Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Cancer ; 130(1): 29-39, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21480217

ABSTRACT

Identification of cancer stem cells is crucial for advancing cancer biology and therapy. Several markers including CD24, CD44, CD117, CD133, the G subfamily of ATP-binding cassette transporters (ABCG), epithelial specific antigen (ESA) and aldehyde dehydrogenase (ALDH) are used to identify and investigate human epithelial cancer stem cells in the literature. We have now systemically analyzed and compared the expression of these markers in fresh ovarian epithelial carcinomas. Although the expression levels of these markers were unexpectedly variable and partially overlapping in fresh ovarian cancer cells from different donors, we reliably detected important levels of CD133 and ALDH in the majority of fresh ovarian cancer. Furthermore, most of these stem cell markers including CD133 and ALDH were gradually lost following in vitro passage of primary tumor cells. However, the expression of ALDH and CD133, but not CD24, CD44 and CD117, could be partially rescued by the in vitro serum-free and sphere cultures and by the in vivo passage in the immune-deficient xenografts. ALDH+ and CD133+ cells formed three-dimensional spheres more efficiently than their negative counterparts. These sphere-forming cells expressed high levels of stem cell core gene transcripts and could be expanded and form additional spheres in long-term culture. ALDH+ , CD133+ and ALDH+ CD133+ cells from fresh tumors developed larger tumors more rapidly than their negative counterparts. This property was preserved in the xenografted tumors. Altogether, the data suggest that ALDH+ and CD133+ cells are enriched with ovarian cancer-initiating (stem) cells and that ALDH and CD133 may be widely used as reliable markers to investigate ovarian cancer stem cell biology.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antigens, CD/metabolism , Glycoproteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peptides/metabolism , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Antigens, CD/genetics , Carcinoma, Ovarian Epithelial , Cell Proliferation , Female , Flow Cytometry , Fluorescent Antibody Technique , Glycoproteins/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Peptides/genetics , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
2.
Sci Transl Med ; 3(104): 104ra100, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21998407

ABSTRACT

T helper 17 (TH17) cells have been shown to contribute to multiple disease systems. However, the functional phenotype and survival pattern of TH17 cells as well as the underlying mechanisms that control TH17 cells have been poorly investigated in humans, significantly hampering the clinical targeting of these cells. Here, we studied human TH17 cells in the pathological microenvironments of graft-versus-host disease, ulcerative colitis, and cancer; TH17 cell numbers were increased in the chronic phase of these diseases. Human TH17 cells phenotypically resembled terminally differentiated memory T cells but were distinct from central memory, exhausted, and senescent T cells. Despite their phenotypic markers of terminal differentiation, TH17 cells mediated and promoted long-term antitumor immunity in in vivo adoptive transfer experiments. Furthermore, TH17 cells had a high capacity for proliferative self-renewal, potent persistence, and apoptotic resistance in vivo, as well as plasticity-converting into other types of TH cells. These cells expressed a relatively specific gene signature including abundant antiapoptotic genes. We found that hypoxia-inducible factor-1α and Notch collaboratively controlled key antiapoptosis Bcl-2 family gene expression and function in TH17 cells. Together, these data indicate that human TH17 cells may be a long-lived proliferating effector memory T cell population with unique genetic and functional characteristics. Targeting TH17-associated signaling pathway would be therapeutically meaningful for treating patients with autoimmune disease and advanced tumor.


Subject(s)
Immunologic Memory , Th17 Cells/cytology , Animals , Apoptosis , Autoimmune Diseases/immunology , Cell Differentiation , Cell Line, Tumor , Cellular Senescence , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Phenotype , Receptors, Notch/metabolism
3.
J Immunol ; 186(7): 4388-95, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21357259

ABSTRACT

Foxp3(+)CD4(+) regulatory T (Treg) cells inhibit immune responses and temper inflammation. IL-17(+)CD4(+) T (Th17) cells mediate inflammation of autoimmune diseases. A small population of IL-17(+)Foxp3(+)CD4(+) T cells has been observed in peripheral blood in healthy human beings. However, the biology of IL-17(+)Foxp3(+)CD4(+) T cells remains poorly understood in humans. We investigated their phenotype, cytokine profile, generation, and pathological relevance in patients with ulcerative colitis. We observed that high levels of IL-17(+)Foxp3(+)CD4(+) T cells were selectively accumulated in the colitic microenvironment and associated colon carcinoma. The phenotype and cytokine profile of IL-17(+)Foxp3(+)CD4(+) T cells was overlapping with Th17 and Treg cells. Myeloid APCs, IL-2, and TGF-ß are essential for their induction from memory CCR6(+) T cells or Treg cells. IL-17(+)Foxp3(+)CD4(+) T cells functionally suppressed T cell activation and stimulated inflammatory cytokine production in the colitic tissues. Our data indicate that IL-17(+)Foxp3(+) cells may be "inflammatory" Treg cells in the pathological microenvironments. These cells may contribute to the pathogenesis of ulcerative colitis through inducing inflammatory cytokines and inhibiting local T cell immunity, and in turn may mechanistically link human chronic inflammation to tumor development. Our data therefore challenge commonly held beliefs of the anti-inflammatory role of Treg cells and suggest a more complex Treg cell biology, at least in the context of human chronic inflammation and associated carcinoma.


Subject(s)
Inflammation Mediators/physiology , Interleukin-17/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cells, Cultured , Chronic Disease , Coculture Techniques , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytokines/biosynthesis , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/physiology , Growth Inhibitors/biosynthesis , Growth Inhibitors/physiology , Humans , Immune Tolerance/immunology , Inflammation Mediators/metabolism , Interleukin-17/biosynthesis , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , T-Lymphocytes, Regulatory/metabolism
4.
Blood ; 114(6): 1141-9, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19470694

ABSTRACT

Th17 cells play an active role in autoimmune diseases. However, the nature of Th17 cells is poorly understood in cancer patients. We studied Th17 cells, the associated mechanisms, and clinical significance in 201 ovarian cancer patients. Tumor-infiltrating Th17 cells exhibit a polyfunctional effector T-cell phenotype, are positively associated with effector cells, and are negatively associated with tumor-infiltrating regulatory T cells. Tumor-associated macrophages promote Th17 cells through interleukin-1beta (IL-1beta), whereas tumor-infiltrating regulatory T cells inhibit Th17 cells through an adenosinergic pathway. Furthermore, through synergistic action between IL-17 and interferon-gamma, Th17 cells stimulate CXCL9 and CXCL10 production to recruit effector T cells to the tumor microenvironment. The levels of CXCL9 and CXCL10 are associated with tumor-infiltrating effector T cells. The levels of tumor-infiltrating Th17 cells and the levels of ascites IL-17 are reduced in more advanced diseases and positively predict patient outcome. Altogether, Th17 cells may contribute to protective human tumor immunity through inducing Th1-type chemokines and recruiting effector cells to the tumor microenvironment. Inhibition of Th17 cells represents a novel immune evasion mechanism. This study thus provides scientific and clinical rationale for developing novel immune-boosting strategies based on promoting the Th17 cell population in cancer patients.


Subject(s)
Interleukin-17/immunology , Macrophages/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Aged , Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Female , Humans , Immunotherapy , Interleukin-1beta/immunology , Macrophages/pathology , Middle Aged , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , T-Lymphocytes, Helper-Inducer/pathology
5.
Cancer Res ; 69(9): 3995-4000, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19383912

ABSTRACT

Activated T cells may express FOXP3. It is thought that FOXP3 is not a specific marker to determine regulatory T cells (Treg) in humans. Here, we examined the functional phenotype and cytokine profile of the in vitro induced FOXP3(+) T cells, primary FOXP3(+) and FOXP3(-) T cells in patients with ulcerative colitis and tumors including colon carcinoma, melanoma, hepatic carcinoma, ovarian carcinoma, pancreatic cancer, and renal cell carcinoma. We observed similar levels of suppressive capacity of primary FOXP3(+) T cells in blood, tumors, and colitic tissues. Compared with primary FOXP3(-) T cells in the same microenvironment, these primary FOXP3(+) T cells expressed minimal levels of effector cytokines, negligible amount of cytotoxic molecule granzyme B, and levels of suppressive molecules interleukin-10 and PD-1. Although the in vitro activated T cells expressed FOXP3, these induced FOXP3(+) T cells expressed high levels of multiple effector cytokines and were not functionally suppressive. The data reinforce the fact that FOXP3 remains an accurate marker to define primary Tregs in patients with cancer and autoimmune disease. We suggest that the combination of FOXP3 and cytokine profile is useful for further functionally distinguishing primary Tregs from activated conventional T cells.


Subject(s)
Colitis, Ulcerative/immunology , Forkhead Transcription Factors/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Colitis, Ulcerative/blood , Cytokines/immunology , Flow Cytometry , Forkhead Transcription Factors/blood , Humans , Neoplasms/blood
6.
Blood ; 114(2): 357-9, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19289853

ABSTRACT

It has been reported that ectopically expressed interleukin-17 (IL-17) in tumor cells suppresses tumor progression through enhanced antitumor immunity in immune competent mice or promote tumor progression through an increase in inflammatory angiogenesis in immune-deficient mice. The role of endogenous IL-17 in tumor immunity remains undefined. Here we showed that tumor growth and lung metastasis were enhanced in IL-17-deficient mice, associated with decreased interferon-gamma(+) natural killer cells and tumor specific interferon-gamma(+) T cells in the tumor draining lymph nodes and tumors. Together with the published data showing that in vitro transforming growth factor-beta and IL-6-polarized Th17 cells induce tumor regression, our work supports the notion that endogenous IL-17 or/and Th17 cells may play a protective role in tumor immunity.


Subject(s)
Interleukin-17/immunology , Neoplasms/immunology , Neoplasms/pathology , Animals , Disease Progression , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasms/genetics
7.
J Immunol ; 181(9): 5842-6, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941172

ABSTRACT

Th1-derived IFN-gamma targets naive T cells and inhibits Th17 development. However, Th1, Th17, and memory but not naive T cells are colocalized in an inflammatory environment. To demonstrate the kinetic relationship between these T cell subsets, we investigated the role of IFN-gamma in regulating the development and balance between Th17 and Th1 in humans. We show that IFN-gamma stimulates B7-H1 expression on APC subsets and abates their Th1 polarization capacity in a B7-H1-dependent manner. Interestingly, IFN-gamma triggers APCs to produce IL-1 and IL-23 and enables them to induce memory Th17 expansion via IL-1 and IL-23 in a B7-H1-independent manner. We propose a novel dynamic between Th1 and Th17 in the course of inflammation as follows: Th1-mediated inflammation is attenuated by IFN-gamma-induced B7-H1 on APCs and is evolved toward Th17-mediated chronic inflammation by IFN-gamma-induced, APC-derived IL-1 and IL-23. Our study challenges the dogma that IFN-gamma suppresses Th17 and enhances Th1 development.


Subject(s)
Antigen-Presenting Cells/immunology , Cell Differentiation/immunology , Growth Inhibitors/physiology , Immunologic Memory , Interferon-gamma/physiology , Interleukin-17/physiology , Th1 Cells/cytology , Antigen-Presenting Cells/metabolism , Antigens, CD/physiology , B7-H1 Antigen , Cells, Cultured , Coculture Techniques , Humans , Interleukin-1/physiology , Interleukin-17/biosynthesis , Interleukin-23/physiology , Th1 Cells/immunology , Th1 Cells/metabolism
8.
J Immunol ; 181(7): 4733-41, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802076

ABSTRACT

Th1 and Th17 T cells are often colocalized in pathological environments, yet Th1-derived IFN-gamma inhibits Th17 cell development in vitro. We explored the physiologic basis of this paradox in humans. In this study, we demonstrate increased the number of CD4(+) and CD8(+) IL-17(+) T cells in skin lesions of psoriasis. Furthermore, we show that myeloid APCs potently support induction of IL-17(+) T cells, and that this activity is greatly increased in psoriasis. We tested stimuli that might account for this activity. Th1 cells and IFN-gamma are increased in psoriatic blood and lesional skin. We show that IFN-gamma programs myeloid APCs to induce human IL-17(+) T cells via IL-1 and IL-23. IFN-gamma also stimulates APC production of CCL20, supporting migration of IL-17(+) T cells, and synergizes with IL-17 in the production of human beta-defensin 2, an antimicrobial and chemotactic protein highly overexpressed by psoriatic keratinocytes. This study reveals a novel mechanistic interaction between Th1 and IL-17(+) T cells, challenges the view that Th1 cells suppress Th17 development through IFN-gamma, and suggests that Th1 and IL-17(+) T cells may collaboratively contribute to human autoimmune diseases.


Subject(s)
Cell Movement/immunology , Interferon-gamma/physiology , Interleukin-17/biosynthesis , Psoriasis/immunology , Psoriasis/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Communication/immunology , Cells, Cultured , Coculture Techniques , Humans , Immunophenotyping , Interleukin-17/metabolism , Interleukin-17/physiology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Psoriasis/metabolism , Skin/cytology , Skin/immunology , Skin/metabolism , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology
9.
J Immunol ; 179(3): 1423-6, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641006

ABSTRACT

In this report, we show that IL-17(+)CD4(+) and IL-17(+)CD8(+) T cells are largely found in lung and digestive mucosa compartments in normal mice. Endogenous and exogenous IL-1 dramatically contribute to IL-17(+) T cell differentiation mediated by TGFbeta and IL-6. IL-1 is capable of stimulating IL-17(+) T cell differentiation in the absence of IL-6. Furthermore, although IL-2 reduces IL-17(+) T cell differentiation, IL-1 completely disables this effect. Mechanistically, IL-1 and IL-2 play opposite roles in regulating the expression of several molecules regulating Th17 cell differentiation, including the orphan nuclear receptor ROR gamma t, the IL-1 receptor, and the IL-23 receptor. IL-1 subverts the effects of IL-2 on the expression of these gene transcripts. Altogether, our work demonstrates that IL-6 is important but not indispensable for IL-17(+) T cell differentiation and that IL-1 plays a predominant role in promoting IL-17(+) T cell induction. Thus, the IL-17(+) T cell pool may be controlled by the local cytokine profile in the microenvironment.


Subject(s)
Cell Differentiation/immunology , Growth Inhibitors/antagonists & inhibitors , Growth Inhibitors/physiology , Interleukin-17/biosynthesis , Interleukin-1/physiology , Interleukin-2/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Interleukin-1/metabolism , Interleukin-2/antagonists & inhibitors , Interleukin-6/deficiency , Interleukin-6/genetics , Interleukin-6/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...