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










Publication year range
1.
Nat Immunol ; 25(2): 268-281, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38195702

ABSTRACT

Melanoma cells, deriving from neuroectodermal melanocytes, may exploit the nervous system's immune privilege for growth. Here we show that nerve growth factor (NGF) has both melanoma cell intrinsic and extrinsic immunosuppressive functions. Autocrine NGF engages tropomyosin receptor kinase A (TrkA) on melanoma cells to desensitize interferon γ signaling, leading to T and natural killer cell exclusion. In effector T cells that upregulate surface TrkA expression upon T cell receptor activation, paracrine NGF dampens T cell receptor signaling and effector function. Inhibiting NGF, either through genetic modification or with the tropomyosin receptor kinase inhibitor larotrectinib, renders melanomas susceptible to immune checkpoint blockade therapy and fosters long-term immunity by activating memory T cells with low affinity. These results identify the NGF-TrkA axis as an important suppressor of anti-tumor immunity and suggest larotrectinib might be repurposed for immune sensitization. Moreover, by enlisting low-affinity T cells, anti-NGF reduces acquired resistance to immune checkpoint blockade and prevents melanoma recurrence.


Subject(s)
Melanoma , Receptor, Nerve Growth Factor , Humans , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Tropomyosin , Melanoma/therapy , Receptor, trkA/genetics , Receptor, trkA/metabolism , Cytoprotection , Immune Checkpoint Inhibitors , Memory T Cells , Immunosuppression Therapy , Immunotherapy , Receptors, Antigen, T-Cell
2.
Adv Sci (Weinh) ; 11(2): e2303489, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37964763

ABSTRACT

The essential branched-chain amino acids (BCAAs) leucine, isoleucine, and valine play critical roles in protein synthesis and energy metabolism. Despite their widespread use as nutritional supplements, BCAAs' full effects on mammalian physiology remain uncertain due to the complexities of BCAA metabolic regulation. Here a novel mechanism linking intrinsic alterations in BCAA metabolism is identified to cellular senescence and the senescence-associated secretory phenotype (SASP), both of which contribute to organismal aging and inflammation-related diseases. Altered BCAA metabolism driving the SASP is mediated by robust activation of the BCAA transporters Solute Carrier Family 6 Members 14 and 15 as well as downregulation of the catabolic enzyme BCAA transaminase 1 during onset of cellular senescence, leading to highly elevated intracellular BCAA levels in senescent cells. This, in turn, activates the mammalian target of rapamycin complex 1 (mTORC1) to establish the full SASP program. Transgenic Drosophila models further indicate that orthologous BCAA regulators are involved in the induction of cellular senescence and age-related phenotypes in flies, suggesting evolutionary conservation of this metabolic pathway during aging. Finally, experimentally blocking BCAA accumulation attenuates the inflammatory response in a mouse senescence model, highlighting the therapeutic potential of modulating BCAA metabolism for the treatment of age-related and inflammatory diseases.


Subject(s)
Amino Acids, Branched-Chain , Senescence-Associated Secretory Phenotype , Animals , Mice , Amino Acids, Branched-Chain/metabolism , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Energy Metabolism , Mammals/metabolism
3.
Cell Res ; 33(7): 516-532, 2023 07.
Article in English | MEDLINE | ID: mdl-37169907

ABSTRACT

Cellular senescence is a stress-induced, stable cell cycle arrest phenotype which generates a pro-inflammatory microenvironment, leading to chronic inflammation and age-associated diseases. Determining the fundamental molecular pathways driving senescence instead of apoptosis could enable the identification of senolytic agents to restore tissue homeostasis. Here, we identify thrombomodulin (THBD) signaling as a key molecular determinant of the senescent cell fate. Although normally restricted to endothelial cells, THBD is rapidly upregulated and maintained throughout all phases of the senescence program in aged mammalian tissues and in senescent cell models. Mechanistically, THBD activates a proteolytic feed-forward signaling pathway by stabilizing a multi-protein complex in early endosomes, thus forming a molecular basis for the irreversibility of the senescence program and ensuring senescent cell viability. Therapeutically, THBD signaling depletion or inhibition using vorapaxar, an FDA-approved drug, effectively ablates senescent cells and restores tissue homeostasis in liver fibrosis models. Collectively, these results uncover proteolytic THBD signaling as a conserved pro-survival pathway essential for senescent cell viability, thus providing a pharmacologically exploitable senolytic target for senescence-associated diseases.


Subject(s)
Endothelial Cells , Thrombomodulin , Animals , Cellular Senescence , Liver Cirrhosis/drug therapy , Signal Transduction , Apoptosis , Mammals
4.
Trends Cell Biol ; 33(5): 403-412, 2023 05.
Article in English | MEDLINE | ID: mdl-36114091

ABSTRACT

Traditionally, γ-aminobutyric acid (GABA) is best known for its role as a primary inhibitory neurotransmitter reducing neuronal excitability in the mammalian central nervous system (CNS), thereby producing calming effects. However, an emerging body of data now supports a function for GABA beyond neurotransmission as a potent factor regulating cancer cell growth and metastasis, as well as the antitumor immune response, by shaping the tumor microenvironment (TME). Here, we review the current knowledge on GABA's effects on the function of tumor cells, tumor-immune interactions, and the underlying molecular mechanisms. Since altered GABAergic signaling is now recognized as a feature of certain types of solid tumors, we also discuss the potential of repurposing existing GABAergic agents as a new class of anticancer therapy.


Subject(s)
Neoplasms , gamma-Aminobutyric Acid , Animals , Humans , gamma-Aminobutyric Acid/metabolism , Neurons/metabolism , Signal Transduction/physiology , Neurotransmitter Agents , Synapses/metabolism , Mammals/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism
5.
Cancer Cell ; 40(6): 674-693.e7, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35594863

ABSTRACT

Despite the unprecedented success of immune checkpoint inhibitors (ICIs) as anti-cancer therapy, it remains a prevailing clinical need to identify additional mechanisms underlying ICI therapeutic efficacy and potential drug resistance. Here, using lineage tracking in cancer patients and tumor-bearing mice, we demonstrate that erythroid progenitor cells lose their developmental potential and switch to the myeloid lineage. Single-cell transcriptome analyses reveal that, notwithstanding quantitative differences in erythroid gene expression, erythroid differentiated myeloid cells (EDMCs) are transcriptionally indistinguishable from their myeloid-originated counterparts. EDMCs possess multifaceted machinery to curtail T cell-mediated anti-tumor responses. Consequently, EDMC content within tumor tissues is negatively associated with T cell inflammation for the majority of solid cancers; moreover, EDMC enrichment, in accordance with anemia manifestation, is predictive of poor prognosis in various cohorts of patients undergoing ICI therapy. Together, our findings reveal a feedforward mechanism by which tumors exploit anemia-triggered erythropoiesis for myeloid transdifferentiation and immunosuppression.


Subject(s)
Anemia , Neoplasms , Anemia/genetics , Anemia/metabolism , Animals , B7-H1 Antigen/metabolism , Erythroid Precursor Cells , Humans , Immunosuppression Therapy , Mice , Myeloid Cells/metabolism , Neoplasms/therapy , Treatment Outcome , Tumor Microenvironment
6.
Nat Cell Biol ; 24(2): 230-241, 2022 02.
Article in English | MEDLINE | ID: mdl-35145222

ABSTRACT

Many cancers have an unusual dependence on glutamine. However, most previous studies have focused on the contribution of glutamine to metabolic building blocks and the energy supply. Here, we report that cancer cells with aberrant expression of glutamate decarboxylase 1 (GAD1) rewire glutamine metabolism for the synthesis of γ-aminobutyric acid (GABA)-a prominent neurotransmitter-in non-nervous tissues. An analysis of clinical samples reveals that increased GABA levels predict poor prognosis. Mechanistically, we identify a cancer-intrinsic pathway through which GABA activates the GABAB receptor to inhibit GSK-3ß activity, leading to enhanced ß-catenin signalling. This GABA-mediated ß-catenin activation both stimulates tumour cell proliferation and suppresses CD8+ T cell intratumoural infiltration, such that targeting GAD1 or GABABR in mouse models overcomes resistance to anti-PD-1 immune checkpoint blockade therapy. Our findings uncover a signalling role for tumour-derived GABA beyond its classic function as a neurotransmitter that can be targeted pharmacologically to reverse immunosuppression.


Subject(s)
Cell Proliferation , Neoplasms/metabolism , Tumor Escape , Tumor Microenvironment/immunology , beta Catenin/metabolism , gamma-Aminobutyric Acid/metabolism , A549 Cells , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Mice, Nude , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Receptors, GABA-B/metabolism , Tumor Burden , Tumor Escape/drug effects , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/genetics
7.
Oncoimmunology ; 10(1): 1955545, 2021.
Article in English | MEDLINE | ID: mdl-34377592

ABSTRACT

The past decade has witnessed the gradual and steady progress of adoptive T cell therapy in treating various types of cancer. In combination with gemcitabine and carboplatin chemotherapy, we previously conducted a clinical trial, NCT00690872, to treat Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) patients with autologous EBV-expanded cytotoxic T lymphocytes (CTLs). While achieving a 2-year overall survival rate of 62.9%, this trial failed to induce an anti-tumor response in a sizable fraction of patients. Thus, the identification of benchmarks capable of evaluating CTL products and predicting clinical immunotherapeutic efficacy remains an urgent need. We conducted T cell receptor (TCR) repertoire sequencing to assess EBV-expanded infusion-ready CTL products. To depict the overall repertoire landscape, we evaluated the individual repertoire diversity by Shannon entropy, and, compared the inter-patient CDR3 similarity to estimate T cells expanded by common antigens. With a recently developed bioinformatics algorithm, termed Motif Analysis, we made a machine-learning prediction of structural regions within the CDR3 of TCRß that associate with CTL therapy prognosis. We found that long term survivors, defined as patients surviving longer than two years, had a higher CTL repertoire diversity with reduced inter-patient similarity. Furthermore, TCR Motif Analysis identified 11 structural motifs distinguishing long term survivors from short term survivors. Specifically, two motifs with a high area under the curve (AUC) values were identified as potential predictive benchmarks for efficacious CTL production. Together, these results reveal that the presence of diverse TCR sequences containing a common core motif set is associated with a favorable response to CTL immunotherapy against EBV-positive NPC.


Subject(s)
Epstein-Barr Virus Infections , Nasopharyngeal Neoplasms , Herpesvirus 4, Human/genetics , Humans , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic
8.
Theranostics ; 11(10): 4699-4709, 2021.
Article in English | MEDLINE | ID: mdl-33754022

ABSTRACT

Rationale: The onset of cytokine release syndrome (CRS) and in vivo persistence of anti-CD19 chimeric antigen receptor T (CAR-T) cells after infusion correlate with clinical responsiveness. However, there are no known baseline biomarkers that can predict the prognosis of patients with B-lineage non-Hodgkin lymphoma (B-NHL). The aim of this study was to identify blood cell populations associated with beneficial outcomes in B-NHL patients administered CAR-T cell immunotherapies. Methods: We enumerated peripheral blood and CAR-T cells by retrospectively analyzing three CAR-T cell trials involving 65 B-NHL patients. We used a preclinical model to elucidate the eosinophil mechanism in CAR-T cell therapy. Results: During an observation period up to 30 mo, B-NHL patients with higher baseline eosinophil counts had higher objective response rates than those with low eosinophil counts. Higher baseline eosinophil counts were also significantly associated with durable progression-free survival (PFS). The predictive significance of baseline eosinophil counts was validated in two independent cohorts. A preclinical model showed that eosinophil depletion impairs the intratumoral infiltration of transferred CAR-T cells and reduces CAR-T cell antitumor efficacy. Conclusion: The results of this study suggest that peripheral eosinophils could serve as stratification biomarkers and a recruitment machinery to facilitate anti-CD19 CAR-T cell therapy in B-NHL patients.


Subject(s)
Eosinophils , Immunotherapy, Adoptive , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen , Adult , Aged , Animals , Antigens, CD19 , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Leukocyte Count , Lymphoma, B-Cell/blood , Male , Mice , Middle Aged , Prognosis , Progression-Free Survival , Young Adult
9.
Sci Adv ; 7(5)2021 01.
Article in English | MEDLINE | ID: mdl-33514544

ABSTRACT

The efficacy of cancer immunotherapy is dictated by CD8+ T cell infiltration and the nature of the tumor microenvironment (TME). By inflaming the TME to favor CD8+ T cell immunity, radiation is now widely considered as a neoadjuvant for immunomodulation. Here, we observed that local irradiation enhances the infiltration of intratumoral eosinophils, and depletion of eosinophil dampens CD8+ T cell infiltration and diminishes the anti-tumor effectiveness of radiation. Retrospectively, we identified a strong correlation between eosinophilia and survival benefit in radiation-treated cancer patients. Experimentally, we further show that radiation enhances the intratumoral infiltration of adoptive transferred T cells therapy, bolstering eosinophils by intravenous interleukin-5 administration promotes the efficacy of radiation-induced abscopal effect. Together, these results suggest that eosinophil mobilization can be considered as a mechanistically relevant biomarker for predicting the effectiveness of pre-immunotherapy radiation, as well as a new strategy to enhance T cell-mediated immunotherapy against cancers.

10.
Oncoimmunology ; 9(1): 1806009, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32923168

ABSTRACT

The tumoricidal efficiency of human CAR-T cells is generally evaluated using immune-deficient mouse models; however, due to their immune-incompetency and the species-specific reactivity of a target antigen, these models are problematic to imitate CAR-T-induced adverse effects in the clinic. Epithelial cell adhesion molecule (EpCAM) is a tumor-associated antigen overtly presented on the cell surface of various carcinomas, making it an attractive target for CAR-T therapy. Here, we developed an anti-mouse EpCAM CAR to evaluate its safety and efficacy in immunocompetent mouse models. As previously reported for their human equivalents, murine EpCAM CAR-T cells exhibit promising anti-tumor efficacy in vitro and in vivo. However, after CAR-T infusion, various dose-depended toxicities including body weight loss, cytokine-release syndrome (CRS), and death were observed in both tumor-bearing and tumor-free mice. Pathological examination revealed unexpected and severe pulmonary immunopathology due to basal EpCAM expression in normal lung. While our study validates EpCAM CAR-T's potent anti-tumor efficacy, it also reveals that EpCAM CAR-T cells used for the treatment of solid tumors may cause lethal toxicity and should, therefore, be evaluated in patients with caution.


Subject(s)
Receptors, Chimeric Antigen , Animals , Epithelial Cell Adhesion Molecule , Humans , Immunotherapy, Adoptive , Lung , Mice , T-Lymphocytes
11.
Oncoimmunology ; 9(1): 1749476, 2020.
Article in English | MEDLINE | ID: mdl-32313731

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults with a dismal prognosis. We previously reported that vaccination with heat shock protein peptide complex-96 (HSPPC-96) improves survival in patients with newly diagnosed GBM (NCT02122822). Especially for patients with a strong antitumor immune response after vaccination, a durable survival benefit can be achieved. Here, we conducted T cell receptor (TCR) sequencing to retrospectively examine the TCR repertoires of tumor-infiltrating lymphocytes in long-term survivors (LTS) and short-term survivors (STS). We found that LTS exhibit lower TCR repertoire diversity compared with STS, indicating the prevalence of dominant TCR clones in LTS tumors. Accordingly, the LTS group showed increased inter-patient similarity, especially among high-frequency TCR clones, implying some of these dominant clones are shared among LTS. Indeed, we discovered four TCR clones significantly enriched in the LTS group: the presence of these clones has predictive value for stratifying patients prior to vaccination. Together, these findings uncover a group of preexisting TCR clones shared in LTS that can be utilized as candidate biomarkers to select GBM patients most likely to durably respond to HSPPC-96 treatment.


Subject(s)
Brain Neoplasms , Cancer Vaccines , Glioblastoma , Heat-Shock Proteins , Receptors, Antigen, T-Cell , Adult , Aged , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Female , Glioblastoma/immunology , Glioblastoma/therapy , Heat-Shock Proteins/administration & dosage , Heat-Shock Proteins/adverse effects , Humans , Male , Middle Aged , Prognosis , Receptors, Antigen, T-Cell/immunology , Retrospective Studies
12.
Sci China Life Sci ; 63(3): 332-342, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32060861

ABSTRACT

Cellular senescence (CS) is a state of stable cell cycle arrest characterized by the production and secretion of inflammatory molecules. Early studies described oncogene-induced senescence (OIS) as a barrier to tumorigenesis, such that the therapeutic induction of CS might represent a rational anti-cancer strategy. Indeed, the validity of this approach has been borne out by the development and approval of the cyclin-dependent kinase (CDK) inhibitor palbociclib for the treatment of breast cancer. Apart from tumors, senescent cells have also been shown to accumulate during natural mammalian aging, where they produce detrimental effects on the physiology of surrounding tissues. Thus, pharmacological senescent cell depletion has been proposed as an approach to delay age-related functional decline; this has been formally demonstrated in animal models. In this review article, we describe the current mechanistic understanding of cellular senescence at the molecular level and how it informs the development of new therapeutic strategies to combat cancer and aging.


Subject(s)
Aging/drug effects , Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Animals , Antineoplastic Agents/metabolism , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Discovery , Female , Humans , Piperazines/metabolism , Protein Kinase Inhibitors/metabolism , Pyridines/metabolism
13.
Nat Commun ; 9(1): 5361, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30560866

ABSTRACT

Combining whole exome sequencing, transcriptome profiling, and T cell repertoire analysis, we investigate the spatial features of surgically-removed biopsies from multiple loci in tumor masses of 15 patients with non-small cell lung cancer (NSCLC). This revealed that the immune microenvironment has high spatial heterogeneity such that intratumoral regional variation is as large as inter-personal variation. While the local total mutational burden (TMB) is associated with local T-cell clonal expansion, local anti-tumor cytotoxicity does not directly correlate with neoantigen abundance. Together, these findings caution against that immunological signatures can be predicted solely from TMB or microenvironmental analysis from a single locus biopsy.


Subject(s)
Antigens, Neoplasm/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Antigens, Neoplasm/immunology , Biopsy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , DNA Mutational Analysis , Gene Expression Profiling , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Tumor Microenvironment/genetics , Exome Sequencing
14.
Nat Commun ; 9(1): 4274, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323337

ABSTRACT

Intrinsic resistance to anti-HER2 therapy in breast cancer remains an obstacle in the clinic, limiting its efficacy. However, the biological basis for intrinsic resistance is poorly understood. Here we performed a CRISPR/Cas9-mediated loss-of-function genetic profiling and identified TALDO1, which encodes the rate-limiting transaldolase (TA) enzyme in the non-oxidative pentose phosphate pathway, as essential for cellular survival following pharmacological HER2 blockade. Suppression of TA increases cell susceptibility to HER2 inhibition in two intrinsically resistant breast cancer cell lines with HER2 amplification. Mechanistically, TA depletion combined with HER2 inhibition significantly reduces cellular NADPH levels, resulting in excessive ROS production and deficient lipid and nucleotide synthesis. Importantly, higher TA expression correlates with poor response to HER2 inhibition in a breast cancer patient cohort. Together, these results pinpoint TA as a novel metabolic enzyme possessing synthetic lethality with HER2 inhibition that can potentially be exploited as a biomarker or target for combination therapy.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Receptor, ErbB-2/genetics , Synthetic Lethal Mutations/genetics , Transaldolase/genetics , Breast Neoplasms/metabolism , CRISPR-Cas Systems , Cell Death/drug effects , Cell Line, Tumor , Female , Genetic Testing , HEK293 Cells , Humans , Lapatinib/pharmacology , Metabolic Flux Analysis , NADP/metabolism , Pentose Phosphate Pathway
15.
Nat Med ; 24(10): 1536-1544, 2018 10.
Article in English | MEDLINE | ID: mdl-30297899

ABSTRACT

Impaired immunity in patients with late-stage cancer is not limited to antitumor responses, as demonstrated by poor vaccination protection and high susceptibility to infection1-3. This has been largely attributed to chemotherapy-induced impairment of innate immunity, such as neutropenia2, whereas systemic effects of tumors on hematopoiesis and adoptive immunity remain incompletely understood. Here we observed anemia associated with severe deficiency of CD8+ T cell responses against pathogens in treatment-naive mice bearing large tumors. Specifically, we identify CD45+ erythroid progenitor cells (CD71+TER119+; EPCs) as robust immunosuppressors. CD45+ EPCs, induced by tumor growth-associated extramedullary hematopoiesis, accumulate in the spleen to become a major population, outnumbering regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). The CD45+ EPC transcriptome closely resembles that of MDSCs, and, like MDSCs, reactive oxygen species production is a major mechanism underlying CD45+ EPC-mediated immunosuppression. Similarly, an immunosuppressive CD45+ EPC population was detected in patients with cancer who have anemia. These findings identify a major population of immunosuppressive cells that likely contributes to the impaired T cell responses commonly observed in patients with advanced cancer.


Subject(s)
Anemia/immunology , Erythroid Precursor Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Sarcoma, Myeloid/immunology , Anemia/genetics , Anemia/pathology , Animals , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Humans , Immune Tolerance , Immunity, Innate/genetics , Leukocyte Common Antigens/immunology , Mice , Neoplasm Staging , Reactive Oxygen Species/metabolism , Receptors, Transferrin/immunology , Sarcoma, Myeloid/metabolism , Sarcoma, Myeloid/pathology , T-Lymphocytes, Regulatory/immunology , Xenograft Model Antitumor Assays
16.
EMBO Rep ; 19(6)2018 06.
Article in English | MEDLINE | ID: mdl-29777051

ABSTRACT

Cellular senescence is a unique cell fate characterized by stable proliferative arrest and the extensive production and secretion of various inflammatory proteins, a phenomenon known as the senescence-associated secretory phenotype (SASP). The molecular mechanisms responsible for generating a SASP in response to senescent stimuli remain largely obscure. Here, using unbiased gene expression profiling, we discover that the scavenger receptor CD36 is rapidly upregulated in multiple cell types in response to replicative, oncogenic, and chemical senescent stimuli. Moreover, ectopic CD36 expression in dividing mammalian cells is sufficient to initiate the production of a large subset of the known SASP components via activation of canonical Src-p38-NF-κB signaling, resulting in the onset of a full senescent state. The secretome is further shown to be ligand-dependent, as amyloid-beta (Aß) is sufficient to drive CD36-dependent NF-κB and SASP activation. Finally, loss-of-function experiments revealed a strict requirement for CD36 in secretory molecule production during conventional senescence reprogramming. Taken together, these results uncover the Aß-CD36-NF-κB signaling axis as an important regulator of the senescent cell fate via induction of the SASP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , CD36 Antigens/physiology , Cellular Senescence/physiology , NF-kappa B/metabolism , CD36 Antigens/genetics , Cells, Cultured , Cellular Senescence/genetics , Fibroblasts/metabolism , Humans , Loss of Function Mutation , Signal Transduction
17.
Cell Discov ; 3: 17019, 2017.
Article in English | MEDLINE | ID: mdl-28626588

ABSTRACT

Cellular senescence is a cell fate characterized by an irreversible cell cycle arrest, but the molecular mechanism underlying this senescence hallmark remains poorly understood. Through an unbiased search for novel senescence regulators in airway basal cells, we discovered that the epigenetic regulator ubiquitin-like with PHD and ring finger domain-containing protein 1 (UHRF1) is critical for regulating cell cycle progression. Upon injury, basal cells in the mouse airway rapidly induce the expression of UHRF1 in order to stimulate stem cell proliferation and tissue repair. Targeted depletion of Uhrf1 specifically in airway basal cells causes a profound defect in cell cycle progression. Consistently, cultured primary human basal cells lacking UHRF1 do not exhibit cell death or differentiation phenotypes but undergo a spontaneous program of senescence. Mechanistically, UHRF1 loss induces G1 cell cycle arrest by abrogating DNA replication factory formation as evidenced by loss of proliferating cell nuclear antigen (PCNA) puncta and an inability to enter the first cell cycle. This proliferation defect is partially mediated by the p15 pathway. Overall, our study provides the first evidence of an indispensable role of UHRF1 in somatic stem cells proliferation during the process of airway regeneration.

18.
Article in English | MEDLINE | ID: mdl-27920038

ABSTRACT

The transforming growth factor ß (TGF-ß) family controls many fundamental aspects of cellular behavior. With advances in the molecular details of the TGF-ß signaling cascade and its cross talk with other signaling pathways, we now have a more coherent understanding of the cytostatic program induced by TGF-ß. However, the molecular mechanisms are still largely elusive for other cellular processes that are regulated by TGF-ß and determine a cell's proliferation and survival, apoptosis, dormancy, autophagy, and senescence. The difficulty in defining TGF-ß's roles partly stems from the context-dependent nature of TGF-ß signaling. Here, we review our current understanding and recent progress on the biological effects of TGF-ß at the cellular level, with the hope of providing a framework for understanding how cells respond to TGF-ß signals in specific contexts, and why disruption of such mechanisms may result in different human diseases including cancer.


Subject(s)
Cell Proliferation , Cell Survival , Signal Transduction , Transforming Growth Factor beta/metabolism , Apoptosis , Autophagy , Cell Cycle Checkpoints , Humans
19.
J Biol Chem ; 292(2): 748-759, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-27903634

ABSTRACT

Targeted inhibitors of the human epidermal growth factor receptor 2 (HER2), such as trastuzumab and lapatinib, are among the first examples of molecularly targeted cancer therapy and have proven largely effective for the treatment of HER2-positive breast cancers. However, approximately half of those patients either do not respond to these therapies or develop secondary resistance. Although a few signaling pathways have been implicated, a comprehensive understanding of mechanisms underlying HER2 inhibitor drug resistance is still lacking. To address this critical question, we undertook a concerted approach using patient expression data sets, HER2-positive cell lines, and tumor samples biopsied both before and after trastuzumab treatment. Together, these methods revealed that high expression and activation of a specific subset of receptor tyrosine kinases (RTKs) was strongly associated with poor clinical prognosis and the development of resistance. Mechanistically, these RTKs are capable of maintaining downstream signal transduction to promote tumor growth via the suppression of cellular senescence. Consequently, these findings provide the rationale for the design of therapeutic strategies for overcoming drug resistance in breast cancer via combinational inhibition of the limited number of targets from this specific subset of RTKs.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Quinazolines/pharmacology , Signal Transduction , Trastuzumab/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Lapatinib , Receptor, ErbB-2 , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Oncotarget ; 7(15): 20381-94, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26967387

ABSTRACT

Triple-negative breast cancer (TNBC) presents a major challenge in the clinic due to its lack of reliable prognostic markers and targeted therapies. Accumulating evidence strongly supports the notion that microRNAs (miRNAs) are involved in tumorigenesis and could serve as biomarkers for diagnostic purposes. To identify miRNAs that functionally suppress metastasis of TNBC, we employed a concerted approach with selecting miRNAs that display differential expression profiles from bioinformatic analyses of breast cancer patient databases and validating top candidates with functional assays using breast cancer cell lines and mouse models. We have found that miR-148a exhibits properties as a tumor suppressor as its expression is inversely correlated with the ability of both human and mouse breast cancer cells to colonize the lung in mouse xenograft tumor models. Mechanistically, miR-148a appears to suppress the extravasation process of cancer cells, likely by targeting two genes WNT1 and NRP1 in a cell non-autonomous manner. Importantly, lower expression of miR-148a is detected in higher-grade tumor samples and correlated with increased likelihood to develop metastases and poor prognosis in subsets of breast cancer patients, particularly those with TNBC. Thus, miR-148a is functionally defined as a suppressor of breast cancer metastasis and may serve as a prognostic biomarker for this disease.


Subject(s)
Biomarkers, Tumor/metabolism , Lung Neoplasms/secondary , MicroRNAs/genetics , Neuropilin-1/metabolism , Triple Negative Breast Neoplasms/pathology , Wnt1 Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neuropilin-1/genetics , Prognosis , Survival Rate , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Wnt1 Protein/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...