Curr Protoc Stem Cell Biol ; 44: 2B.9.1-2B.9.22, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29512111

ABSTRACT

In the field of orthopedics, translational research of novel therapeutic approaches involves the use of large animal models (such as sheep, goat, pig, dog, and horse) due to the similarities with humans in weight, size, joint structure, and bone/cartilage healing mechanisms. Particularly in the development of cell-based therapies, the lack of manageable immunocompromised preclinical large animal models prevents the use of human cells, which makes it necessary to produce equivalent homologous cell types for the study of their pharmacodynamics, pharmacokinetics, and toxicology. The methods described herein allow for the isolation, expansion, manipulation, and characterization of fibroblastic-like ovine bone marrow-derived multipotent mesenchymal stromal cells (BM-MSC) that, similar to human BM-MSC, adhere to standard plastic surfaces; express specific surface markers such as CD44, CD90, CD140a, CD105, and CD166; and display trilineage differentiation potential in vitro. Homogeneous cell cultures result from a 3-week bioprocess yielding cell densities in the range of 2-4 × 104 MSC/cm2 at passage 2, which corresponds to ∼8 cumulative population doublings. Large quantities of BM-MSC resulting from following this methodology can be readily used in proof of efficacy and safety studies in the preclinical development stage. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Bone Marrow Cells/cytology , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Mesenchymal Stem Cells/enzymology , Phenotype , Sheep , Stromal Cells/cytology
5.
Cytotechnology ; 70(1): 31-44, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29322348

ABSTRACT

Clinical use of multipotent Mesenchymal Stromal Cell (MSC)-based medicinal products requires their production in compliance with Good Manufacturing Practices, thus ensuring that the final drug product meets specifications consistently from batch to batch in terms of cell viability, identity, purity and potency. Potency relates to the efficacy of the medicine in its target clinical indication, so adequate release tests need to be defined and validated as quality controls. Herein we report the design and optimisation of parameters affecting the performance of an in vitro cell-based assay for assessing immunomodulatory potential of clinical grade MSC for human use, based on their capacity to inhibit proliferation of T lymphocytes under strong polyclonal stimuli. The resulting method was demonstrated to be reproducible and relatively simple to execute. Two case studies using clinical grade MSC are presented as examples to illustrate the applicability of the methodology described in this work.

6.
J Tissue Eng Regen Med ; 12(1): e532-e540, 2018 01.
Article in English | MEDLINE | ID: mdl-27684058

ABSTRACT

Pseudoarthrosis is a relatively frequent complication of fractures, in which the lack of mechanical stability and biological stimuli results in the failure of bone union, most frequently in humerus and tibia. Treatment of recalcitrant pseudoarthrosis relies on the achievement of satisfactory mechanical stability combined with adequate local biology. Herein we present two cases of atrophic pseudoarthrosis that received a tissue-engineering product (TEP) composed of autologous bone marrow-derived mesenchymal stromal cells (BM-MSC) combined with deantigenized trabecular bone particles from a tissue bank. The feasibility of the treatment and osteogenic potential of the cell-based medicine was first demonstrated in an ovine model of critical size segmental tibial defect. Clinical-grade autologous BM-MSC were produced following a good manufacturing practice-compliant bioprocess. Results were successful in one case, with pseudoarthrosis resolution, and inconclusive in the other one. The first patient presented atrophic pseudoarthrosis of the humeral diaphysis and was treated with osteosynthesis and TEP resulting in satisfactory consolidation at month 6. The second case presented a recalcitrant pseudoarthrosis of the proximal tibia and the Masquelet technique was followed before filling the defect with the TEP. This patient presented a neuropathic pain syndrome unrelated to the treatment that forced the amputation of the extremity 3 months later. In this case, the histological analysis of the tissue formed at the defect site provided evidence of neovascularization but no overt bone remodelling activity. It is concluded that the use of expanded autologous BM-MSC to treat pseudoarthrosis was demonstrated to be feasible and safe, provided that no clinical complications were reported, and early signs of effectiveness were observed. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Pseudarthrosis/pathology , Pseudarthrosis/therapy , Translational Research, Biomedical , Adult , Animals , Atrophy , Bone Marrow Cells/cytology , Disease Models, Animal , Female , Humans , Male , Middle Aged , Osteogenesis , Sheep , Tibia/pathology , Tibia/surgery , Tissue Engineering
7.
Cytotherapy ; 19(9): 1060-1069, 2017 09.
Article in English | MEDLINE | ID: mdl-28734679

ABSTRACT

BACKGROUND AIMS: Biodistribution of candidate cell-based therapeutics is a critical safety concern that must be addressed in the preclinical development program. We aimed to design a decision tree based on a series of studies included in actual dossiers approved by competent regulatory authorities, noting that the design, execution and interpretation of pharmacokinetics studies using this type of therapy is not straightforward and presents a challenge for both developers and regulators. METHODS: Eight studies were evaluated for the definition of a decision tree, in which mesenchymal stromal cells (MSCs) were administered to mouse, rat and sheep models using diverse routes (local or systemic), cell labeling (chemical or genetic) and detection methodologies (polymerase chain reaction [PCR], immunohistochemistry [IHC], fluorescence bioimaging, and magnetic resonance imaging [MRI]). Moreover, labeling and detection methodologies were compared in terms of cost, throughput, speed, sensitivity and specificity. RESULTS: A decision tree was defined based on the model chosen: (i) small immunodeficient animals receiving heterologous MSC products for assessing biodistribution and other safety aspects and (ii) large animals receiving homologous labeled products; this contributed to gathering data not only on biodistribution but also on pharmacodynamics. PCR emerged as the most convenient technique despite the loss of spatial information on cell distribution that can be further assessed by IHC. DISCUSSION: This work contributes to the standardization in the design of biodistribution studies by improving methods for accurate assessment of safety. The evaluation of different animal models and screening of target organs through a combination of techniques is a cost-effective and timely strategy.


Subject(s)
Algorithms , Decision Support Techniques , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Animals , Humans , Immunohistochemistry/methods , Magnetic Resonance Imaging , Mesenchymal Stem Cells/physiology , Mice , Polymerase Chain Reaction/methods , Rats , Research Design , Sheep
8.
N Biotechnol ; 35: 19-29, 2017 Mar 25.
Article in English | MEDLINE | ID: mdl-27810336

ABSTRACT

Umbilical cord blood (UCB) transplantation is associated with long periods of aplastic anaemia. This undesirable situation is due to the low cell dose available per unit of UCB and the immaturity of its progenitors. To overcome this, we present a cell culture strategy aimed at the expansion of the CD34+ population and the generation of granulocyte lineage-committed progenitors. Two culture products were produced after either 6 or 14days of in vitro expansion, and their characteristics compared to non-expanded UCB CD34+ controls in terms of phenotype, colony-forming activity and multilineage repopulation potential in NOD-scid IL2Rγnull mice. Both expanded cell products maintained rapid SCID repopulation activity similar to the non-expanded control, but 14-day cultured cells showed impaired long term SCID repopulation activity. The process was successfully scaled up to clinically relevant doses of 89×106 CD34+ cells committed to the granulocytic lineage and 3.9×109 neutrophil precursors in different maturation stages. Cell yields and biological properties presented by the cell product obtained after 14days in culture were superior and therefore this is proposed as the preferred production setup in a new type of dual transplant strategy to reduce aplastic periods, producing a transient repopulation before the definitive engraftment of the non-cultured UCB unit. Importantly, human telomerase reverse transcriptase activity was undetectable, c-myc expression levels were low and no genetic abnormalities were found, as determined by G-banding karyotype, further confirming the safety of the expanded product.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Fetal Blood/cytology , Anemia, Aplastic/blood , Anemia, Aplastic/etiology , Anemia, Aplastic/prevention & control , Animals , Antigens, CD34/blood , Biotechnology , Cell Differentiation , Cell Lineage , Colony-Forming Units Assay , Cord Blood Stem Cell Transplantation/adverse effects , Female , Fetal Blood/immunology , Graft Enhancement, Immunologic/methods , Granulocytes/cytology , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neutrophils/cytology
9.
Regen Med ; 11(6): 521-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27513321

ABSTRACT

AIM: Computerized systems (CS) are essential in the development and manufacture of cell-based medicines and must comply with good manufacturing practice, thus pushing academic developers to implement methods that are typically found within pharmaceutical industry environments. MATERIALS & METHODS: Qualitative and quantitative risk analyses were performed by Ishikawa and Failure Mode and Effects Analysis, respectively. RESULTS: A process for qualification of a CS that keeps track of environmental conditions was designed and executed. The simplicity of the Ishikawa analysis permitted to identify critical parameters that were subsequently quantified by Failure Mode Effects Analysis, resulting in a list of test included in the qualification protocols. CONCLUSION: The approach presented here contributes to simplify and streamline the qualification of CS in compliance with pharmaceutical quality standards.


Subject(s)
Cell- and Tissue-Based Therapy/standards , Computers , Drug Industry/standards , Regenerative Medicine/standards , Total Quality Management , Commerce , Humans
10.
Cytotherapy ; 18(9): 1197-208, 2016 09.
Article in English | MEDLINE | ID: mdl-27424149

ABSTRACT

BACKGROUND: Multipotent mesenchymal stromal cells (MSC) have achieved a notable prominence in the field of regenerative medicine, despite the lack of common standards in the production processes and suitable quality controls compatible with Good Manufacturing Practice (GMP). Herein we describe the design of a bioprocess for bone marrow (BM)-derived MSC isolation and expansion, its validation and production of 48 consecutive batches for clinical use. METHODS: BM samples were collected from the iliac crest of patients for autologous therapy. Manufacturing procedures included: (i) isolation of nucleated cells (NC) by automated density-gradient centrifugation and plating; (ii) trypsinization and expansion of secondary cultures; and (iii) harvest and formulation of a suspension containing 40 ± 10 × 10(6) viable cells. Quality controls were defined as: (i) cell count and viability assessment; (ii) immunophenotype; and (iii) sterility tests, Mycoplasma detection, endotoxin test and Gram staining. RESULTS: A 3-week manufacturing bioprocess was first designed and then validated in 3 consecutive mock productions, prior to producing 48 batches of BM-MSC for clinical use. Validation included the assessment of MSC identity and genetic stability. Regarding production, 139.0 ± 17.8 mL of BM containing 2.53 ± 0.92 × 10(9) viable NC were used as starting material, yielding 38.8 ± 5.3 × 10(6) viable cells in the final product. Surface antigen expression was consistent with the expected phenotype for MSC, displaying high levels of CD73, CD90 and CD105, lack of expression of CD31 and CD45 and low levels of HLA-DR. Tests for sterility, Mycoplasma, Gram staining and endotoxin had negative results in all cases. DISCUSSION: Herein we demonstrated the establishment of a feasible, consistent and reproducible bioprocess for the production of safe BM-derived MSC for clinical use.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Animals , Cell Culture Techniques/standards , Female , Humans , Immunophenotyping , Mesenchymal Stem Cells/immunology , Mice, Inbred NOD , Quality Control
12.
Cytotherapy ; 18(1): 25-35, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26549383

ABSTRACT

BACKGROUND AIMS: Umbilical cord (UC) has been proposed as a source of mesenchymal stromal cells (MSCs) for use in experimental cell-based therapies provided that its collection does not raise any risk to the donor, and, similar to bone marrow and lipoaspirates, UC-MSCs are multipotent cells with immuno-modulative properties. However, some of the challenges that make a broader use of UC-MSCs difficult include the limited availability of fresh starting tissue, time-consuming processing for successful derivation of cell lines, and the lack of information on identity, potency and genetic stability in extensively expanded UC-MSCs, which are necessary for banking relevant cell numbers for preclinical and clinical studies. METHODS: Factors affecting the success of the derivation process (namely, time elapsed from birth to processing and weight of fragments), and methods for establishing a two-tiered system of Master Cell Bank and Working Cell Bank of UC-MSCs were analyzed. RESULTS: Efficient derivation of UC-MSCs was achieved by using UC fragments larger than 7 g that were processed within 80 h from birth. Cells maintained their immunophenotype (being highly positive for CD105, CD90 and CD73 markers), multi-potentiality and immuno-modulative properties beyond 40 cumulative population doublings. No genetic abnormalities were found, as determined by G-banding karyotype, human telomerase reverse transcriptase activity was undetectable and no toxicity was observed in vivo after intravenous administration of UC-MSCs in athymic rats. DISCUSSION: This works demonstrates the feasibility of the derivation and large-scale expansion of UC-MSCs from small and relatively old fragments of UC typically discarded from public cord blood banking programs.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Tissue Banks , Wharton Jelly/cytology , Animals , Cell Proliferation , Cells, Cultured , Humans , Immunophenotyping , Male , Mesenchymal Stem Cells/metabolism , Rats, Nude , Telomerase/metabolism , Tissue Distribution , Umbilical Cord/cytology
13.
Cytotherapy ; 17(8): 1009-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25769789

ABSTRACT

Along with academic and charitable organizations, transfusion centers have ventured into the stem cell field, with the aim of testing of novel cell-based therapeutics in a clinical setting for future marketing approval. The fact that quality management structures, which are required for compliance with good scientific practice regulations, were originally designed for product development in corporate environments represents a major challenge for many developers. In this Commentary, challenges that non-pharmaceutical institutions must overcome to translate cell-based products into clinical therapies will be discussed from a quality standpoint. Furthermore, our development experience for a mesenchymal stromal cell-based therapy will be shared as a case study.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Humans , Mesenchymal Stem Cells/cytology , Quality Control
14.
J Immunol ; 187(11): 5577-86, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22068234

ABSTRACT

CD84 is a self-binding receptor from the CD150 (or signaling lymphocyte activation molecule [SLAM]) family that is broadly expressed in hematopoietic cells. It has been described that the adaptors SLAM-associated protein (SAP) and EWS-FLI1-activated transcript 2 (EAT-2) are critical for CD150 family members' signaling and function. We observed that human mast cells express CD84 but lack SAP or EAT-2, that CD84 is tyrosine phosphorylated upon FcεRI engagement, and that the release of granule contents is reduced when FcεRI is coengaged with CD84 in LAD2 and human CD34(+)-derived mast cells. In addition, we observed that the release of IL-8 and GM-CSF was also reduced in FcεRI/CD84-costimulated cells as compared with FcεRI/Ig control. To understand how CD84 downregulates FcεRI-mediated function, we analyzed signaling pathways affected by CD84 in human mast cells. Our results showed that CD84 dampens FcεRI-mediated calcium mobilization after its co-cross-linking with the receptor. Furthermore, FcεRI-mediated Syk-linker for activation of T cells-phospholipase C-γ1 axis activity is downregulated after CD84 stimulation, compared with FcεRI/Ig control. The inhibitory kinase Fes phosphorylates mainly the inhibitory motif for CD84. Moreover, Fes, which has been described to become phosphorylated after substrate binding, also gets phosphorylated when coexpressed with CD84. Consistently, Fes was observed to be more phosphorylated after CD84 and FcεRI co-cross-linking. The phosphorylation of the protein phosphatase Src homology region 2 domain-containing phosphatase-1 also increases after CD84 and FcεRI coengagement. Taken together, our results show that CD84 is highly expressed in mast cells and that it contributes to the regulation of FcεRI signaling in SAP- and EAT-2-independent and Fes- and Src homology region 2 domain-containing phosphatase-1-dependent mechanisms.


Subject(s)
Antigens, CD/immunology , Mast Cells/immunology , Receptors, IgE/immunology , Signal Transduction/immunology , Antigens, CD/metabolism , Cell Degranulation/immunology , Cell Line , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Mast Cells/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signaling Lymphocytic Activation Molecule Family , Transfection , Two-Hybrid System Techniques
15.
Mol Immunol ; 45(12): 3446-53, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18479751

ABSTRACT

Natural killer (NK) cell cytotoxicity requires triggering of activation receptors over inhibitory receptors. CD244, a member of CD150 receptor family, positively regulates NK-mediated lyses by activating an intracellular multiproteic signaling network that involves the adaptors X-linked lymphoproliferative gene product SAP and 3BP2. However, the exact mechanisms used by 3BP2 to enhance CD244-mediated cytotoxicity are still not fully understood. Here using the human NK cell line YT-overexpressing 3BP2, we found that the adaptor increases CD244, PI3K, and Vav phosphorylation upon CD244 engagement. The use of enzymatic inhibitors revealed that 3BP2-dependent cytolysis enhancement was PKC-dependent and PI3K-ERK independent. Furthermore, 3BP2 overexpression enhanced PKC delta phosphorylation. SAP knockdown expression inhibited PKC delta activation, indicating that the activating role played by 3BP2 depends upon the presence of SAP. In conclusion, our data show that 3BP2 acts downstream of SAP, increases CD244 phosphorylation and links the receptor with PI3K, Vav, PLC gamma, and PKC downstream events in order to achieve maximum NK killing function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , Cytotoxicity, Immunologic , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase C-delta/metabolism , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Androstadienes/pharmacology , Animals , Cell Line , Cytotoxicity, Immunologic/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoprecipitation , Mice , Models, Immunological , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction/drug effects , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Wortmannin
16.
Mol Immunol ; 45(8): 2138-49, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18243321

ABSTRACT

Signaling through the high-affinity receptor for immunoglobulin E (Fc epsilon RI) results in the coordinated activation of tyrosine kinases, thus leading to calcium mobilization, degranulation, and leukotriene and cytokine synthesis. Here, we show that CD84, a member of the CD150 family of leukocyte receptors, inhibits Fc epsilon RI-mediated mast cell degranulation in CD84-transfected rat basophilic leukaemia-2H3 mast cell line cells (RBL-2H3) through homophilic interaction. There was no reduction in overall protein phosphorylation following IgE triggering in CD84 RBL-2H3 cells. Indeed, phosphorylation of Dok-1 and c-Cbl increased in CD84 RBL-2H3, suggesting that inhibition is mediated by these molecules. MAP kinase phosphorylation (ERK1/2, JNK and p38) and cytokine synthesis were impaired in CD84 RBL-2H3. This inhibitory mechanism was independent of SAP and SHP-2 recruitment. Interestingly, CD84 mutants in tyrosines (Y279F and DeltaY324) reversed this inhibitory profile. These data suggest that CD84 may play a role in modulating Fc epsilon RI-mediated signaling in mast cells. Thus, CD84 could play a protective role against undesired allergic and inflammatory responses.


Subject(s)
Antigens, CD/immunology , Receptors, IgE/antagonists & inhibitors , Receptors, IgE/immunology , Signal Transduction , Amino Acid Sequence , Animals , Antigens, CD/chemistry , COS Cells , Cell Degranulation/immunology , Cell Line, Tumor , Chlorocebus aethiops , Cytokines/biosynthesis , Humans , Immunoglobulin E/immunology , Intracellular Signaling Peptides and Proteins/immunology , Mast Cells/immunology , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutant Proteins/immunology , Phosphorylation , Protein Binding , Rats , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Transfection , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...