Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 525: 84-96, 2022 01 28.
Article in English | MEDLINE | ID: mdl-34740608

ABSTRACT

Wnt/ß-catenin signaling is a highly conserved pathway that regulates cell proliferation, differentiation, apoptosis, stem cell self-renewal, tissue homeostasis, and wound healing. Dysregulation of the Wnt pathway is intricately involved in almost all stages of tumorigenesis in various cancers. Through direct and/or indirect effects on effector T cells, T-regulatory cells, T-helper cells, dendritic cells, and other cytokine-expressing immune cells, abnormal activation of Wnt/ß-catenin signaling benefits immune exclusion and hinders T-cell-mediated antitumor immune responses. Activation of Wnt signaling results in increased resistance to immunotherapies. In this review, we summarize the process by which Wnt signaling affects cancer and immune surveillance, and the potential for targeting the Wnt-signaling pathway via cancer immunotherapy.


Subject(s)
Carcinogenesis/genetics , Immunotherapy , Neoplasms/immunology , Tumor Microenvironment/immunology , Carcinogenesis/immunology , Cell Proliferation/genetics , Cell Self Renewal/immunology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/therapy , Th1 Cells/immunology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/immunology
2.
Immunity ; 54(6): 1200-1218.e9, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33951416

ABSTRACT

Tissue macrophages self-renew during homeostasis and produce inflammatory mediators upon microbial infection. We examined the relationship between proliferative and inflammatory properties of tissue macrophages by defining the impact of the Wnt/ß-catenin pathway, a central regulator of self-renewal, in alveolar macrophages (AMs). Activation of ß-catenin by Wnt ligand inhibited AM proliferation and stemness, but promoted inflammatory activity. In a murine influenza viral pneumonia model, ß-catenin-mediated AM inflammatory activity promoted acute host morbidity; in contrast, AM proliferation enabled repopulation of reparative AMs and tissue recovery following viral clearance. Mechanistically, Wnt treatment promoted ß-catenin-HIF-1α interaction and glycolysis-dependent inflammation while suppressing mitochondrial metabolism and thereby, AM proliferation. Differential HIF-1α activities distinguished proliferative and inflammatory AMs in vivo. This ß-catenin-HIF-1α axis was conserved in human AMs and enhanced HIF-1α expression associated with macrophage inflammation in COVID-19 patients. Thus, inflammatory and reparative activities of lung macrophages are regulated by ß-catenin-HIF-1α signaling, with implications for the treatment of severe respiratory diseases.


Subject(s)
COVID-19/immunology , COVID-19/virology , Cell Self Renewal/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , SARS-CoV-2/immunology , Biomarkers , COVID-19/metabolism , Cytokines/metabolism , Disease Susceptibility/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Signal Transduction
3.
Nat Immunol ; 22(6): 723-734, 2021 06.
Article in English | MEDLINE | ID: mdl-33958784

ABSTRACT

Continuous supply of immune cells throughout life relies on the delicate balance in the hematopoietic stem cell (HSC) pool between long-term maintenance and meeting the demands of both normal blood production and unexpected stress conditions. Here we identified distinct subsets of human long-term (LT)-HSCs that responded differently to regeneration-mediated stress: an immune checkpoint ligand CD112lo subset that exhibited a transient engraftment restraint (termed latency) before contributing to hematopoietic reconstitution and a primed CD112hi subset that responded rapidly. This functional heterogeneity and CD112 expression are regulated by INKA1 through direct interaction with PAK4 and SIRT1, inducing epigenetic changes and defining an alternative state of LT-HSC quiescence that serves to preserve self-renewal and regenerative capacity upon regeneration-mediated stress. Collectively, our data uncovered the molecular intricacies underlying HSC heterogeneity and self-renewal regulation and point to latency as an orchestrated physiological response that balances blood cell demands with preserving a stem cell reservoir.


Subject(s)
Cell Self Renewal/immunology , Hematopoietic Stem Cells/physiology , Immune Reconstitution , Multipotent Stem Cells/physiology , Stress, Physiological/immunology , Adult , Animals , Cell Self Renewal/genetics , Cells, Cultured , Epigenesis, Genetic/immunology , Female , Fetal Blood/cytology , Flow Cytometry , Gene Knockdown Techniques , Hematopoiesis , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunomagnetic Separation , Infant, Newborn , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Nectins/metabolism , Primary Cell Culture , RNA-Seq , Single-Cell Analysis , Sirtuin 1/metabolism , Stress, Physiological/genetics , Transplantation, Heterologous , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
5.
Theranostics ; 10(26): 11950-11962, 2020.
Article in English | MEDLINE | ID: mdl-33204322

ABSTRACT

Rationale: Mesenchymal stem cells (MSCs) have been the focus of many studies because of their abilities to modulate immune responses, angiogenesis, and promote tumor growth and metastasis. Our previous work showed that gastric cancer MSCs (GCMSCs) promoted immune escape by secreting of IL-8, which induced programmed cell death ligand 1 (PD-L1) expression in GC cells. Mounting evidence has revealed that PD-L1 expression is related to intrinsic tumor cell properties. Here, we investigated whether GCMSCs maintained a pool of cancer stem cells (CSCs) through PD-L1 signaling and the specific underlying molecular mechanism. Methods: Stem cell surface markers, aldehyde dehydrogenase (ALDH) activity, migration and sphere formation abilities were tested to evaluate the stemness of GC cells. PD-L1-expressing lentivirus and PD-L1 specific siRNA were used to analyze the effects of PD-L1 on GC cells stemness. Annexin V/PI double staining was used to assess apoptosis of GC cells induced by chemotherapy. Co-Immunoprecipitation (Co-IP) and Mass spectrometry were employed to determine the PD-L1 binding partner in GC cells. PD-L1Negative and PD-L1Positive cells were sorted by flow cytometry and used for limiting dilution assays to verify the effect of PD-L1 on tumorigenic ability in GC cells. Results: The results showed that GCMSCs enhanced the CSC-like properties of GC cells through PD-L1, which led to the resistance of GC cells to chemotherapy. PD-L1 associated with CTCF to contribute to the stemness and self-renewal of GC cells. In vivo, PD-L1Positive GC cells had greater stemness potential and tumorigenicity than PD-L1Negative GC cells. The results also indicated that GC cells were heterogeneous, and that PD-L1 in GC cells had different reactivity to GCMSCs. Conclusions: Overall, our data indicated that GCMSCs enriched CSC-like cells in GC cells, which gives a new insight into the mechanism of GCMSCs prompting GC progression and provides a potential combined therapeutic target.


Subject(s)
B7-H1 Antigen/metabolism , CCCTC-Binding Factor/metabolism , Mesenchymal Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Stomach Neoplasms/immunology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Self Renewal/immunology , Culture Media, Conditioned/metabolism , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Gastric Mucosa/cytology , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic/immunology , Gene Knockdown Techniques , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Male , Mice , Neoplastic Stem Cells/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
J Invest Dermatol ; 140(12): 2433-2441.e5, 2020 12.
Article in English | MEDLINE | ID: mdl-32311397

ABSTRACT

Mast cells (MCs) are tissue-resident hematopoietic cells intensely studied for their role as effectors in allergic immune responses. Yolk sac-derived embryonic MCs first populate tissues and are later replaced by definitive MCs. We show that definitive MC progenitors expand locally in skin and form clonal colonies that cover stable territories. In MC-deficient skin, colonies grow by proliferation of MCs at the border of the clonal territory. Clonal growth ceases at common borders of neighboring colonies. In steady state, colony self-renewal is independent of bone marrow contribution, and the clonal architecture remains fixed if not disturbed by skin inflammation. Inflammatory cues increase MC density setpoint, stimulating the influx of new progenitors from the bone marrow as well as proliferation of skin-resident cells. The expanding new arrivals disrespect territories of preexisting MC clones. We conclude that during a limited window early in development, definitive MC precursors efficiently enter the skin, expand, and self-maintain, occupying stable territories. In adulthood, circulating progenitors, excluded from steady-state skin, are recruited only into inflamed skin where they clonally expand alongside proliferating skin-resident MCs, disorganizing the original architecture of clonal territories.


Subject(s)
Adult Stem Cells/physiology , Cell Self Renewal/immunology , Dermatitis/immunology , Mast Cells/immunology , Skin/pathology , Animals , Bone Marrow , Cells, Cultured , Dermatitis/pathology , Disease Models, Animal , Embryo, Mammalian , Embryonic Stem Cells/physiology , Female , Humans , Male , Mice , Mice, Transgenic , Morula/cytology , Skin/cytology , Skin/immunology , Tetradecanoylphorbol Acetate/immunology
7.
Cancer Discov ; 10(3): 371-381, 2020 03.
Article in English | MEDLINE | ID: mdl-31919052

ABSTRACT

Glioblastoma (GBM) is a lethal brain tumor containing a subpopulation of glioma stem cells (GSC). Pan-cancer analyses have revealed that stemness of cancer cells correlates positively with immunosuppressive pathways in many solid tumors, including GBM, prompting us to conduct a gain-of-function screen of epigenetic regulators that may influence GSC self-renewal and tumor immunity. The circadian regulator CLOCK emerged as a top hit in enhancing stem-cell self-renewal, which was amplified in about 5% of human GBM cases. CLOCK and its heterodimeric partner BMAL1 enhanced GSC self-renewal and triggered protumor immunity via transcriptional upregulation of OLFML3, a novel chemokine recruiting immune-suppressive microglia into the tumor microenvironment. In GBM models, CLOCK or OLFML3 depletion reduced intratumoral microglia density and extended overall survival. We conclude that the CLOCK-BMAL1 complex contributes to key GBM hallmarks of GSC maintenance and immunosuppression and, together with its downstream target OLFML3, represents new therapeutic targets for this disease. SIGNIFICANCE: Circadian regulator CLOCK drives GSC self-renewal and metabolism and promotes microglia infiltration through direct regulation of a novel microglia-attracting chemokine, OLFML3. CLOCK and/or OLFML3 may represent novel therapeutic targets for GBM.This article is highlighted in the In This Issue feature, p. 327.


Subject(s)
ARNTL Transcription Factors/genetics , CLOCK Proteins/genetics , Glioblastoma/genetics , Glycoproteins/genetics , Animals , Cell Line, Tumor , Cell Self Renewal/genetics , Cell Self Renewal/immunology , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/therapy , Heterografts , Humans , Immunity, Cellular/immunology , Mice , Microglia/immunology , Microglia/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
8.
Int J Cancer ; 146(2): 424-438, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31241171

ABSTRACT

Stem cell chemoresistance remains challenging the efficacy of the front-line temozolomide against glioblastoma. Novel therapies are urgently needed to fight those cells in order to control tumor relapse. Here, we report that anti-O-acetyl-GD2 adjuvant immunotherapy controls glioma stem-like cell-driven chemoresistance. Using patient-derived glioblastoma cells, we found that glioma stem-like cells overexpressed O-acetyl-GD2. As a result, monoclonal antibody 8B6 immunotherapy significantly increased temozolomide genotoxicity and tumor cell death in vitro by enhancing temozolomide tumor uptake. Furthermore, the combination therapy decreased the expression of the glioma stem-like cell markers CD133 and Nestin and compromised glioma stem-like cell self-renewal capabilities. When tested in vivo, adjuvant 8B6 immunotherapy prevented the extension of the temozolomide-resistant glioma stem-like cell pool within the tumor bulk in vivo and was more effective than the single agent therapies. This is the first report demonstrating that anti-O-acetyl-GD2 monoclonal antibody 8B6 targets glioblastoma in a manner that control temozolomide-resistance driven by glioma stem-like cells. Together our results offer a proof of concept for using anti-O-acetyl GD2 reagents in glioblastoma to develop more efficient combination therapies for malignant gliomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gangliosides/antagonists & inhibitors , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Self Renewal/drug effects , Cell Self Renewal/immunology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Drug Synergism , Gangliosides/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/immunology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
9.
Stem Cells Dev ; 29(6): 327-335, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31856674

ABSTRACT

Head and neck squamous cell carcinomas (HNSCCs) are malignancies that originate in the mucosal lining of the upper aerodigestive tract. Despite advances in therapeutic interventions, survival rates among HNSCC patients have remained static for years. Cancer stem cells (CSCs) are tumor-initiating cells that are highly resistant to treatment, and are hypothesized to contribute to a significant fraction of tumor recurrences. Consequently, further investigations of how CSCs mediate recurrence may provide insights into novel druggable targets. A key element of recurrence involves the tumor's ability to evade immunosurveillance. Recent published reports suggest that CSCs possess immunosuppressive properties, however, the underlying mechanism have yet to be fully elucidated. To date, most groups have focused on the role of CSC-derived secretory proteins, such as cytokines and growth factors. Here, we review the established immunoregulatory role of exosomes derived from mixed tumor cell populations, and propose further study of CSC-derived exosomes may be warranted. Such studies may yield novel insights into new druggable targets, or lay the foundation for future exosome-based diagnostics.


Subject(s)
Carcinoma, Squamous Cell/immunology , Exosomes/immunology , Head and Neck Neoplasms/immunology , Neoplastic Stem Cells/immunology , Carcinoma, Squamous Cell/pathology , Cell Differentiation/immunology , Cell Self Renewal/immunology , Head and Neck Neoplasms/pathology , Humans , Neoplasm Recurrence, Local , Signal Transduction/immunology , Tumor Microenvironment/immunology
10.
J Exp Med ; 216(8): 1791-1808, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31164392

ABSTRACT

Chronic infection and cancer are associated with suppressed T cell responses in the presence of cognate antigen. Recent work identified memory-like CXCR5+ TCF1+ CD8+ T cells that sustain T cell responses during persistent infection and proliferate upon anti-PD1 treatment. Approaches to expand these cells are sought. We show that blockade of interferon type 1 (IFN-I) receptor leads to CXCR5+ CD8+ T cell expansion in an IL-27- and STAT1-dependent manner. IFNAR1 blockade promoted accelerated cell division and retention of TCF1 in virus-specific CD8+ T cells. We found that CD8+ T cell-intrinsic IL-27 signaling safeguards the ability of TCF1hi cells to maintain proliferation and avoid terminal differentiation or programmed cell death. Mechanistically, IL-27 endowed rapidly dividing cells with IRF1, a transcription factor that was required for sustained division in a cell-intrinsic manner. These findings reveal that IL-27 opposes IFN-I to uncouple effector differentiation from cell division and suggest that IL-27 signaling could be exploited to augment self-renewing T cells in chronic infections and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Self Renewal/immunology , Interleukins/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunologic Memory , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interleukins/genetics , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptors, CXCR5/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Transcriptome
11.
Nat Immunol ; 20(2): 183-194, 2019 02.
Article in English | MEDLINE | ID: mdl-30643264

ABSTRACT

Intestinal stem cells (ISCs) are maintained by stemness signaling for precise modulation of self-renewal and differentiation under homeostasis. However, the way in which intestinal immune cells regulate the self-renewal of ISCs remains elusive. Here we found that mouse and human Lgr5+ ISCs showed high expression of the immune cell-associated circular RNA circPan3 (originating from the Pan3 gene transcript). Deletion of circPan3 in Lgr5+ ISCs impaired their self-renewal capacity and the regeneration of gut epithelium in a manner dependent on immune cells. circPan3 bound mRNA encoding the cytokine IL-13 receptor subunit IL-13Rα1 (Il13ra1) in ISCs to increase its stability, which led to the expression of IL-13Rα1 in ISCs. IL-13 produced by group 2 innate lymphoid cells in the crypt niche engaged IL-13Rα1 on crypt ISCs and activated signaling mediated by IL-13‒IL-13R, which in turn initiated expression of the transcription factor Foxp1. Foxp1 is associated with ß-catenin in rendering its nuclear translocation, which caused activation of the ß-catenin pathway and the maintenance of Lgr5+ ISCs.


Subject(s)
Cell Self Renewal/immunology , Interleukin-13/metabolism , Intestinal Mucosa/immunology , RNA/metabolism , Stem Cells/physiology , Animals , Carrier Proteins/genetics , Cell Differentiation/immunology , Cell Self Renewal/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Interleukin-13/immunology , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-13 Receptor alpha1 Subunit/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , RNA/genetics , RNA/immunology , RNA, Circular , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration/genetics , Regeneration/immunology , Signal Transduction/genetics , Signal Transduction/immunology , beta Catenin/immunology , beta Catenin/metabolism
12.
Arthritis Rheumatol ; 71(1): 133-142, 2019 01.
Article in English | MEDLINE | ID: mdl-29984480

ABSTRACT

OBJECTIVE: A major characteristic of the autoimmune disease primary Sjögren's syndrome (SS) is salivary gland (SG) hypofunction. The inability of resident SG stem cells (SGSCs) to maintain homeostasis and saliva production has never been explained and limits our comprehension of mechanisms underlying primary SS. The present study was undertaken to investigate the role of salivary gland stem cells in hyposalivation in primary SS. METHODS: SGSCs were isolated from parotid biopsy samples from controls and patients classified as having primary SS or incomplete primary SS, according to the American College of Rheumatology/European League Against Rheumatism criteria. Self-renewal and differentiation assays were used to determine SGSC regenerative potential, RNA was extracted for sequencing analysis, single telomere length analysis was conducted to determine telomere length, and frozen tissue samples were used for immunohistochemical analysis. RESULTS: SGSCs isolated from primary SS parotid gland biopsy samples were regeneratively inferior to healthy control specimens. We demonstrated that SGSCs from samples from patients with primary SS are not only lower in number and less able to differentiate, but are likely to be senescent, as revealed by telomere length analysis, RNA sequencing, and immunostaining. We further found that SGSCs exposed to primary SS-associated proinflammatory cytokines we induced to proliferate, express senescence-associated genes, and subsequently differentiate into intercalated duct cells. We also localized p16+ senescent cells to the intercalated ducts in primary SS SG tissue, suggesting a block in SGSC differentiation into acinar cells. CONCLUSION: This study represents the first characterization of SGSCs in primary SS, and also the first demonstration of a linkage between an autoimmune disease and a parenchymal premature-aging phenotype. The knowledge garnered in this study indicates that disease-modifying antirheumatic drugs used to treat primary SS are not likely to restore saliva production, and should be supplemented with fresh SGSCs to recover saliva production.


Subject(s)
Cell Self Renewal/immunology , Cellular Senescence/immunology , Parotid Gland/immunology , Sjogren's Syndrome/immunology , Stem Cells/immunology , Case-Control Studies , Cell Self Renewal/genetics , Cellular Senescence/genetics , Cytokines/immunology , Humans , Immunohistochemistry , Parotid Gland/cytology , Parotid Gland/metabolism , Salivary Glands , Sequence Analysis, RNA , Stem Cells/metabolism , Telomere/metabolism
13.
Biomed Pharmacother ; 107: 1318-1325, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30257347

ABSTRACT

Colorectal carcinoma (CRC) is the third most common malignant tumor in the world. In recent years, the morbidity and mortality of CRC have increased in the world due to increasingly ageing population, modern dietary habits, environmental change, genetic disorders and chronic intestinal inflammation. Despite recent advances in earlier detection and improvements in chemotherapy, the 5-year survival rate of patients with metastatic CRC remains low. Therefore, novel effective treatment strategies for primary or metastatic CRC have emerged to enhance cure rate as well as elongation of patient's survival. Immunotherapy has been proposed for a potentially effective therapeutic approach to the treatment of CRC. Tumor vaccination in preclinical and clinical studies has supported the antitumor activity induced by immunization with CRC cell vaccines. Epithelial cell molecule Mucin 1 (MUC1), a transmembrane glycoprotein aberrantly overexpressed in various cancers including CRC, has been used as a candidate target antigen in the peptide, dendritic cell, and whole tumor vaccines. Several clinical trials in progress reveal the immunogenicity and suitability of MUC1 that acted as immunotherapeutic vaccines for CRC/colorectal cancer stem cells (CCSC). The present review summarizes the potential roles of MUC1 on CRC/CCSC vaccines according to the latest data. Moreover, this review also discusses the novel strategies for targeting CCSC via inducing an immune response against MUC1 to achieve the best prevention and treatment effects in animal models and clinical trails.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Colorectal Neoplasms/metabolism , Immunotherapy/methods , Mucin-1/metabolism , Cell Proliferation , Cell Self Renewal/immunology , Clinical Trials as Topic , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Humans , Mucin-1/immunology , Survival Rate
14.
Mamm Genome ; 29(11-12): 771-776, 2018 12.
Article in English | MEDLINE | ID: mdl-30182300

ABSTRACT

Emerging evidence indicates that thymocyte self-renewal induced by progenitor deprivation carries an oncogenic risk that is modulated by intra-thymic competition from differentiation-committed cells. Here we discuss formative studies demonstrating that, in mice, early thymocytes acquire self-renewing potential when thymic progenitor supply is sub-physiological and the importance of cellular competition with this at-risk cell population to prevent lymphoid malignancy. We also consider the possibility that increased thymic residency time, established under conditions of limited cellular competition, may have contributed to oncogenesis observed in early SCID-X1 trials when combined with insertional activation of proto-oncogenes such as LMO2.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinogenesis/genetics , LIM Domain Proteins/genetics , Neoplasms/immunology , Thymocytes/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Carcinogenesis/immunology , Cell Self Renewal/immunology , Cell Transformation, Neoplastic/immunology , Disease Models, Animal , Genetic Therapy , Hematopoietic Stem Cells/immunology , Humans , LIM Domain Proteins/immunology , Mice , Neoplasms/genetics , Neoplastic Stem Cells/immunology
15.
Front Immunol ; 9: 1064, 2018.
Article in English | MEDLINE | ID: mdl-29868023

ABSTRACT

Studies performed in animal models and in humans indicate that the innate arm of the immune system provides an essential role in the initial protection against potential insults and in maintaining tolerance to self-antigens. In the B cell compartment, several subsets engage in both adaptive and innate functions. Whereas B cell subsets are recognized to play important roles in autoimmune diseases, understanding the intricacies of their effector functions remains challenging. In addition to B-1a cells and marginal zone B cells, the B cell compartment comprises other B cells with innate-like functions, including innate response activator B cells, T-bet positive B cells, natural killer-like B cells, IL-17-producing B cells, and human self-reactive VH4-34-expressing B cells. Herein, we summarize the functions of recently described B cell populations that can exert innate-like roles in both animal models and humans. We also highlight the importance of the cross talk between innate-like B cells and other adaptive and innate branches of the immune system in various autoimmune and inflammatory diseases. In as much as innate immunity seems to be important in resolving inflammation, it is possible that targeting certain innate-like B cell subsets could represent a novel therapeutic approach for inducing resolution of inflammation of autoimmune and inflammatory responses.


Subject(s)
Autoimmunity , B-Lymphocyte Subsets/immunology , Cell Communication/immunology , Immunity, Innate , Animals , Antibodies/immunology , Antibody-Dependent Cell Cytotoxicity , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Cell Self Renewal/immunology , Chemotaxis, Leukocyte/immunology , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation , Signal Transduction , T-Box Domain Proteins/metabolism
16.
PLoS Biol ; 16(6): e2005523, 2018 06.
Article in English | MEDLINE | ID: mdl-29933397

ABSTRACT

Adaptive immunity relies on the generation and maintenance of memory T cells to provide protection against repeated antigen exposure. It has been hypothesised that a self-renewing population of T cells, named stem cell-like memory T (TSCM) cells, are responsible for maintaining memory. However, it is not clear if the dynamics of TSCM cells in vivo are compatible with this hypothesis. To address this issue, we investigated the dynamics of TSCM cells under physiological conditions in humans in vivo using a multidisciplinary approach that combines mathematical modelling, stable isotope labelling, telomere length analysis, and cross-sectional data from vaccine recipients. We show that, unexpectedly, the average longevity of a TSCM clone is very short (half-life < 1 year, degree of self-renewal = 430 days): far too short to constitute a stem cell population. However, we also find that the TSCM population is comprised of at least 2 kinetically distinct subpopulations that turn over at different rates. Whilst one subpopulation is rapidly replaced (half-life = 5 months) and explains the rapid average turnover of the bulk TSCM population, the half-life of the other TSCM subpopulation is approximately 9 years, consistent with the longevity of the recall response. We also show that this latter population exhibited a high degree of self-renewal, with a cell residing without dying or differentiating for 15% of our lifetime. Finally, although small, the population was not subject to excessive stochasticity. We conclude that the majority of TSCM cells are not stem cell-like but that there is a subpopulation of TSCM cells whose dynamics are compatible with their putative role in the maintenance of T cell memory.


Subject(s)
Cell Self Renewal/immunology , Immunologic Memory , T-Lymphocyte Subsets/immunology , Adult , Aged, 80 and over , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Humans , Kinetics , Mathematical Concepts , Middle Aged , Models, Immunological , T-Lymphocyte Subsets/cytology , Telomere Homeostasis/immunology , Yellow fever virus/immunology
17.
Immunity ; 48(4): 730-744.e5, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669251

ABSTRACT

Although characterization of T cell exhaustion has unlocked powerful immunotherapies, the mechanisms sustaining adaptations of short-lived innate cells to chronic inflammatory settings remain unknown. During murine chronic viral infection, we found that concerted events in bone marrow and spleen mediated by type I interferon (IFN-I) and Toll-like receptor 7 (TLR7) maintained a pool of functionally exhausted plasmacytoid dendritic cells (pDCs). In the bone marrow, IFN-I compromised the number and the developmental capacity of pDC progenitors, which generated dysfunctional pDCs. Concurrently, exhausted pDCs in the periphery were maintained by self-renewal via IFN-I- and TLR7-induced proliferation of CD4- subsets. On the other hand, pDC functional loss was mediated by TLR7, leading to compromised IFN-I production and resistance to secondary infection. These findings unveil the mechanisms sustaining a self-perpetuating pool of functionally exhausted pDCs and provide a framework for deciphering long-term exhaustion of other short-lived innate cells during chronic inflammation.


Subject(s)
Cell Self Renewal/immunology , Dendritic Cells/immunology , Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 7/immunology , 3T3 Cells , Animals , Carrier Proteins/biosynthesis , Cell Line , Cell Proliferation , DNA-Binding Proteins/biosynthesis , Dendritic Cells/cytology , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/biosynthesis , Repressor Proteins , Signal Transduction/immunology , Transcription Factor 4/biosynthesis , Transcription Factors/biosynthesis
18.
Cell Stem Cell ; 20(4): 450-461.e4, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28089910

ABSTRACT

Tumors contain hostile inflammatory signals generated by aberrant proliferation, necrosis, and hypoxia. These signals are sensed and acted upon acutely by the Toll-like receptors (TLRs) to halt proliferation and activate an immune response. Despite the presence of TLR ligands within the microenvironment, tumors progress, and the mechanisms that permit this growth remain largely unknown. We report that self-renewing cancer stem cells (CSCs) in glioblastoma have low TLR4 expression that allows them to survive by disregarding inflammatory signals. Non-CSCs express high levels of TLR4 and respond to ligands. TLR4 signaling suppresses CSC properties by reducing retinoblastoma binding protein 5 (RBBP5), which is elevated in CSCs. RBBP5 activates core stem cell transcription factors, is necessary and sufficient for self-renewal, and is suppressed by TLR4 overexpression in CSCs. Our findings provide a mechanism through which CSCs persist in hostile environments because of an inability to respond to inflammatory signals.


Subject(s)
Cell Self Renewal/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Immune Evasion , Immunity, Innate , Neoplastic Stem Cells/pathology , Toll-Like Receptor 4/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins , Female , Humans , Mice , Models, Biological , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
19.
Nat Med ; 23(1): 18-27, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28060797

ABSTRACT

T memory stem (TSCM) cells are a rare subset of memory lymphocytes endowed with the stem cell-like ability to self-renew and the multipotent capacity to reconstitute the entire spectrum of memory and effector T cell subsets. Cumulative evidence in mice, nonhuman primates and humans indicates that TSCM cells are minimally differentiated cells at the apex of the hierarchical system of memory T lymphocytes. Here we describe emerging findings demonstrating that TSCM cells, owing to their extreme longevity and robust potential for immune reconstitution, are central players in many physiological and pathological human processes. We also discuss how TSCM cell stemness could be leveraged therapeutically to enhance the efficacy of vaccines and adoptive T cell therapies for cancer and infectious diseases or, conversely, how it could be disrupted to treat TSCM cell driven and sustained diseases, such as autoimmunity, adult T cell leukemia and HIV-1.


Subject(s)
Cell Self Renewal/immunology , Immunologic Memory/immunology , Stem Cells/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cell Differentiation , HIV Infections/immunology , HIV Infections/therapy , Humans , Leukemia, T-Cell/immunology , Leukemia, T-Cell/therapy , T-Lymphocytes/transplantation
20.
Clin Cancer Res ; 23(10): 2516-2527, 2017 May 15.
Article in English | MEDLINE | ID: mdl-27780858

ABSTRACT

Purpose: Locoregional recurrence is a frequent treatment outcome for patients with advanced head and neck squamous cell carcinoma (HNSCC). Emerging evidence suggests that tumor recurrence is mediated by a small subpopulation of uniquely tumorigenic cells, that is, cancer stem cells (CSC), that are resistant to conventional chemotherapy, endowed with self-renewal and multipotency.Experimental Design: Here, we evaluated the efficacy of MEDI0641, a novel antibody-drug conjugate targeted to 5T4 and carrying a DNA-damaging "payload" (pyrrolobenzodiazepine) in preclinical models of HNSCC.Results: Analysis of a tissue microarray containing 77 HNSCC with follow-up of up to 12 years revealed that patients with 5T4high tumors displayed lower overall survival than those with 5T4low tumors (P = 0.038). 5T4 is more highly expressed in head and neck CSC (ALDHhighCD44high) than in control cells (non-CSC). Treatment with MEDI0641 caused a significant reduction in the CSC fraction in HNSCC cells (UM-SCC-11B, UM-SCC-22B) in vitro Notably, a single intravenous dose of 1 mg/kg MEDI0641 caused long-lasting tumor regression in three patient-derived xenograft (PDX) models of HNSCC. MEDI0641 ablated CSC in the PDX-SCC-M0 model, reduced it by five-fold in the PDX-SCC-M1, and two-fold in the PDX-SCC-M11 model. Importantly, mice (n = 12) treated with neoadjuvant, single administration of MEDI0641 prior to surgical tumor removal showed no recurrence for more than 200 days, whereas the control group had 7 recurrences (in 12 mice; P = 0.0047).Conclusions: Collectively, these findings demonstrate that an anti-5T4 antibody-drug conjugate reduces the fraction of CSCs and prevents local recurrence and suggest a novel therapeutic approach for patients with HNSCC. Clin Cancer Res; 23(10); 2516-27. ©2016 AACR.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Immunoconjugates/administration & dosage , Membrane Glycoproteins/immunology , Animals , Benzodiazepines/administration & dosage , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Self Renewal/genetics , Cell Self Renewal/immunology , DNA Damage/drug effects , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/pathology , Humans , Immunoconjugates/immunology , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Pyrroles/administration & dosage , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...