Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell Death Dis ; 14(5): 316, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37160920

ABSTRACT

Extranodal NK/T-cell lymphoma (NKTL) is a rare and aggressive form of extranodal lymphoma with a poor prognosis. Currently, there are very limited treatment options for patients with advanced-stage disease or those with relapsed/recurrent disease. Here we show that Chiauranib, an orally small molecule inhibitor of select serine-threonine kinases (aurora B, VEGFRs, PDGFR, CSF1R, c-Kit), inhibited NKTL cell proliferation, induced cell cycle arrest, as well as suppressed the microvessel density in vitro and in vivo similar as in other types of cancer cells. Surprisingly, Chiauranib unfolded a new effect to induce apoptosis of NKTL cells by triggering AIF-dependent apoptosis other than the traditional cyt-c/caspase mitochondrial apoptosis pathway. The knockdown of AIF in vitro and in vivo dramatically blocked the efficacy of Chiauranib on NKTL. Mechanistically, the release of AIF from mitochondria is due to the upregulation of VDAC1 by the AKT-GSK3ß pathway and activation of calcium-dependent m-calpain, which promotes the cleavage of VDAC1 and therefore permits the release of AIF. Notably, the low expression of Bax in both NKTL cells and patient tissues restrained the cyt-c release. It resulted in the inhibition of cyt-c/caspase mitochondrial pathway, suggesting that drugs targeting this traditional pathway may not be effective in NKTL. Furthermore, we found that L-asparaginase triggered CD95 (Fas/Apo-1)-caspase 8-caspase 3 apoptotic pathway in NKTL cells, and combination of Chiauranib and L-asparaginase exhibited a synergistic effect, suggesting a feasibility to combine these two drugs for effective treatment of NKTL. This study demonstrates Chiauranib's positive efficacy toward NKTL through the activation of the AIF-dependent apoptosis pathway for the first time. The novel and multi-targets of Chiauranib and the synergistic effect with L-asparaginase may provide a promising therapy for NKTL patients.


Subject(s)
Lymphoma, T-Cell , Quinolines , Humans , Asparaginase/pharmacology , Asparaginase/therapeutic use , Naphthalenes
2.
J Clin Neurosci ; 105: 31-36, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36063751

ABSTRACT

OBJECTIVES: This study aimed to investigate the role of peripheral neutrophil to lymphocyte ratio (NLR), monocyte to lymphocyte ratio (MLR), and platelet to lymphocyte ratio (PLR) in the progression and severity of the Guillain-Barré syndrome (GBS). METHODS: 47 GBS patients and 50 age and sex-matched healthy controls (HC) were retrospectively included. Demographic and clinical assessment data were reviewed and abstracted. NLR, MLR, and PLR were calculated based on the peripheral blood tests by reviewing clinical data. The relationship between the Hughes' score and NLR, MLR, PLR levels was investigated. RESULTS: The GBS patients had higher NLR levels (P < 0.001), MLR levels (P = 0.001) and PLR levels (P < 0.001) than those in HC. And patients with severe disability score (Hughes' score ≥ 3) had significantly higher NLR (P = 0.007), MLR (P = 0.04), PLR (P = 0.013). Spearman correlation analysis indicated that NLR was positively associated with the Hughes' score (r = 0.331, P = 0.023). In the patients with acute inflammatory demyelinating polyneuropathy (AIDP), Spearman correlation analysis indicated that NLR, MLR and PLR were positively associated to the Hughes' score (r = 0.825, P = 0.001 for NLR, r = 0.727, P = 0.005 for MLR, and r = 0.723, P = 0.005 for PLR). CONCLUSIONS: NLR, MLR, and PLR may be indicators of disease activity in patients with GBS or AIDP. These parameters may benefit the active treatment of GBS patients with a high degree of disability.


Subject(s)
Guillain-Barre Syndrome , Lymphocytes , Biomarkers , Blood Platelets , Guillain-Barre Syndrome/diagnosis , Humans , Neutrophils , Prognosis , Retrospective Studies
3.
Nat Cell Biol ; 24(7): 1165-1176, 2022 07.
Article in English | MEDLINE | ID: mdl-35773432

ABSTRACT

CD8+ T cells are central mediators of immune responses against infections and cancer. Here we identified Dapl1 as a crucial regulator of CD8+ T cell responses to cancer and infections. Dapl1 deficiency promotes the expansion of tumour-infiltrating effector memory-like CD8+ T cells and prevents their functional exhaustion, coupled with increased antitumour immunity and improved efficacy of adoptive T cell therapy. Dapl1 controls activation of NFATc2, a transcription factor required for the effector function of CD8+ T cells. Although NFATc2 mediates induction of the immune checkpoint receptor Tim3, competent NFATc2 activation prevents functional exhaustion of CD8+ T cells. Interestingly, exhausted CD8+ T cells display attenuated NFATc2 activation due to Tim3-mediated feedback inhibition; Dapl1 deletion rescues NFATc2 activation and thereby prevents dysfunction of exhausted CD8+ T cells in chronic infection and cancer. These findings establish Dapl1 as a crucial regulator of CD8+ T cell immunity and a potential target for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Membrane Proteins , NFATC Transcription Factors/genetics , Neoplasms/genetics , Persistent Infection , Transcription Factors
4.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35074921

ABSTRACT

Proinflammatory cytokine production by innate immune cells plays a crucial role in inflammatory diseases, but the molecular mechanisms controlling the inflammatory responses are poorly understood. Here, we show that TANK-binding kinase 1 (TBK1) serves as a vital regulator of proinflammatory macrophage function and protects against tissue inflammation. Myeloid cell-conditional Tbk1 knockout (MKO) mice spontaneously developed adipose hypertrophy and metabolic disorders at old ages, associated with increased adipose tissue M1 macrophage infiltration and proinflammatory cytokine expression. When fed with a high-fat diet, the Tbk1-MKO mice also displayed exacerbated hepatic inflammation and insulin resistance, developing symptoms of nonalcoholic steatohepatitis. Furthermore, myeloid cell-specific TBK1 ablation exacerbates inflammation in experimental colitis. Mechanistically, TBK1 functions in macrophages to suppress the NF-κB and MAP kinase signaling pathways and thus attenuate induction of proinflammatory cytokines, particularly IL-1ß. Ablation of IL-1 receptor 1 (IL-1R1) eliminates the inflammatory symptoms of Tbk1-MKO mice. These results establish TBK1 as a pivotal anti-inflammatory mediator that restricts inflammation in different disease models.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Biomarkers , Colitis/etiology , Colitis/metabolism , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Disease Susceptibility/immunology , Gene Expression Regulation , Glucose/metabolism , Hypertrophy , Immunomodulation/genetics , Inflammation/pathology , Inflammation Mediators/metabolism , Insulin Resistance , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Organ Specificity , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-1/deficiency , Signal Transduction
5.
Neurol Sci ; 43(4): 2785-2790, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34709479

ABSTRACT

OBJECTIVE: Myasthenia gravis (MG) is an autoimmune disorder whose main symptoms are muscle weakness and fatigue. Irisin is a novel skeletal muscle-derived myokine participating in several physiological and pathological processes. The initial objective of the project was to explore serum levels of irisin in patients with MG, as well as its correlation with disease severity. METHODS: We retrospectively evaluated serum levels of irisin in 77 MG patients and 57 healthy controls (HCs) by enzyme-linked immunosorbent assay. Further, clinical parameters were measured properly. RESULTS: Serum irisin levels were significantly elevated in MG patients compared with HCs (p < 0.001). Furthermore, serum irisin levels were associated with the myasthenia gravis activities of daily living score in ocular myasthenia gravis (OMG) patients (r = 0.476, p = 0.004), but there was no relationship to be considered of any relevant value in generalized myasthenia gravis (GMG) patients. Acetylcholine receptor antibody-positive MG patients had higher serum irisin levels compared with HCs. Thymoma, endotracheal intubation, or intensive care unit treatments subsequently were not found to have effect on serum levels of irisin, but tendencies of increase were observed in negative ones. CONCLUSIONS: Serum irisin levels were elevated in patients with MG, suggesting its possible involvement in MG. And irisin is expected to be a signal to evaluate the activities of daily living of OMG patients, while its effect needs further study.


Subject(s)
Activities of Daily Living , Fibronectins , Myasthenia Gravis , Autoantibodies/blood , Fibronectins/blood , Humans , Myasthenia Gravis/blood , Myasthenia Gravis/diagnosis , Receptors, Cholinergic/immunology , Retrospective Studies
6.
Cell Rep ; 37(4): 109904, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706239

ABSTRACT

Inflammasomes are crucial for innate immunity against infections and, when deregulated, also contribute to inflammatory diseases. Here, we identify a critical function of the E3 ubiquitin ligase Peli1 in regulating the activation of NLRP3 inflammasome. Peli1 deficiency impairs induction of interleukin-1ß (IL-1ß) secretion by different NLRP3 inducers, but not by inducers of the Aim2, NLRP1, and NLRC4 inflammasomes. Peli1-deficient mice have alleviated peritonitis induction by alum and display increased resistance to lipopolysaccharide (LPS) endotoxin shock, coupled with decreased serum concentration of IL-1ß. Peli1 is required for NLRP3-induced caspase-1 activation and IL-1ß maturation. Mechanistically, Peli1 conjugates K63 ubiquitin chain to lysine 55 of the inflammasome adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), which in turn facilitates ASC/NLRP3 interaction and ASC oligomerization, thereby contributing to inflammasome activation. Peli1 deficiency impairs the ubiquitination of ASC and inhibits inflammasome activation. Our findings establish Peli1 as an important inflammasome regulator and suggest a mechanism by which Peli1 mediates inflammatory responses.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nuclear Proteins/metabolism , Protein Multimerization , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Cell Line , Humans , Inflammation/metabolism , Mice , Mice, Transgenic
7.
Sci Adv ; 7(36): eabh0609, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34516909

ABSTRACT

Microglia have been implicated in neuroinflammatory diseases, including multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). We demonstrate that microglia mediate EAE disease progression via a mechanism relying on the noncanonical nuclear factor kB (NF-κB) pathway. Microglia-specific deletion of the noncanonical NF-κB-inducing kinase (NIK) impairs EAE disease progression. Although microglial NIK is dispensable for the initial phase of T cell infiltration into the central nervous system (CNS) and EAE disease onset, it is critical for the subsequent CNS recruitment of inflammatory T cells and monocytes. Our data suggest that following their initial CNS infiltration, T cells activate the microglial noncanonical NF-κB pathway, which synergizes with the T cell-derived cytokine granulocyte-macrophage colony-stimulating factor to induce expression of chemokines involved in the second-wave of T cell recruitment and disease progression. These findings highlight a mechanism of microglial function that is dependent on NIK signaling and required for EAE disease progression.

8.
Mol Oncol ; 15(12): 3490-3512, 2021 12.
Article in English | MEDLINE | ID: mdl-34288405

ABSTRACT

Distant metastasis is, unfortunately, the leading cause of death in colorectal cancer (CRC). Approximately 50% of CRC patients develop liver metastases, while 10-30% of patients develop pulmonary metastases. The occurrence of metastasis is considered to be almost exclusively driven by cancer stem cells (CSCs) formation. However, the key molecules that confer the transformation to stem cells in CRC, and subsequent metastasis, remain unclear. Far upstream element-binding protein 1 (FUBP1), a transcriptional regulator of c-Myc, was screened in CSCs of CRC by mass spectrometry and was examined by immunohistochemistry in a cohort of CRC tissues. FUBP1 was upregulated in 85% of KRAS-mutant and 25% of wild-type CRC patients. Further, whether in KRAS-mutant or wild-type patients, elevated FUBP1 was positively correlated with CRC lymph node metastasis and clinical stage, and negatively associated with overall survival. Overexpression of FUBP1 significantly enhanced CRC cell migration, invasion, tumor sphere formation, and CD133 and ALDH1 expression in vitro, and tumorigenicity in vivo. Mechanistically, FUBP1 promoted the initiation of CSCs by activating Wnt/ß-catenin signaling via directly binding to the promoter of DVL1, a potent activator of ß-catenin. Knockdown of DVL1 significantly inhibited the transformation to stem cells in, as well as the tumorigenicity of, CRC. Activation of Wnt/ß-catenin signaling by DVL1 increased pluripotent transcription factors, including c-Myc, NANOG, and SOX2. Moreover, FUBP1 was upregulated at the post-transcriptional level. Elevated FUBP1 levels in KRAS wild-type CRC patients is due to the decrease in Smurf2, which promotes ubiquitin-mediated degradation of FUBP1. In contrast, FUBP1 was upregulated in KRAS-mutant patients through both inhibition of caspase 3-dependent cleavage and decreased Smurf2. Our results demonstrate, for the first time, that FUBP1 is an oncogene, initiating the development of CSCs, as well as a new powerful endogenous Wnt-signaling agonist that could provide an important prognostic factor and therapeutic target for metastasis in both KRAS-mutant and wild-type CRC.


Subject(s)
Colorectal Neoplasms , DNA-Binding Proteins , Dishevelled Proteins , Neoplastic Stem Cells , RNA-Binding Proteins , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/pathology , Oncogenes , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
9.
Blood ; 138(23): 2360-2371, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34255829

ABSTRACT

B-cell-activating factor (BAFF) mediates B-cell survival and, when deregulated, contributes to autoimmune diseases and B-cell malignancies. The mechanism connecting BAFF receptor (BAFFR) signal to downstream pathways and pathophysiological functions is not well understood. Here we identified DYRK1a as a kinase that responds to BAFF stimulation and mediates BAFF-induced B-cell survival. B-cell-specific DYRK1a deficiency causes peripheral B-cell reduction and ameliorates autoimmunity in a mouse model of lupus. An unbiased screen identified DYRK1a as a protein that interacts with TRAF3, a ubiquitin ligase component mediating degradation of the noncanonical nuclear factor (NF)-κB-inducing kinase (NIK). DYRK1a phosphorylates TRAF3 at serine-29 to interfere with its function in mediating NIK degradation, thereby facilitating BAFF-induced NIK accumulation and noncanonical NF-κB activation. Interestingly, B-cell acute lymphoblastic leukemia (B-ALL) cells express high levels of BAFFR and respond to BAFF for noncanonical NF-κB activation and survival in a DYRK1a-dependent manner. Furthermore, DYRK1a promotes a mouse model of B-ALL through activation of the noncanonical NF-κB pathway. These results establish DYRK1a as a critical BAFFR signaling mediator and provide novel insight into B-ALL pathogenesis.


Subject(s)
Autoimmunity , B-Cell Activating Factor/immunology , Leukemia, B-Cell/immunology , NF-kappa B/immunology , Protein Serine-Threonine Kinases/immunology , Protein-Tyrosine Kinases/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Line, Tumor , Humans , Leukemia, B-Cell/pathology , Mice , Mice, Inbred C57BL , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Dyrk Kinases
10.
Ann Hematol ; 100(9): 2293-2302, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33991204

ABSTRACT

Despite the clinical value of HMGB1 in non-Hodgkin lymphoma (NHL), the impact of HMGB1 protein expression on survival of patients with mature T-cell and NK-cell lymphoma (T/NK-CL) is unknown. Here, we evaluated correlations of HMGB1 expression in tumor tissues with pathophysiological characteristics of disease and determined the prognostic value of HMGB1 expression in relapsed/refractory T/NK-CL. HMGB1 expression was detected by immunohistochemistry (IHC) in 66 cases of relapsed/refractory T/NK-CL, and specimens were classified as high or low HMGB1 expression. Univariate and multivariate Cox regression analyses identified prognostic factors associated with progression-free survival (PFS) and overall survival (OS). High HMGB1 expression was significantly correlated with increased Ki67 levels and progressive lymphoma subtypes. Univariate Cox regression analysis showed that high HMGB1 expression was associated with unfavorable PFS (P = 0.006) and poorer OS (P < 0.001). Prognostic factors identified by univariate analysis were prognostic index for peripheral T-cell lymphoma non-specified (PIT) score ≥ 2, bone marrow involvement, Ki67 ≥ 70%, and high HMGB1 expression. Multivariate Cox regression analysis revealed that high HMGB1 expression was an independent prognostic factor for poorer PFS [hazard ratio (HR) 3.593; 95% confidence interval (CI) 1.171-11.027; P = 0.025] and OS [HR 7.663; 95% CI 2.367-24.803; P = 0.001]. A proposal prognostic model combining HMGB1 and Ki67 expression showed improved prognostic capacity and may help guide treatment planning. High HMGB1 expression may be a promising prognostic predictor and a potential therapeutic target for relapsed/refractory T/NK-CL. Furthermore, to apply HMGB1 as one of the best bio-maker, an external independent control cohort is needed.


Subject(s)
HMGB1 Protein/analysis , Lymphoma, Extranodal NK-T-Cell/diagnosis , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Doxorubicin/therapeutic use , Female , Humans , Ki-67 Antigen/analysis , Lymphoma, Extranodal NK-T-Cell/drug therapy , Lymphoma, Extranodal NK-T-Cell/pathology , Lymphoma, Extranodal NK-T-Cell/radiotherapy , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Prednisone/therapeutic use , Prognosis , Survival Analysis , Vincristine/therapeutic use , Young Adult
11.
Cell Death Dis ; 12(4): 295, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731707

ABSTRACT

Nasopharyngeal carcinoma (NPC) is one of the most malignant tumors in southern China and Asia, and lymph node metastasis is an important cause for treatment failure. Lymphangiogenesis is a crucial step in lymphatic metastasis of NPC, while little is known about lymphangiogenesis in NPC. Similar to angiogenesis, lymphangitic neovascularization is a process of balance between pro-lymphangiogenesis and anti-lymphangiogenesis factors, but there are few studies on endogenous lymphangiogenesis inhibitors. Pigment epithelium-derived factor (PEDF) is a well-known effective endogenous angiogenesis inhibitor. However, the relationship between PEDF and lymphangiogenesis remains unknown. Our present study reveals that PEDF is lowly expressed in human NPC tissues with poor prognosis and is negatively correlated with lymphatic vessel density (LVD). Consistently, PEDF inhibits lymphangiogenesis and lymphatic metastasis of NPC in vivo experiments. Mechanistically, PEDF inhibits the proliferation, migration, and tube formation of lymphatic endothelial cells and promotes cell apoptosis. On the other hand, PEDF reduces the expression and secretion of vascular endothelial growth factor C (VEGF-C) of NPC cells through the nuclear factor-κB (NF-κB) signaling pathway. Our findings indicate that PEDF plays a vital role in lymphatic metastasis by targeting both lymphatic endothelial cells and NPC cells, and PEDF may represent a novel therapeutic target for NPC.


Subject(s)
Eye Proteins/therapeutic use , Lymphatic Metastasis/drug therapy , Nasopharyngeal Carcinoma/drug therapy , Nerve Growth Factors/therapeutic use , Protease Inhibitors/therapeutic use , Serpins/therapeutic use , Animals , Eye Proteins/pharmacology , Humans , Mice , Nerve Growth Factors/pharmacology , Protease Inhibitors/pharmacology , Serpins/pharmacology , Transfection
12.
EMBO J ; 40(2): e104532, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33215753

ABSTRACT

Metabolic fitness of T cells is crucial for immune responses against infections and tumorigenesis. Both the T cell receptor (TCR) signal and environmental cues contribute to the induction of T cell metabolic reprogramming, but the underlying mechanism is incompletely understood. Here, we identified the E3 ubiquitin ligase Peli1 as an important regulator of T cell metabolism and antitumor immunity. Peli1 ablation profoundly promotes tumor rejection, associated with increased tumor-infiltrating CD4 and CD8 T cells. The Peli1-deficient T cells display markedly stronger metabolic activities, particularly glycolysis, than wild-type T cells. Peli1 controls the activation of a metabolic kinase, mTORC1, stimulated by both the TCR signal and growth factors, and this function of Peli1 is mediated through regulation of the mTORC1-inhibitory proteins, TSC1 and TSC2. Peli1 mediates non-degradative ubiquitination of TSC1, thereby promoting TSC1-TSC2 dimerization and TSC2 stabilization. These results establish Peli1 as a novel regulator of mTORC1 and downstream mTORC1-mediated actions on T cell metabolism and antitumor immunity.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Cell Line, Tumor , Glycolysis/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism
13.
Liver Cancer ; 9(3): 338-357, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32647635

ABSTRACT

BACKGROUND AND AIMS: Combining anti-angiogenic therapy with immune checkpoint blockade with anti-programmed cell death-1 (PD-1) antibodies is a promising treatment for hepatocellular carcinoma (HCC). Tyrosine kinase inhibitors are well-known anti-angiogenic agents and offer potential for combination with anti-PD-1 antibodies. This study investigated the possible underlying immunomodulatory mechanisms of combined therapy. METHODS: HCC tissue samples for RNA-sequencing (RNA-seq) were obtained from patients with differential prognoses following anti-PD-1 treatment. Recombinant basic fibroblast growth factor (bFGF) and vascular endothelial growth factor A (VEGFA) were used to stimulate T cells following lenvatinib or sorafenib treatment, respectively. T cell function was analyzed by flow cytometry and lactate dehydrogenase assay. In vivo experiments were conducted in murine H22 and Hepa 1-6 competent models of HCC. Local immune infiltration in the tumor microenvironment (TME) was assessed using multicolor flow cytometry. Gene regulation was evaluated by RNA-seq. Microvascular density was measured by immunohistochemistry, and PD-1 ligand (PD-L1) induction was quantified by western blot. RESULTS: The baseline expression of VEGF and fibroblast growth factor (FGF) in patients with progressive disease was significantly higher than in patients achieving stable disease following anti-PD-1 treatment. VEGFA and bFGF significantly upregulated the expression of PD-1, cytotoxic T-lymphocyte-associated protein-4, and Tim-3 on T cells, while inhibiting the secretion of interferon gamma (IFNG) and granzyme B and suppressing T cell cytotoxicity. This immunosuppressive effect was reverted by lenvatinib but not sorafenib. Furthermore, dual lenvatinib/anti-PD-1 antibody therapy led to better antitumor effects than either sorafenib or fibroblast growth factor receptor (FGFR) inhibitor (BGJ398) in H22 murine models of HCC. Combined lenvatinib/anti-PD-1 treatment also led to long-term immune memory formation, while synergistically modulating the TME and enhancing the cytotoxic effect of T cells. Finally, lenvatinib inhibited PD-L1 expression on human umbilical vein endothelial cells, which improved the function of T cells. CONCLUSIONS: Inhibition of vascular endothelial growth factor receptor and FGFR augmented the efficacy of anti-PD-1 antibodies. Combined lenvatinib/anti-PD-1 treatment appears to exert antitumor activity by synergistically modulating effector T cell function in the TME and by mutually regulating tumor vessel normalization.

14.
Proc Natl Acad Sci U S A ; 117(23): 13012-13022, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32444490

ABSTRACT

Metastatic colorectal cancer (mCRC) patients have poor overall survival despite using irinotecan- or oxaliplatin-based chemotherapy combined with anti-EGFR (epidermal growth factor receptor) drugs, especially those with the oncogene mutation of KRAS Metformin has been reported as a potentially novel antitumor agent in many experiments, but its therapeutic activity is discrepant and controversial so far. Inspiringly, the median survival time for KRAS-mutation mCRC patients with diabetes on metformin is 37.8 mo longer than those treated with other hypoglycemic drugs in combination with standard systemic therapy. In contrast, metformin could not improve the survival of mCRC patients with wild-type KRAS Interestingly, metformin is preferentially accumulated in KRAS-mutation mCRC cells, but not wild-type ones, in both primary cell cultures and patient-derived xenografts, which is in agreement with its tremendous effect in KRAS-mutation mCRC. Mechanistically, the mutated KRAS oncoprotein hypermethylates and silences the expression of multidrug and toxic compound extrusion 1 (MATE1), a specific pump that expels metformin from the tumor cells by up-regulating DNA methyltransferase 1 (DNMT1). Our findings provide evidence that KRAS-mutation mCRC patients benefit from metformin treatment and targeting MATE1 may provide a strategy to improve the anticancer response of metformin.


Subject(s)
Colorectal Neoplasms/drug therapy , Metformin/pharmacology , Organic Cation Transport Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Metformin/therapeutic use , Mice , Middle Aged , Proto-Oncogene Proteins p21(ras)/metabolism , Xenograft Model Antitumor Assays
15.
Article in English | MEDLINE | ID: mdl-34267412

ABSTRACT

Gaining environmental awareness through lateral head scanning (yaw rotations) is important for driving safety, especially when approaching intersections. Therefore, head scanning movements could be an important behavioral metric for driving safety research and driving risk mitigation systems. Tracking head scanning movements with a single in-car camera is preferred hardware-wise, but it is very challenging to track the head over almost a 180° range. In this paper we investigate two state-of-the-art methods, a multi-loss deep residual learning method with 50 layers (multi-loss ResNet-50) and an ORB feature-based simultaneous localization and mapping method (ORB-SLAM). While deep learning methods have been extensively studied for head pose detection, this is the first study in which SLAM has been employed to innovatively track head scanning over a very wide range. Our laboratory experimental results showed that ORB-SLAM was more accurate than multi-loss ResNet-50, which often failed when many facial features were not in the view. On the contrary, ORB-SLAM was able to continue tracking as it doesn't rely on particular facial features. Testing with real driving videos demonstrated the feasibility of using ORB-SLAM for tracking large lateral head scans in naturalistic video data.

16.
Nat Cell Biol ; 21(12): 1604-1614, 2019 12.
Article in English | MEDLINE | ID: mdl-31792381

ABSTRACT

TANK-binding kinase 1 (TBK1) responds to microbial stimuli and mediates the induction of type I interferon (IFN). Here, we show that TBK1 is also a central mediator of growth factor signalling; this function of TBK1 relies on a specific adaptor-TBK-binding protein 1 (TBKBP1). TBKBP1 recruits TBK1 to protein kinase C-theta (PKCθ) through a scaffold protein, CARD10. This enables PKCθ to phosphorylate TBK1 at Ser 716, a crucial step for TBK1 activation by growth factors but not by innate immune stimuli. Although the TBK1-TBKBP1 signalling axis is not required for the induction of type I IFN, it mediates mTORC1 activation and oncogenesis. Conditional deletion of either TBK1 or TBKBP1 in lung epithelial cells inhibits tumourigenesis in a mouse model of lung cancer. In addition to promoting tumour growth, the TBK1-TBKBP1 axis facilitates tumour-mediated immunosuppression through a mechanism that involves induction of the checkpoint molecule PD-L1 and stimulation of glycolysis. These findings suggest a PKCθ-TBKBP1-TBK1 growth factor signalling axis that mediates both tumour growth and immunosuppression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinogenesis/genetics , Immune Tolerance/genetics , Intercellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , A549 Cells , Animals , CARD Signaling Adaptor Proteins/genetics , Cells, Cultured , Epithelial Cells/pathology , HEK293 Cells , Humans , Immunity, Innate/genetics , Interferon Type I/genetics , Lung/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL
17.
Cell Death Dis ; 10(10): 742, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31582735

ABSTRACT

Pigment epithelium-derived factor (PEDF), a classic angiogenic inhibitor, has been reported to function as a tumor suppression protein and to downregulate in many types of solid tumors. However, the expression level of PEDF and its role in hepatocellular carcinoma (HCC) are contradictory. The present study investigates the expression and different activities of secreted and intracellular PEDF during HCC development, as well as the underlying mechanism of PEDF on HCC lipid disorders. We found that PEDF had no association with patients' prognosis, although PEDF was highly expressed and inhibited angiogenesis in HCC tumor tissues. The animal experiments indicated that full-length PEDF exhibited equalizing effects on tumor growth activation and tumor angiogenesis inhibition in the late stage of HCC progression. Importantly, the pro-tumor activity was mediated by the intracellular PEDF, which causes accumulation of free fatty acids (FFAs) in vivo and in vitro. Based on the correlation analysis of PEDF and lipid metabolic indexes in human HCC tissues, we demonstrated that the intracellular PEDF led to the accumulation of FFA and eventually promoted HCC cell growth by inhibiting the activation of AMPK via ubiquitin-proteasome-mediated degradation, which causes increased de novo fatty acid synthesis and decreased FFA oxidation. Our findings revealed why elevated PEDF did not improve the patients' prognosis as the offsetting intracellular and extracellular activities. This study will lead to a comprehensive understanding of the diverse role of PEDF in HCC and provide a new selective strategy by supplement of extracellular PEDF and downregulation of intracellular PEDF for the prevention and treatment of liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Extracellular Space/metabolism , Eye Proteins/metabolism , Intracellular Space/metabolism , Liver Neoplasms/metabolism , Nerve Growth Factors/metabolism , Serpins/metabolism , Adenylate Kinase/metabolism , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Eye Proteins/genetics , Fatty Acids/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Models, Biological , Neoplasm Staging , Nerve Growth Factors/genetics , Prognosis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Serpins/genetics , Ubiquitin/metabolism , Xenograft Model Antitumor Assays
18.
J Exp Clin Cancer Res ; 38(1): 400, 2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31511046

ABSTRACT

BACKGROUND: Neuroblastoma (NB) is one of the deadliest paediatric solid tumours due to its rapid proliferative characteristics. Amplified copies of MYCN are considered the most important marker for the prediction of tumour relapse and progression in NB, but they were only detected in 20-30% of NB patients, indicating there might be other oncogenes in the development of NB. The far upstream element binding protein 1 (FUBP1) was first identified as a transcriptional regulator of the proto-oncogene MYC. However, the expression and role of FUBP1 in NB have not been documented. METHODS: FUBP1 expression was analysed from GEO database and verified by immunohistochemistry (IHC) and western blotting (WB) in NB tissues and cell lines. Cell proliferation and apoptosis were detected by Cell Counting Kit-8, Colony formation assay, EDU, TUNEL staining and flow cytometric analysis. Several glycolytic metabolites production was confirmed by ELISA and oxygen consuming rate (OCR). Luciferase assay, WB, chromatin immunoprecipitation (CHIP) were used to explore the mechanisms of the effect of FUBP1 on NB. RESULTS: FUBP1 mRNA levels were increased along with the increase in International Neuroblastoma Staging System (INSS) stages. High expression of FUBP1 with low N-Myc expression accounted for 44.6% of NB patient samples (n = 65). In addition, FUBP1 protein levels were remarkably increased with NB malignancy in the NB tissue microarray (NB: n = 65; ganglioneuroblastoma: n = 31; ganglioneuroma: n = 27). Furthermore, FUBP1 expression was negatively correlated with patient survival rate but positively correlated with ki67 content. In vitro experiments showed that FUBP1 promotes NB cell proliferation and inhibits cell apoptosis via enhancing glycolysis and ATP production. Mechanistically, FUBP1 inhibited the degradation of HIF1α via downregulation of Von Hippel-Lindau (VHL), the E3 ligase for HIF1α, resulting in upregulation of lactate dehydrogenase isoform B (LDHB) expression to enhance glycolysis. Overexpressed or silenced N-Myc could not regulate FUBP1 or LDHB levels. CONCLUSIONS: Taken together, our findings demonstrate for the first time that elevated FUBP1 promotes NB glycolysis and growth by targeting HIF1α rather than N-Myc, suggesting that FUBP1 is a novel and powerful oncogene in the development of NB independent of N-Myc and may have potential in the diagnosis and treatment of NB.


Subject(s)
Biomarkers, Tumor , DNA-Binding Proteins/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , RNA-Binding Proteins/genetics , Apoptosis/genetics , Biomarkers , Cell Line, Tumor , Cell Proliferation , Databases, Genetic , Gene Expression , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Models, Biological , Neuroblastoma/pathology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
19.
Cancer Med ; 8(10): 4688-4698, 2019 08.
Article in English | MEDLINE | ID: mdl-31218841

ABSTRACT

Patients with Non-Hodgkin lymphoma (NHL) treated by conventional chemotherapeutic drugs usually require a long recovery period. However, metronomic combination chemotherapy (MCC) enhances therapeutic efficacy and decreases side effects in the treatment of NHL. In this study, we tested and compared the effects of metronomic chemotherapy (MC) using podophyllotoxin derivative etoposide (VP-16) alone and that of MCC using both VP-16 and everolimus (RAD001) in the treatment of NHL. Two types of NHL cells, OCI-LY-10 and SU-DHL-6, were employed for the experiments. Cell proliferation, apoptosis, and cell senescence were measured to test the effects of drugs in each experiment. In addition, the influences of MC and MCC on the cell cycle and autophagy pathway were evaluated to study the functional mechanisms behind their effects. Finally, we conducted analyses of the growth inhibitory effect and synergistic activity for different MCC. The results showed that MC using low-dose VP-16 alone demonstrated strong treatment effects in terms of inducing apoptosis, cell senescence, and reducing tumor cell proliferation, and this treatment also led to changes of the cell cycle. Compared with MC, MCC using VP-16 and RAD001 together demonstrated even stronger treatment effects, with both the cell cycle and autophagy-related proteins being affected. Considering the synergistic activity, our results showed the MCC of VP-16 48 hours + RAD001 24 hours is the optimal method for treating NHL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy-Related Proteins/metabolism , Etoposide/pharmacology , Everolimus/pharmacology , Lymphoma, Non-Hodgkin/metabolism , Administration, Metronomic , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Non-Hodgkin/drug therapy , Survival Analysis , Treatment Outcome
20.
Gastric Cancer ; 21(4): 617-631, 2018 07.
Article in English | MEDLINE | ID: mdl-29243194

ABSTRACT

BACKGROUND: Tumor-induced lymphangiogenesis and lymphatic metastasis are predominant during the metastasis of many types of cancers. However, the endogenous inhibitors that counterbalance the lymphangiogenesis and lymphatic metastasis of tumors have not been well evaluated. Kallistatin has been recognized as an endogenous angiogenesis inhibitor. METHODS AND RESULTS: Our recent study showed for the first time that the lymphatic vessel density (LVD) was reduced in lung and stomach sections from kallistatin-overexpressing transgenic mice. Kallistatin expresses anti-lymphangiogenic activity by inhibiting the proliferation, migration, and tube formation of human lymphatic endothelial cells (hLECs). Therefore, the present study focuses on the relationships of changes in kallistatin expression with the lymphangiogenesis and lymphatic metastasis of gastric cancer and its underlying mechanisms. Our results revealed that the expression of kallistatin in cancer tissues, metastatic lymph nodes, and plasma of gastric cancer patients was significantly downregulated and that the plasma level of kallistatin was negatively associated with the phase of lymph node metastasis. Furthermore, treatment with kallistatin recombinant protein decreased LVD and lymph node metastases in the implanted gastric xenograft tumors of nude mice. Mechanically, kallistatin suppressed the lymphangiogenesis and lymphatic metastasis by downregulating VEGF-C expression and secretion through the LRP6/IKK/IÒ¡B/NF-Ò¡B signaling pathway in gastric cancer cells. CONCLUSIONS: These findings demonstrated that kallistatin functions as an endogenous lymphangiogenesis inhibitor and has an important part in the lymphatic metastasis of gastric cancer.


Subject(s)
Lymphangiogenesis/physiology , Serpins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor C/metabolism , Aged , Animals , Cell Line, Tumor , Down-Regulation , Female , Humans , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Lymphatic Metastasis/pathology , Male , Mice, Inbred BALB C , Middle Aged , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Serpins/blood , Serpins/genetics , Serpins/pharmacology , Stomach Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...