Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
iScience ; 26(6): 106904, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37275530

ABSTRACT

CD200 is overexpressed in many solid tumors and considered as an immune checkpoint molecule dampening cancer immunity. In this study, we found that CD200R-/- mice were significantly more potent in rejecting these CD200+ tumors. scRNA sequencing demonstrated that tumors from CD200R-/- mice had more infiltration of CD4+ and CD8+ T cells, and NK cells but less infiltration of neutrophils. Antibody depletion experiments revealed that immune effector cells are crucial in inhibiting tumor growth in CD200R-/- mice. Mechanistically, we found that CD200R signaling regulates the expression of chemokines in tumor-associated myeloid cells (TAMCs). In the absence of CD200R, TAMCs increased expression of CCL24 and resulted in increased infiltration of eosinophils, which contributes to anti-tumor activity. Overall, we conclude that CD200R signaling contributes to unfavorable TME through chemokine-dependent recruitment of immune suppressive neutrophils and exclusion of anti-cancer immune effectors. Our study has implications in developing CD200-CD200R targeted immunotherapy of solid tumors.

2.
Am J Transl Res ; 14(4): 2291-2300, 2022.
Article in English | MEDLINE | ID: mdl-35559385

ABSTRACT

CD24 is a glycosyl-phosphatidylinositol (GPI) anchored cell surface glycoprotein with a variety of immunomodulatory functions such as inhibition of thymic generation of autoreactive T cells, regulation of antigen presenting cell functions, and mediation of autoimmunity. Given the autoimmune nature of FoxP3+ regulatory T cells and their importance in autoimmune diseases, we hypothesize that CD24 regulates the generation and functions of Treg cells. Through the analysis of the Treg repertoire in two strains of CD24-deficient mice, we found that CD24 does not globally affect the thymic generation of Treg cells. However, CD24 is abundantly expressed on Treg cells, and CD24 antibody treatment of Treg cells enhances their suppressive functions. Concurrently, we observed CD24-deficient Treg cells exhibit increased suppressive functions and produce more IL-10 compared to their wild type counterparts. In addition, CD24-deficient Treg cells exhibited more potent suppressive capacity in inhibiting the development of experimental autoimmune encephalomyelitis (EAE) in mice. Thus, CD24 on Treg cells regulates their suppressive functions. Our findings can partially explain the resistance of EAE development in CD24-deficient mice and CD24 polymorphism-associated susceptibility of human autoimmune diseases. Further investigations regarding mechanisms of CD24 regulation of Treg function may lead to a new approach for the immunotherapy of human autoimmune diseases.

3.
J Immunol ; 208(9): 2239-2245, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35418466

ABSTRACT

IL-27 is a pleiotropic cytokine that exhibits stimulatory/regulatory functions on multiple lineages of immune cells including T lymphocytes. In this study, we demonstrate that IL-27 directly induces CCL5 production by T lymphocytes, particularly CD8+ T cells in vitro and in vivo. IL-27-induced CCL5 production is IL-27R-dependent. In CD4+ T cells, IL-27-induced CCL5 production was primarily dependent on Stat1 activation, whereas in CD8+ T cells, Stat1 deficiency does not abrogate CCL5 induction. A chromatin immunoprecipitation assay revealed that in the CCL5 promoter region, both putative Stat3 binding sites exhibit significant binding to Stat3, whereas only one out of four Stat1 binding sites displays moderate binding to Stat1. In tumor-bearing mice, IL-27 induced dramatic production of CCL5 in tumor-infiltrating T cells. IL-27-induced CCL5 appears to contribute to an IL-27-mediated antitumor effect. This is signified by diminished tumor inhibition in anti-CCL5- and IL-27-treated mice. Additionally, intratumor delivery of CCL5 mRNA using lipid nanoparticles significantly inhibited tumor growth. Thus, IL-27 induces robust CCL5 production by T cells, which contributes to antitumor activity.


Subject(s)
Interleukin-27 , Animals , CD8-Positive T-Lymphocytes , Cytokines , Gene Expression , Liposomes , Mice , Nanoparticles
4.
J Control Release ; 345: 306-313, 2022 05.
Article in English | MEDLINE | ID: mdl-35301053

ABSTRACT

Cytokines are important immunotherapeutics with approved drugs for the treatment of human cancers. However, systemic administration of cytokines often fails to achieve adequate concentrations to immune cells in tumors due to dose-limiting toxicity. Thus, developing localized therapy that directly delivers immune-stimulatory cytokines to tumors may improve the therapeutic efficacy. In this study, we generated novel lipid nanoparticles (LNPs) encapsulated with mRNAs encoding cytokines including IL-12, IL-27 and GM-CSF, and tested their anti-tumor activity. We first synthesized ionizable lipid materials containing di-amino groups with various head groups (DALs). The novel DAL4-LNP effectively delivered different mRNAs in vitro to tumor cells and in vivo to tumors. Intratumoral injection of DAL4-LNP loaded with IL-12 mRNA was most potent in inhibiting B16F10 melanoma tumor growth compared to IL-27 or GM-CSF mRNAs in monotherapy. Furthermore, intratumoral injection of dual DAL4-LNP-IL-12 mRNA and IL-27 mRNA showed a synergistic effect in suppressing tumor growth without causing systematic toxicity. Most importantly, intratumoral delivery of IL-12 and IL-27 mRNAs induced robust infiltration of immune effector cells, including IFN-γ and TNF-α producing NK and CD8+ T cells into tumors. Thus, intratumoral administration of DAL-LNP loaded with IL-12 and IL-27 mRNA provides a new treatment strategy for cancer.


Subject(s)
Interleukin-27 , Nanoparticles , Neoplasms , CD8-Positive T-Lymphocytes , Cytokines , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Immunotherapy , Interleukin-12/genetics , Liposomes , Neoplasms/drug therapy , RNA, Messenger/genetics , RNA, Messenger/therapeutic use
5.
Front Cell Dev Biol ; 9: 739816, 2021.
Article in English | MEDLINE | ID: mdl-34692697

ABSTRACT

CD200-CD200R pathway regulates immune responses and has been implicated in the pathogenesis of a number of cancer types. CD200 blockade is considered a strategy for immunotherapy of CD200-positive cancers such as melanoma. Thus, it is critical to understand the potential impacts of CD200 blockade in a more human relevant tumor model. In this study, we evaluated these issues using the CD200+ Yumm1.7 mouse melanoma model. Yumm1.7 cells bear Braf/Pten mutations resembling human melanoma. We found that Yumm1.7 tumors grow significantly faster in CD200R-/- mice compared to wild type mice. Analysis of tumor immune microenvironment (TIME) revealed that tumors from CD200R-/- or anti-CD200 treated mice had downregulated immune cell contents and reduced TCR clonality compared to tumors from untreated wild type mice. T cells also showed impaired effector functions, as reflected by reduced numbers of IFN-γ+ and TNF-α+ T cells. Mechanistically, we found upregulation of the CCL8 gene in CD200R-/- tumors. In vitro co-culture experiments using Yumm1.7 tumor cells with bone marrow derived macrophages (BMDM) from WT and CD200R-/- mice confirmed upregulation of macrophage CCL8 in the absence of CD200-CD200R interaction. Finally, we found that anti-CD200 therapy failed to show efficacy either alone or in combination with checkpoint inhibitors such as anti-PD-1 or anti-CTLA4 in inhibiting Yumm1.7 tumor growth. Given that CD200R-deficiency or anti-CD200 treatment leads to reduced T cell responses in TME, using blockade of CD200 as an immunotherapy for cancers such as melanoma should be practiced with caution.

6.
Adv Exp Med Biol ; 1223: 155-165, 2020.
Article in English | MEDLINE | ID: mdl-32030689

ABSTRACT

Tumor-associated inflammation and immune responses are key components in the tumor microenvironment (TME) which regulate tumor growth, progression, and metastasis. Tumor-associated myeloid cells (TAMCs) are a group of cells that play multiple key roles including induction of tumor-associated inflammation/angiogenesis and regulation of tumor-specific T-cell responses. Thus, identification and characterization of key pathways that can regulate TAMCs are of critical importance for developing cancer immunotherapy. Recent studies suggest that CD200-CD200 receptor (CD200R) interaction may be important in regulating the TME via affecting TAMCs. In this chapter, we will give a brief overview of the CD200-CD200R axis, including the biology behind CD200-CD200R interaction and the role(s) it plays in tumor microenvironment and tumor growth, and activation/effector functions of T cells. We will also discuss CD200-CD200R's role as potential checkpoint molecules for cancer immunotherapy. Further investigation of the CD200-CD200R pathway will not only advance our understanding of tumor pathogenesis and immunity but also provide the rationale for CD200-CD200R-targeted immunotherapy of human cancer.


Subject(s)
Antigens, CD/metabolism , Immunotherapy , Neoplasms/therapy , Orexin Receptors/metabolism , Tumor Microenvironment/immunology , Antigens, CD/immunology , Humans , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Orexin Receptors/immunology
7.
Metab Brain Dis ; 35(2): 327-333, 2020 02.
Article in English | MEDLINE | ID: mdl-31728886

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder associated with different epidemiological, genetic, epigenetic, and environmental factors. Although its etiology is not fully understood, immune dysfunction is implicated in this disease. Recently, a large number of genes encoding long noncoding RNAs (lncRNAs) were discovered which act as positive or negative regulators of neighboring target genes. The lncRNA, Interferon gamma-antisense RNA (IFNG-AS1), regulates expression levels of the Interferon gamma (IFNG) gene. In the present study, we investigated expression of IFNG and IFNG-AS1 in 50 children with ASD (15 females and 35 males, mean age: 6 ± 1.4 years) and 50 healthy controls (14 females and 36 males, mean age: 6 ± 1.74 years) by real time PCR technique. The results showed significant up-regulation of IFNG and down-regulation of IFNG-AS1 expression in children with ASD compared to controls (Fold change = 1.5, P < 0.0001; Fold change = -0.143, P = 0.013, respectively). The IFNG expression level increase was more pronounced in male ASD children (Fold change = 1.621; p < 0.0001). Our data reveal a functional disruption in the interactive network of IFNG/IFNG-AS1 regulation, which could be a contributing factor in the chronic inflammatory aspect of ASD. Our findings can help understanding the underlying contributors to ASD pathogenesis and find novel treatment options for children with ASD.


Subject(s)
Autism Spectrum Disorder/metabolism , Interferon-gamma/biosynthesis , RNA, Long Noncoding/biosynthesis , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/genetics , Case-Control Studies , Child , Female , Gene Expression , Humans , Interferon-gamma/genetics , Male , RNA, Long Noncoding/genetics
8.
J Immunol ; 197(4): 1489-97, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27385779

ABSTRACT

CD200 is a cell surface glycoprotein that functions through engaging CD200R on cells of the myeloid lineage and inhibits their functions. Expression of CD200 was implicated in a variety of human cancer cells, including melanoma cells; however, its roles in tumor growth and immunity are not clearly understood. In this study, we used CD200R-deficient mice and the B16 tumor model to evaluate this issue. We found that CD200R-deficient mice exhibited accelerated growth of CD200(+), but not CD200(-), B16 tumors. Strikingly, CD200R-deficient mice receiving CD200(+) B16 cells i.v. exhibited massive tumor growth in multiple organs, including liver, lung, kidney, and peritoneal cavity, whereas the growth of the same tumors in wild-type mice was limited. CD200(+) tumors grown in CD200R-deficient mice contained higher numbers of CD11b(+)Ly6C(+) myeloid cells, exhibited increased expression of VEGF and HIF1α genes with increased angiogenesis, and showed significantly reduced infiltration of CD4(+) and CD8(+) T cells, presumably as the result of reduced expression of T cell chemokines, such as CXCL9 and CXCL16. The liver from CD200R-deficient mice, under metastatic growth of CD200(+) tumors, contained significantly increased numbers of CD11b(+)Gr1(-) myeloid cells and Foxp3(+) regulatory T cells and reduced numbers of NK cells. Liver T cells also had a reduced capacity to produce IFN-γ or TNF-α. Taken together, we revealed a critical role for CD200R signaling in limiting the growth and metastasis of CD200(+) tumors. Thus, targeting CD200R signaling may potentially interfere with the metastatic growth of CD200(+) tumors, like melanoma.


Subject(s)
Antigens, CD/metabolism , Melanoma, Experimental/pathology , Neoplasm Invasiveness/pathology , Signal Transduction/physiology , Animals , Antigens, CD/immunology , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockout Techniques , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/immunology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Real-Time Polymerase Chain Reaction , Tumor Microenvironment/physiology
9.
Oncoimmunology ; 4(7): e1014232, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26140236

ABSTRACT

Interleukin-10 (IL-10) is a potent anti-inflammatory cytokine that regulates immune responses. IL-10 has also been shown to enhance antitumor CD8+ T-cell responses in tumor models although the underlying mechanisms are not fully understood. In this study, we used a series of genetic mouse models and the mouse plasmacytoma J558 model to investigate this issue. J558 tumors grew significantly faster in IL-10-/- mice than in wild type (WT) mice, but similarly in IL-10 -/- Rag2 -/- and Rag2 -/- mice. Tumors from IL-10 -/- mice contained fewer IFN-γ-producing CD8+ and CD4+ T cells than tumors from WT mice. Strikingly, depletion of total CD4+ T cells, but not CD25+ cells, resulted in tumor eradication in IL-10 -/- mice. Adoptive transfer studies revealed that CD4+ T cells from IL-10 -/- mice exhibited more potent suppression of cytotoxic T lymphocyte (CTL)-mediated tumor rejection than their WT counterparts, and IL-10-deficient tumor-infiltrating CD4+ T cells expressed higher levels of PD-L1 and CTLA-4 inhibitory molecules. Although IL-10-deficient CD8+ T cells are not defective in activation and initial rejection of tumors, adoptive transfer studies using IL-10-deficient P1CTL transgenic T cells that recognize the tumor rejection antigen P1A reveal that IL-10 is required for long-term persistence of CTLs and control of tumor growth. Thus, we have found that IL-10 enhances antitumor CTL responses by inhibiting highly suppressive CD4+ T cells and promoting CTL persistence. These data have important implications for the design of immunotherapy for human cancer.

10.
Eur J Immunol ; 43(2): 468-79, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23225163

ABSTRACT

IL-27 is a member of the IL-12 family of cytokines that is comprised of an IL-12 p40-related protein subunit, EBV-induced gene 3, and a p35-related subunit, p28. IL-27 functions through IL-27R and has been shown to have potent antitumor activity via activation of a variety of cellular components, including antitumor CD8(+) T-cell responses. However, the exact mechanisms of how IL-27 enhances antitumor CD8(+) T-cell responses remain unclear. Here we show that IL-27 significantly enhances the survival of activated tumor antigen-specific CD8(+) T cells in vitro and in vivo, and programs tumor antigen-specific CD8(+) T cells into memory precursor-like effector cells, characterized by upregulation of Bcl-6, SOCS3, Sca-1, and IL-10. While STAT3 activation and the CTL survival-enhancing effects can be independent of CTL IL-10 production, we show here that IL-27-induced CTL IL-10 production contributes to memory precursor cell phenotype induction, CTL memory, and tumor rejection. Thus, IL-27 enhances antitumor CTL responses via programming tumor antigen-specific CD8(+) T cells into a unique memory precursor type of effector cells characterized by a greater survival advantage. Our results have important implications for designing immunotherapy against human cancer.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-10/immunology , Interleukins/immunology , Animals , Antigens, Ly/immunology , Antigens, Ly/metabolism , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival/immunology , Interleukin-10/metabolism , Interleukins/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-bcl-6/immunology , Proto-Oncogene Proteins c-bcl-6/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Up-Regulation/immunology
11.
Oncoimmunology ; 1(6): 971-973, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-23162775

ABSTRACT

CD200 is a cell surface glycoprotein that has been implicated in a variety of human cancer cells and has been thought to play a pro-tumor role. However, in our recent study we have revealed that CD200 on cancer cells inhibits tumor formation and metastasis through inhibition of myeloid cells.

12.
J Immunol ; 188(7): 3099-106, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22387555

ABSTRACT

EBV-induced gene 3 (EBI3)-encoded protein can form heterodimers with IL-27P28 and IL-12P35 to form IL-27 and IL-35. IL-27 and IL-35 may influence autoimmunity by inhibiting Th17 differentiation and facilitating the inhibitory roles of Foxp3(+) regulatory T (Treg) cells, respectively. In this study, we have evaluated the development of experimental autoimmune encephalomyelitis (EAE) in EBI3-deficient mice that lack both IL-27 and IL-35. We found that myelin oligodendrocyte glycoprotein peptide immunization resulted in marginally enhanced EAE development in EBI3-deficient C57BL6 and 2D2 TCR-transgenic mice. EBI3 deficiency resulted in significantly increased Th17 and Th1 responses in the CNS and increased T cell production of IL-2 and IL-17 in the peripheral lymphoid organs. EBI3-deficient and -sufficient 2D2 T cells had equal ability in inducing EAE in Rag1(-/-) mice; however, more severe disease was induced in EBI3(-/-)Rag1(-/-) mice than in Rag1(-/-) mice by 2D2 T cells. EBI3-deficient mice had increased numbers of CD4(+)Foxp3(+) Treg cells in peripheral lymphoid organs. More strikingly, EBI3-deficient Treg cells had more potent suppressive functions in vitro and in vivo. Thus, our data support an inhibitory role for EBI3 in Th17, Th1, IL-2, and Treg responses. Although these observations are consistent with the known functions of IL-27, the IL-35 contribution to the suppressive functions of Treg cells is not evident in this model. Increased Treg responses in EBI3(-/-) mice may explain why the EAE development is only modestly enhanced compared with wild-type mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Receptors, Cytokine/physiology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Encephalomyelitis, Autoimmune, Experimental/etiology , Forkhead Transcription Factors/analysis , Gene Expression Regulation/immunology , Homeodomain Proteins/immunology , Immunosuppression Therapy , Interleukin-2/immunology , Interleukins/deficiency , Interleukins/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens , RNA, Messenger/biosynthesis , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , T-Cell Antigen Receptor Specificity , Th1 Cells/immunology
13.
PLoS One ; 7(2): e31442, 2012.
Article in English | MEDLINE | ID: mdl-22319630

ABSTRACT

CD200 is a cell surface glycoprotein that functions through engaging CD200 receptor on cells of the myeloid lineage and inhibits their functions. Expression of CD200 has been implicated in a variety of human cancer cells including melanoma cells and has been thought to play a protumor role. To investigate the role of cancer cell expression of CD200 in tumor formation and metastasis, we generated CD200-positive and CD200-negative B16 melanoma cells. Subcutaneous injection of CD200-positive B16 melanoma cells inhibited tumor formation and growth in C57BL/6 mice but not in Rag1⁻/⁻C57BL/6 mice. However, i.v. injection of CD200-positive B16 melanoma cells dramatically inhibited tumor foci formation in the lungs of both C57BL/6 and Rag1⁻/⁻C57BL6 mice. Flow cytometry analysis revealed higher expression of CD200R in Gr1⁺ myeloid cells in the lung than in peripheral myeloid cells. Depletion of Gr1⁺ cells or stimulation of CD200R with an agonistic antibody in vivo dramatically inhibited tumor foci formation in the lungs. In addition, treatment with tumor antigen specific CD4 or CD8 T cells or their combination yielded a survival advantage for CD200 positive tumor bearing mice over mice bearing CD200-negative tumors. Taken together, we have revealed a novel role for CD200-CD200R interaction in inhibiting tumor formation and metastasis. Targeting CD200R may represent a novel approach for cancer immunotherapy.


Subject(s)
Antigens, CD/biosynthesis , Lung Neoplasms/secondary , Melanoma/pathology , Myeloid Cells/pathology , Neoplasm Metastasis/prevention & control , Animals , Antigens, Surface , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Humans , Melanoma/metabolism , Mice , Neoplasms/pathology , Orexin Receptors , Receptors, Cell Surface
14.
Eur J Immunol ; 40(9): 2569-79, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20662098

ABSTRACT

CD200 is a cell-surface glycoprotein that functions through interaction with the CD200 receptor on myeloid lineage cells to regulate myeloid cell functions. Expression of CD200 has been implicated in multiple types of human cancer; however, the impact of tumor expression of CD200 on tumor immunity remains poorly understood. To evaluate this issue, we generated CD200-positive mouse plasmacytoma J558 and mastocytoma P815 cells. We found that established CD200-positive tumors were often completely rejected by adoptively transferred CTL without tumor recurrence; in contrast, CD200-negative tumors were initially rejected by adoptively transferred CTL but the majority of tumors recurred. Tumor expression of CD200 significantly inhibited suppressive activity and IL-10 production by tumor-associated myeloid cells (TAMC), and as a result, more CTL accumulated in the tumor and exhibited a greater capacity to produce IFN-gamma in CD200-positive tumors than in CD200-negative tumors. Neutralization of IL-10 significantly inhibited the suppressor activity of TAMC, and IL-10-deficiency allowed TAMC to kill cancer cells and their antigenic variants, which prevented tumor recurrence during CTL therapy. Thus, tumor expression of CD200 prevents tumor recurrence via inhibiting IL-10 production by TAMC.


Subject(s)
Antigens, CD/metabolism , Cancer Vaccines , Mastocytoma/immunology , Plasmacytoma/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Graft vs Tumor Effect/genetics , Graft vs Tumor Effect/immunology , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-10/metabolism , Mastocytoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/immunology , Neoplasm Transplantation , Plasmacytoma/pathology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Transgenes/genetics , Tumor Escape/genetics , Tumor Escape/immunology
15.
Iran J Immunol ; 4(1): 50-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17652844

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) with their potential to differentiate into mesodermal and non-mesodermal lineages have several immunomodulatory characteristics. These properties make them promising tools in cell and gene therapy. OBJECTIVE: To evaluate the potential therapeutic applications of autologous MSC in improving clinical manifestations of MS patients. METHODS: Ten patients were included in this pilot study. All had progressive disease that had not responded to disease modifying agents including Mitoxantrone. Their Expanded Disability Status Scale (EDSS) score ranged from 3.5 to 6. Patients were injected intrathecally with culture expanded MSCs. They were followed with monthly neurological assessment and a MRI scan at the end of the first year. RESULTS: During 13 to 26 months of follow up (mean: 19 months), the EDSS of one patient improved from 5 to 2.5 score. Four patients showed no change in EDSS. Five patients' EDSS increased from 0.5 to 2.5. In the functional system assessment, six patients showed some degree of improvement in their sensory, pyramidal, and cerebellar functions. One showed no difference in clinical assessment and three deteriorated. The result of MRI assessment after 12 months was as following: seven patients with no difference, two showed an extra plaque, and one patient showed decrease in the number of plaques. CONCLUSION: This preliminary report emphasizes on the feasibility of autologous MSC for treatment of MS patients. However, in order to draw a definitive conclusion a larger sample size is required.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Multiple Sclerosis/therapy , Adult , Cells, Cultured , Feasibility Studies , Female , Follow-Up Studies , Humans , Immunophenotyping , Injections, Spinal , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Pilot Projects
16.
Int Rev Neurobiol ; 79: 13-42, 2007.
Article in English | MEDLINE | ID: mdl-17531836

ABSTRACT

Despite the current limited understanding of the etiology of multiple sclerosis (MS), genetic susceptibility and environmental influences are known driving factors. MS is considered a T-cell-mediated disease given the prevalence of T cells in plaques. Plaque formation is characteristic of this disease attributable to immune mechanisms, triggered by an autoimmune attack aimed at antigens in the myelin sheath or oligodendrocyte proteins. The attack consists of the following: The role of the B cells is twofold: first, as autoreactive B cells they produce autoantibodies, secrete cytokines, clonally replicate memory B cells, and long-living plasma cells which serve to advance the diseased state by their constant production of autoantibodies. Second, as antigen-presenting cells they activate the autoreactive T cells. For this reason, the stipulation that T cell is the cornerstone of MS must be reevaluated. Various studies on pathogenesis of MS have indicated that B cells, as the humoral component of the adaptive immune system, are active participants in pathogenesis and lesion maintenance throughout the disease process. The active role of B cells and autoantibodies makes them an encouraging therapeutic target. Advances in the understanding of B-cell development and activity would allow for an enhanced strategy in the design of autoimmune treatment. For this reason, further investigation is necessary to determine whether depletion of B cells or antibodies may restore immune function.


Subject(s)
B-Lymphocytes/pathology , B-Lymphocytes/physiology , Multiple Sclerosis/pathology , Animals , Autoantigens/immunology , Humans , Multiple Sclerosis/immunology
17.
Int Rev Neurobiol ; 79: 173-202, 2007.
Article in English | MEDLINE | ID: mdl-17531842

ABSTRACT

Scientific advances have demonstrated that autoreactive cells are a component of the healthy immune repertoire. If we define autoimmunity as an active induction of autoreaction, the solution should be an active induction of self-tolerance, and may indicate the direction to explore the future therapies. Microchimerism (MC) refers to the presence of a limited number of nonhost cells in the body of an individual. These cells can enter via blood transfusion and organ transplantation or naturally through pregnancy. Chimeric cells engraft in the host body, develop, proliferate, and are accepted by the immune system as self. These include stem cells that enter the maternal body during fetal stages. These stem cells are also postulated to be helpful reservoirs in protecting the host body. MC has been considered a risk factor in autoimmune disease induction. However, today we know it is a natural phenomenon. MC can be considered a natural model of successful transplantation, the earliest engrafting cells being fetal mesenchymal stem cells (MSCs). MSCs have two notable features. They have an immunosuppressive quality when encountering the adoptive immune system and they display repair-inducing potential within damaged tissues. For the fetus, MC appears to be an effective factor in maternal tolerance induction toward the fetal graft and for the mother; these novel fetal cells might be useful in disease conditions occurring after pregnancy. Hematopoietic stem cell transplantation has become an accepted treatment option for both malignant and nonmalignant diseases and this unique procedure is now being investigated as a potential therapy for multiple sclerosis (MS). Due to the dichotomous properties of MSC, suppressing aggressive immune dysfunction while promoting damaged tissue repair, they may be appropriate therapy for MS.


Subject(s)
Multiple Sclerosis/surgery , Stem Cell Transplantation/methods , Transplantation Chimera/physiology , Animals , Humans
18.
Urol J ; 4(1): 1-9, 2007.
Article in English | MEDLINE | ID: mdl-17514603

ABSTRACT

INTRODUCTION: Microchimerism has become a familiar term in the past few years. Many groups all over the globe, specializing in a diverse array of basic and medical sciences, have turned their attention to microchimerism, its possible role in disease or repair, and its mechanism of action in the host body. We reviewed the current knowledge about this novel term. MATERIALS AND METHODS: We search the PubMed, using all the derivatives of chimera. All papers and their bibliographic information published by December 2005 were reviewed and 61 were selected. RESULTS: Microchimerism is the presence of foreign or nonhost cells in a body. These are cells that live, differentiate, and persist in the host body by definition. These cells can enter the host body in a variety of manners. The most familiar aspect is microchimerism resulting from organ transplant. For many years now scientists have been debating over the interpretation of this phenomenon. We know that donor cell engraftment in the recipient body is a sign of transplantation success. What this means is that the body has developed tolerance toward the foreign organ and created a chimer. CONCLUSION: How long this chimeric state will last, whether these cells will induce or be induced to create a chronic complication in the long run, or will these genetically distinct cell types live peacefully in one body to the end of the host's life are the essence of the ongoing discussion and what probes researchers to continue their search.


Subject(s)
Chimerism , Animals , Autoimmunity/physiology , Female , Humans , Pregnancy , Stem Cells/physiology
19.
BMC Cell Biol ; 7: 14, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16529651

ABSTRACT

BACKGROUND: A hot new topic in medical treatment is the use of mesenchymal stem cells (MSC) in therapy. The low frequency of this subpopulation of stem cells in bone marrow (BM) necessitates their in vitro expansion prior to clinical use. We evaluated the effect of long term culture on the senescence of these cells. RESULTS: The mean long term culture was 118 days and the mean passage number was 9. The average number of PD decreased from 7.7 to 1.2 in the 10th passage. The mean telomere length decreased from 9.19 Kbp to 8.7 kbp in the 9th passage. Differentiation potential dropped from the 6th passage on. The culture's morphological abnormalities were typical of the Hayflick model of cellular aging. CONCLUSION: We believe that MSC enter senescence almost undetectably from the moment of in vitro culturing. Simultaneously these cells are losing their stem cell characteristics. Therefore, it is much better to consider them for cell and gene therapy early on.


Subject(s)
Cellular Senescence/physiology , Mesenchymal Stem Cells/cytology , Adipocytes/cytology , Adolescent , Adult , Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cell Shape , Cells, Cultured , Child , Child, Preschool , Female , Humans , Immunophenotyping , Kinetics , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Osteocytes/cytology , Telomere/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...