Human TH17 Cells Are Long-Lived Effector Memory Cells

Human TH17 cells function as long-lived effector memory cells in the context of chronic disease. Forever Seventeen “It” is that quality possessed by some which draws all others with its magnetic force. Novelist Elinor Glyn In teen television drama parlance, T helper 17 (TH17) cells have been immunology’s “it girl” for the past few years. Discovered in 2007, this beguiling subset of TH cells is associated with autoimmune disease and long-term antitumor immunity, and the presence of these cells in tumors positively correlates with patient survival. Although mouse TH17 cells are believed to be short-lived, studies of advanced human cancers suggest that human TH17 cells persist, but little is known about the nature of these cells in the context of human disease. Now, Kryczek et al. define the detailed phenotype and functional aspects of human TH17 cells in diverse diseased human tissues and find that these cells resemble terminally differentiated, long-lived memory T cells with a twist. The authors used a well-defined human system to study the defining features and behavior of TH17 cells in the pathological microenvironments of human graft-versus-host disease, ulcerative colitis, and cancer. The fraction of TH17 cells was increased in these tissues during the chronic phases of the diseases. Furthermore, the TH17 cells not only displayed characteristics in common with terminally differentiated memory T cells, but also sported some genetic and functional signatures of their own. For example, disease-associated human TH17 cells wore the standard phenotypic markers of terminal differentiation and, in adoptive transfer experiments, drove persistent antitumor immunity. However, these versatile human T cells also displayed a high capacity for proliferative self-renewal, cell-type plasticity, and enriched expression of antiapoptotic genes. The cell membrane signaling protein Notch and the transcriptional regulatory protein hypoxia-inducible factor–1α (HIF-1α) were shown to jointly regulate the expression and function of the Bcl-2 family of antiapoptosis proteins. An intricate understanding of human TH17 cell biology in disease contexts should reveal new “it” molecules—therapeutic targets that permit TH17-directed treatment in patients with autoimmune diseases and advanced tumors. 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.

[1]  W. Zou,et al.  Erratum: TH 17 cells in tumour immunity and immunotherapy (Nature Reviews Immunology (2010) 10 (248-256)) , 2011 .

[2]  Yang Liu,et al.  Targeting HIF1α eliminates cancer stem cells in hematological malignancies. , 2011, Cell stem cell.

[3]  Ke Wu,et al.  IL-17+ Regulatory T Cells in the Microenvironments of Chronic Inflammation and Cancer , 2011, The Journal of Immunology.

[4]  A. Chang,et al.  Intratumoral Expression of IL-17 and Its Prognostic Role in Gastric Adenocarcinoma Patients , 2011, International journal of biological sciences.

[5]  Huanzhong Shi,et al.  Generation and Differentiation of IL-17–Producing CD4+ T Cells in Malignant Pleural Effusion , 2010, The Journal of Immunology.

[6]  C. June,et al.  The Inducible Costimulator (ICOS) Is Critical for the Development of Human TH17 Cells , 2010, Science Translational Medicine.

[7]  M. Suematsu,et al.  Regulation of the HIF-1alpha level is essential for hematopoietic stem cells. , 2010, Cell stem cell.

[8]  Brigitta Stockinger,et al.  Effector T cell plasticity: flexibility in the face of changing circumstances , 2010, Nature Immunology.

[9]  W. Zou,et al.  TH17 cells in tumour immunity and immunotherapy , 2010, Nature Reviews Immunology.

[10]  P. Hwu,et al.  T helper 17 cells promote cytotoxic T cell activation in tumor immunity. , 2009, Immunity.

[11]  Jonathan L. Linehan,et al.  Different routes of bacterial infection induce long-lived TH1 memory cells and short-lived TH-17 cells , 2009, Nature Immunology.

[12]  R. Sprengel,et al.  The death receptor CD95 activates adult neural stem cells for working memory formation and brain repair. , 2009, Cell stem cell.

[13]  M. Banerjee,et al.  Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. , 2009, Blood.

[14]  P. Muranski,et al.  Type 17 CD8+ T cells display enhanced antitumor immunity. , 2009, Blood.

[15]  P. Muranski,et al.  Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells , 2009, Nature Medicine.

[16]  Erin E. Murphy,et al.  Circulating and gut-resident human Th17 cells express CD161 and promote intestinal inflammation , 2009, The Journal of experimental medicine.

[17]  C. Elson,et al.  Late developmental plasticity in the T helper 17 lineage. , 2009, Immunity.

[18]  Yi-hong Wang,et al.  Th17 cells promote pancreatic inflammation but only induce diabetes efficiently in lymphopenic hosts after conversion into Th1 cells , 2009, European journal of immunology.

[19]  James T. Elder,et al.  Induction of IL-17+ T Cell Trafficking and Development by IFN-γ: Mechanism and Pathological Relevance in Psoriasis1 , 2008, The Journal of Immunology.

[20]  P. Muranski,et al.  Tumor-specific Th17-polarized cells eradicate large established melanoma. , 2008, Blood.

[21]  Kristian Helin,et al.  The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. , 2007, Genes & development.

[22]  Susan Morrissey,et al.  BUILDING ON SUCCESS , 2006 .

[23]  S. Rosenberg,et al.  Adoptive immunotherapy for cancer: building on success , 2006, Nature Reviews Immunology.

[24]  Gefeng Zhu,et al.  B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma , 2006, The Journal of experimental medicine.

[25]  C. Dong Diversification of T-helper-cell lineages: finding the family root of IL-17-producing cells , 2006, Nature Reviews Immunology.

[26]  R. J. Hocking,et al.  TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. , 2006, Immunity.

[27]  Jiang Zhu,et al.  Host-reactive CD8+ memory stem cells in graft-versus-host disease , 2005, Nature Medicine.

[28]  R. D. Hatton,et al.  Interleukin 17–producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages , 2005, Nature Immunology.

[29]  Lieve Moons,et al.  CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. , 2005, Cancer research.

[30]  A. Lindén,et al.  Interleukin-17 family members and inflammation. , 2004, Immunity.

[31]  George Coukos,et al.  Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival , 2004, Nature Medicine.

[32]  G. Semenza Targeting HIF-1 for cancer therapy , 2003, Nature Reviews Cancer.

[33]  T. Curiel,et al.  Blockade of B7-H1 improves myeloid dendritic cell–mediated antitumor immunity , 2003, Nature Medicine.

[34]  T. Schumacher,et al.  CD27 is required for generation and long-term maintenance of T cell immunity , 2000, Nature Immunology.

[35]  J. Altman,et al.  Cutting Edge: Increased Expression of Bcl-2 in Antigen-Specific Memory CD8+ T Cells1 , 2000, The Journal of Immunology.

[36]  R. Herberman,et al.  Adoptive Immunotherapy of Cancer , 1994, Clinical Immunotherapeutics.

[37]  N. Suárez Comparative Leaf Anatomy and Pressure-Volume Analysis in Plants of Ipomoea pes-caprae Experimenting Saline and/or Drought Stress , 2011 .

[38]  R. J. Hocking,et al.  TGFb in the Context of an Inflammatory Cytokine Milieu Supports De Novo Differentiation of IL-17-Producing T Cells , 2022 .