Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Zookeys ; 1204: 191-198, 2024.
Article in English | MEDLINE | ID: mdl-38882564

ABSTRACT

Melanasterasinica He & Burckhardt, sp. nov., a new psylloid species developing on Grewia sp., is described from Hainan, China. It is the first Melanastera species reported from Asia and China, and the second species from the Old World. While New World species of Melanastera are mostly associated with the plant families Melastomataceae and Annonaceae, the two Old World species develop on the malvaceous Grewia, a host otherwise used in psylloids by two Haplaphalara species. The new species is described, diagnosed and illustrated, and its host plant and biogeographic ranges are discussed.

2.
J Clin Invest ; 134(8)2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38386414

ABSTRACT

Chromosomal instability is a prominent biological feature of myelodysplastic syndromes (MDS), with over 50% of patients with MDS harboring chromosomal abnormalities or a complex karyotype (CK). Despite this observation, the mechanisms underlying mitotic and chromosomal defects in MDS remain elusive. In this study, we identified ectopic expression of the transcription factor ONECUT3, which is associated with CKs and poorer survival outcomes in MDS. ONECUT3-overexpressing cell models exhibited enrichment of several notable pathways, including signatures of sister chromosome exchange separation and mitotic nuclear division with the upregulation of INCENP and CDCA8 genes. Notably, dysregulation of chromosome passenger complex (CPC) accumulation, besides the cell equator and midbody, during mitotic phases consequently caused cytokinesis failure and defective chromosome segregation. Mechanistically, the homeobox (HOX) domain of ONECUT3, serving as the DNA binding domain, occupied the unique genomic regions of INCENP and CDCA8 and transcriptionally activated these 2 genes. We identified a lead compound, C5484617, that functionally targeted the HOX domain of ONECUT3, inhibiting its transcriptional activity on downstream genes, and synergistically resensitized MDS cells to hypomethylating agents. This study revealed that ONECUT3 promoted chromosomal instability by transcriptional activation of INCENP and CDCA8, suggesting potential prognostic and therapeutic roles for targeting high-risk MDS patients with a CK.


Subject(s)
Myelodysplastic Syndromes , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Ectopic Gene Expression , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Chromosomal Instability , Karyotype
3.
Biomed Pharmacother ; 167: 115584, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37778270

ABSTRACT

Acute myeloid leukemia (AML) is a deadly hematological malignancy characterized by oncogenic translational addiction that results in over-proliferation and apoptosis evasion of leukemia cells. Various chemo- and targeted therapies aim to reverse this hallmark, but most show only modest efficacy. Here we report a single oral pill containing a low-dose triple small molecule-based cocktail, a highly active anti-cancer therapy (HAACT) with unique mechanisms that can effectively control AML. The cocktail comprises oncogenic translation inhibitor HHT, drug efflux pump P-gpi ENC and anti-apoptotic protein Bcl-2i VEN. Mechanistically, the cocktail can potently kill both leukemia stem cells (LSC) and bulk leukemic cells via co-targeting oncogenic translation, apoptosis machinery, and drug efflux pump, resulting in deep and durable remissions of AML in diverse model systems. We also identified EphB4/Bcl-xL as the cocktail response biomarkers. Collectively, our studies provide proof that a single pill containing a triple combination cocktail might be a promising avenue for AML therapy.


Subject(s)
Apoptosis , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/metabolism , Apoptosis Regulatory Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Line, Tumor
4.
Cancers (Basel) ; 14(19)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36230632

ABSTRACT

Acute myeloid leukaemia (AML) is one of the most common types of haematopoietic malignancy. Ribonucleotide reductase (RNR) is a key enzyme required for DNA synthesis and cell proliferation, and its small subunit RRM2 plays a key role for the enzymatic activity. We predicted monobenzone (MB) as a potential RRM2 target compound based on the crystal structure of RRM2. In vitro, MB inhibited recombinant RNR activity (IC50 = 0.25 µM). Microscale thermophoresis indicated that MB inhibited RNR activity by binding to RRM2. MB inhibited cell proliferation (MTT IC50 = 6-18 µM) and caused dose-dependent DNA synthesis inhibition, cell cycle arrest, and apoptosis in AML cells. The cell cycle arrest was reversed by the addition of deoxyribonucleoside triphosphates precursors, suggesting that RNR was the intracellular target of the compound. Moreover, MB overcame drug resistance to the common AML drugs cytarabine and doxorubicin, and treatment with the combination of MB and the Bcl-2 inhibitor ABT-737 exerted a synergistic inhibitory effect. Finally, the nude mice xenografts study indicated that MB administration produced a significant inhibitory effect on AML growth with relatively weak toxicity. Thus, we propose that MB has the potential as a novel anti-AML therapeutic agent in the future.

5.
Nat Commun ; 13(1): 2835, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35595767

ABSTRACT

Cyclin-dependent kinase 2 (CDK2) complex is significantly over-activated in many cancers. While it makes CDK2 an attractive target for cancer therapy, most inhibitors against CDK2 are ATP competitors that are either nonspecific or highly toxic, and typically fail clinical trials. One alternative approach is to develop non-ATP competitive inhibitors; they disrupt interactions between CDK2 and either its partners or substrates, resulting in specific inhibition of CDK2 activities. In this report, we identify two potential druggable pockets located in the protein-protein interaction interface (PPI) between CDK2 and Cyclin A. To target the potential druggable pockets, we perform a LIVS in silico screening of a library containing 1925 FDA approved drugs. Using this approach, homoharringtonine (HHT) shows high affinity to the PPI and strongly disrupts the interaction between CDK2 and cyclins. Further, we demonstrate that HHT induces autophagic degradation of the CDK2 protein via tripartite motif 21 (Trim21) in cancer cells, which is confirmed in a leukemia mouse model and in human primary leukemia cells. These results thus identify an autophagic degradation mechanism of CDK2 protein and provide a potential avenue towards treating CDK2-dependent cancers.


Subject(s)
Autophagy , CDC2-CDC28 Kinases , Cyclin A , Cyclin-Dependent Kinase 2 , Leukemia , Animals , Cell Line, Tumor/metabolism , Cyclin A/antagonists & inhibitors , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Homoharringtonine/metabolism , Homoharringtonine/pharmacology , Humans , Mice , Ribonucleoproteins
6.
Front Immunol ; 13: 888250, 2022.
Article in English | MEDLINE | ID: mdl-35592333

ABSTRACT

Tumor protein 53 (TP53) mutation predicts an unfavorable prognosis in diffuse large B-cell lymphoma (DLBCL), but the molecular basis for this association remains unclear. In several malignancies, the cytidine deaminase apolipoprotein B mRNA editing enzyme catalytic subunit 3B (APOBEC3B) has been reported to be associated with the TP53 G/C-to-A/T mutation. Here, we show that the frequency of this mutation was significantly higher in relapsed/refractory (R/R) than in non-R/R DLBCL, which was positively associated with the APOBEC3B expression level. APOBEC3B overexpression induced the TP53 G/C-to-A/T mutation in vitro, resulting in a phenotype similar to that of DLBCL specimens. Additionally, APOBEC3B-induced p53 mutants promoted the growth of DLBCL cells and enhanced drug resistance. These results suggest that APOBEC3B is a critical factor in mutant p53-driven R/R DLBCL and is therefore a potential therapeutic target.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Tumor Suppressor Protein p53 , Cytidine Deaminase/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Minor Histocompatibility Antigens/genetics , Mutation , Prognosis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Huan Jing Ke Xue ; 43(4): 1920-1929, 2022 Apr 08.
Article in Chinese | MEDLINE | ID: mdl-35393815

ABSTRACT

In order to better understand the water cycle processes in Baiyangdian Lake, samples for precipitation, river water, lake water, and groundwater were collected in 2020 via the analysis of oxygen and hydrogen isotopes and the total dissolved solids. A combined approach including correlation analysis, end-member modeling, and evaporation modeling was used to identify hydrogen and oxygen isotopic characteristics of different water types and their indicative significance to evaporation, hydrodynamics, and lake-groundwater interactions. The results showed that the compositions of hydrogen and oxygen isotopes in surface water and groundwater differed from each other. The average values of δD and δ18O were the highest in samples from lake water in June, secondary for lake water in October, and the lightest in groundwater. The slope of the lake water line was lower than that of the local meteoric water line, which could be attributed to a greater evaporative enrichment in lake water. The water loss ratio was estimated to be 18.8%-42.3% in June and 2.7%-30.3% in October by applying an evaporation model using deuterium excess. Lake water isotopes exhibited highly spatial heterogeneity, which indicated that Baiyangdian Lake was a poor-mixed lake controlled by the complex hydrodynamic conditions. The values of δD and δ18O were lighter around the estuary area and higher in the district far from the estuary. Although the main water isotopes in areas such as channels had a short residence time, the spatial differences in lake water isotopes were not remarkable, which suggested that the well-mixed conditions of the lake water contributed to reducing the heterogeneity of the lake water isotopic compositions on a spatial scale. The contribution ratios of lake leakage to groundwater were 0-91.7% and varied spatially. It had a negative relationship with the buried depth of groundwater and the distance to the lakeshore. No obvious relationship was found between lake leakage magnitude and groundwater level gradient. This study could provide a theoretical basis and technical support for the sustainable development of water resources and ecological environment protection in Baiyangdian Lake.


Subject(s)
Groundwater , Lakes , Environmental Monitoring/methods , Hydrogen/analysis , Lakes/analysis , Oxygen/analysis , Oxygen Isotopes/analysis , Water/analysis
8.
Adv Sci (Weinh) ; 9(8): e2104344, 2022 03.
Article in English | MEDLINE | ID: mdl-35048559

ABSTRACT

MYC oncogene is involved in the majority of human cancers and is often associated with poor outcomes, rendering it an extraordinarily desirable target, but therapeutic targeting of c-Myc protein has been a challenge for >30 years. Here, WBC100, a novel oral active molecule glue that selectively degrades c-Myc protein over other proteins and potently kills c-Myc overexpressing cancer cells is reported. WBC100 targets the nuclear localization signal 1 (NLS1)-Basic-nuclear localization signal 2 (NLS2) region of c-Myc and induces c-Myc protein degradation through ubiquitin E3 ligase CHIP mediated 26S proteasome pathway, leading to apoptosis of cancer cells. In vivo, WBC100 potently regresses multiple lethal c-Myc overexpressing tumors such as acute myeloid leukemia, pancreatic, and gastric cancers with good tolerability in multiple xenograft mouse models. Identification of the NLS1-Basic-NLS2 region as a druggable pocket for targeting the "undruggable" c-Myc protein and that single-agent WBC100 potently regresses c-Myc overexpressing tumors through selective c-Myc proteolysis opens new perspectives for pharmacologically intervening c-Myc in human cancers.


Subject(s)
Proto-Oncogene Proteins c-myc , Ubiquitin-Protein Ligases , Animals , Cell Line, Tumor , Humans , Mice , Proteolysis , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
9.
Signal Transduct Target Ther ; 6(1): 123, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33723219

ABSTRACT

The emergence of SARS-CoV-2 has resulted in the COVID-19 pandemic, leading to millions of infections and hundreds of thousands of human deaths. The efficient replication and population spread of SARS-CoV-2 indicates an effective evasion of human innate immune responses, although the viral proteins responsible for this immune evasion are not clear. In this study, we identified SARS-CoV-2 structural proteins, accessory proteins, and the main viral protease as potent inhibitors of host innate immune responses of distinct pathways. In particular, the main viral protease was a potent inhibitor of both the RLR and cGAS-STING pathways. Viral accessory protein ORF3a had the unique ability to inhibit STING, but not the RLR response. On the other hand, structural protein N was a unique RLR inhibitor. ORF3a bound STING in a unique fashion and blocked the nuclear accumulation of p65 to inhibit nuclear factor-κB signaling. 3CL of SARS-CoV-2 inhibited K63-ubiquitin modification of STING to disrupt the assembly of the STING functional complex and downstream signaling. Diverse vertebrate STINGs, including those from humans, mice, and chickens, could be inhibited by ORF3a and 3CL of SARS-CoV-2. The existence of more effective innate immune suppressors in pathogenic coronaviruses may allow them to replicate more efficiently in vivo. Since evasion of host innate immune responses is essential for the survival of all viruses, our study provides insights into the design of therapeutic agents against SARS-CoV-2.


Subject(s)
Immunity, Innate , Membrane Proteins/immunology , Nucleotidyltransferases/immunology , RNA, Viral/immunology , SARS-CoV-2/immunology , Signal Transduction/immunology , Viral Proteins/immunology , A549 Cells , Animals , Chickens , HEK293 Cells , HeLa Cells , Humans , Ligases/immunology , Mice
10.
Biomed Pharmacother ; 133: 111054, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33254022

ABSTRACT

Acute myeloid leukemia harboring internal tandem duplication of FMS-like tyrosine kinase 3 (FLT3-ITD AML) is a subset of highly aggressive malignancies with poor clinical outcome. Despite some advances in the development of FLT3 tyrosine kinase inhibitors (FLT3 inhibitors), most of FLT3-ITD AML patients suffer from lethal disease relapse, suggesting the requirement of novel targets and agents. Here we describe a natural small molecule, triptonide that can efficiently inhibit FLT3-ITD-driven AML in vitro and in vivo. Mechanistically, triptonide targeted Hedgehog/FLT3 signaling by inhibiting its critical effectors, which are GLI2, c-Myc and FLT3 and induced apoptosis of FLT3-ITD-driven leukemia cells. In addition, we also observed that triptonide activated tumor suppressor p53. In vivo, triptonide treatment markedly suppressed lethal FLT3-ITD-driven AML with good tolerance and prolonged survival time in orthotopic mouse model. Our studies identify Hedgehog/FLT3 axis as a novel target for treating FLT3-ITD-driven leukemia and demonstrate that triptonide is an active lead compound that can kill FLT3-ITD-driven leukemia cells.


Subject(s)
Antineoplastic Agents/pharmacology , Hedgehog Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Tandem Repeat Sequences , Triterpenes/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism , fms-Like Tyrosine Kinase 3/metabolism
11.
Aging (Albany NY) ; 12(23): 23647-23667, 2020 11 21.
Article in English | MEDLINE | ID: mdl-33226963

ABSTRACT

We previously defined the HERV-K Np9 as a viral oncogene. Here we report the discovery of a novel oncogene, Np17, which is homologous to the viral Np9 gene and predominantly present in Hominoidea. Np17 is located on chromosome 8, consists of 7 exons, and encodes a 16.8kDa nuclear protein with149 amino-acid residue. Functionally, knockdown of Np17 induced growth inhibition of leukemia cells, whereas enforced expression of Np17 promoted growth of leukemia cells in vitro and in vivo. In human leukemia, Np17 was detected in 59.65% (34/57) of acute myeloid leukemia (AML) patients examined and associated with refractory/relapsed AML. Mechanistically, Np17 decreased p53 levels and its mechanism might be involved in recruiting nuclear MDM2 to p53 for ubiquitin-mediated degradation. These findings reveal that Np17 is a novel oncogene associated with refractory/relapsed leukemia.


Subject(s)
Leukemia/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins/metabolism , Animals , Case-Control Studies , Cell Proliferation , Female , Gene Expression Regulation, Leukemic , HL-60 Cells , Humans , K562 Cells , Leukemia/genetics , Leukemia/pathology , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Oncogene Proteins/genetics , Proteolysis , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , THP-1 Cells , Tumor Suppressor Protein p53/metabolism , Ubiquitination
12.
J Cancer ; 11(19): 5727-5737, 2020.
Article in English | MEDLINE | ID: mdl-32913466

ABSTRACT

Cell division cycle associated (CDCA) gene family plays an important role in cells. However, some researchers revealed that overexpression of CDCAs might contribute to the tumor progression in several cancers. Here, we analyzed the role of this gene family in hepatocellular carcinoma (HCC). We used several web tools and found that most of CDCAs were highly expressed in tumor tissues compared to the paracancer tissues in HCC. We then used RT-qPCR to confirm our results. The results showed that CDCA2, CDCA3, CDCA5 and CDCA8 were up-regulated in HCC. We also found that these genes were associated with poor overall survival and relapse free survival except CDCA7. The functional analysis showed that this gene family might take part in many processes, including cell division, apoptosis, DNA damage and DNA repair, which might contribute to the tumor progression. The KEGG pathway analysis showed that these genes participated in several important pathways such as PI3K-Akt signaling pathway and hippo signaling pathway. In conclusion, our findings suggested that CDCA2, CDCA3, CDCA4, CDCA5, and CDCA8 might have potential diagnostic and prognostic values for hepatocellular carcinoma.

13.
Aging (Albany NY) ; 12(14): 15037-15049, 2020 07 26.
Article in English | MEDLINE | ID: mdl-32713851

ABSTRACT

Osteosarcoma is the most common malignant bone tumor in children and young adults, and it has a survival rate of only 60% with current cytotoxic chemotherapy combined with aggressive surgery. The aim of this study was to evaluate the therapeutic efficacy of the berbamine derivative 2-methylbenzoyl berbamine (BBD24) for osteosarcoma in vitro and in vivo. We used human osteosarcoma cell lines, primary osteosarcoma cells and mouse models to evaluate the inhibitory effects of BBD24 on osteosarcoma and to determine the molecular mechanism. Our results showed that BBD24 inhibited the growth of the human osteosarcoma cell lines HOS and MG63 in a time- and dose-dependent manner. BBD24 also exhibited significant inhibitory effects on primary osteosarcoma cells. In contrast, BBD24 did not affect normal blood cells under the same conditions. Treatment with BBD24 induced apoptosis, necrosis and autophagy in osteosarcoma cells. Western blot analysis revealed that BBD24 activated the caspase-dependent pathway and downregulated the NF-kB, AKT, and ERK pathways. Finally, BBD24 treatment induced a significant inhibitory effect on the growth of osteosarcoma in nude mice. Our findings indicate that BBD24 is a multitarget inhibitor and may represent a new type of anticancer agent for osteosarcoma treatment.


Subject(s)
Benzylisoquinolines/pharmacology , Bone Neoplasms , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Osteosarcoma , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/pathology
14.
Aging (Albany NY) ; 12(13): 13668-13683, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32658867

ABSTRACT

Aberrant activation of CAMKIIγ has been linked to leukemia and T-cell lymphoma, but not multiple myeloma (MM). The purpose of this study was to explore the role of CaMKIIγ in the pathogenesis and therapy of MM. In this study, we found that CaMKIIγ was aberrantly activated in human MM and its expression level was positively correlated with malignant progression and poor prognosis. Ectopic expression of CaMKIIγ promoted cell growth, colony formation, cell cycle progress and inhibited apoptosis of MM cell lines, whereas, knockdown of CAMKIIγ expression suppressed MM cell growth in vitro and in vivo. Mechanically, we observed that CaMKIIγ overexpression upregulated p-ERK and p-Stat3 levels and suppression of CaMKIIγ had opposite effects. CaMKIIγ is frequently dysregulated in MM and plays a critical role in maintaining MM cell growth through upregulating STAT3 signaling pathway. Furthermore, our preclinical studies suggest that CaMKIIγ is a potential therapeutic target in MM, and could be intervened pharmacologically by small-molecule berbamine analogues.


Subject(s)
Benzylisoquinolines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Multiple Myeloma/pathology , STAT3 Transcription Factor/metabolism , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Benzylisoquinolines/therapeutic use , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , MAP Kinase Signaling System/drug effects , Male , Mice , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Neoplasm Staging , Phosphorylation/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
15.
Nat Microbiol ; 4(12): 2552-2564, 2019 12.
Article in English | MEDLINE | ID: mdl-31659299

ABSTRACT

Innate immunity is the first line of host defence against pathogens. Suppression of innate immune responses is essential for the survival of all viruses. However, the interplay between innate immunity and HIV/SIV is only poorly characterized. We have discovered Vpx as a novel inhibitor of innate immune activation that associates with STING signalosomes and interferes with the nuclear translocation of NF-κB and the induction of innate immune genes. This new function of Vpx could be separated from its role in mediating degradation of the antiviral factor SAMHD1, and is conserved among diverse HIV-2/SIV Vpx. Vpx selectively suppressed cGAS-STING-mediated nuclear factor-κB signalling. Furthermore, Vpx and Vpr had complementary activities against cGAS-STING activity. Since SIVMAC lacking both Vpx and Vpr was less pathogenic than SIV deficient for Vpr or Vpx alone, suppression of innate immunity by HIV/SIV is probably a key pathogenic determinant, making it a promising target for intervention.


Subject(s)
HIV-2/metabolism , Membrane Proteins/metabolism , NF-kappa B/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Simian Immunodeficiency Virus/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Adult , Amino Acid Sequence , Animals , Disease Models, Animal , Female , HEK293 Cells , HIV Infections/immunology , HIV Infections/virology , HIV-2/genetics , HeLa Cells , Humans , Immunity, Innate/genetics , Male , Mice, Inbred BALB C , SAM Domain and HD Domain-Containing Protein 1/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/immunology , Young Adult , vpr Gene Products, Human Immunodeficiency Virus/immunology , vpr Gene Products, Human Immunodeficiency Virus/metabolism
16.
Exp Cell Res ; 384(2): 111653, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31574286

ABSTRACT

Acute myeloid leukemia (AML) is a group of highly aggressive malignancies with a 5-year overall survival of less than 40%. Cell overgrowth with defective apoptosis is a hallmark of AML, but little is known about how it occurs. Here, we show that aberrant activation of the largest subunit of RNA polymerase II (RPB1) encoded by POLR2A gene is critically involved in this hallmark. We retrospectively analyzed the expression profiles of POLR2A and RPB1 in a panel of AML cell lines, primary AML patients and peripheral blood samples. Meanwhile, correlation analysis was used to explore the correlation between the expression of RPB1 with tumor burden and overall survival time in untreated AML samples. RNA-Seq approach was performed to identify the differentially expressed genes between RPB1 silencing AML cells with control cells after knocking out RPB1. Furthermore, orthotopic AML models were established with RPB1 silencing and control cells to investigate the effects of RPB1 protein level on leukemia cell growth. In most AML patients, RPB1 was aberrantly activated and closely associated with poor prognosis, but not in normal hematopoietic cells. Global transcriptomic analysis revealed that POLR2A knockout strongly impaired growth of AML cells by selectively depleting a substantial set of AML-related oncogenic and anti-apoptosis genes such as MYC, RUNX2, MEIS1, CDC25A and BCL-2. Silencing RPB1 by genetic technology led to a potent regression of human refractory AML in mouse models. These findings reveal that dysregulated RPB1 is a central oncogenic hub that drives overgrowth by hijacking an array of oncogenic and anti-apoptosis factors. Targeting RPB1 is a potential therapeutic for treating AML.


Subject(s)
Cell Proliferation/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , RNA Polymerase II/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Cell Line , Cell Line, Tumor , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Leukemic/genetics , HEK293 Cells , HL-60 Cells , Humans , Mice , Retrospective Studies , THP-1 Cells
17.
Biomed Pharmacother ; 117: 109134, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31247466

ABSTRACT

Proto-oncogene Myc, a key transcription factor, is frequently deregulated in human leukemia with aggressive and poor clinical outcome, but the development of MYC inhibitors remains challenging due to MYC helix-loop-helix topology lacking druggable domains. Here we describe a novel oral active small molecule analog of berbamine, tosyl chloride-berbamine (TCB), that efficiently eliminates MYC-positive leukemia in vitro and in vivo. Mechanistically, TCB potently reduced MYC protein by inhibiting CaMKIIγ, a critical enzyme that stabilizes MYC protein, and induces apoptosis of MYC-positive leukemia cells. In vivo, oral administration of TCB markedly eliminated lethal MYC-positive acute lymphoblastic leukemia (ALL) with well tolerability in orthotopic mouse model. Our studies identify CaMKIIγ/Myc axis as a valid target for developing small molecule-based new therapies for treating MYC-mediated leukemia and demonstrate that TCB is an orally active analog of berbamine that kills MYC-positive leukemia cells.


Subject(s)
Benzylisoquinolines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Leukemia/pathology , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tosyl Compounds/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzylisoquinolines/administration & dosage , Benzylisoquinolines/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Inhibitory Concentration 50 , Leukemia/enzymology , Mice , Proto-Oncogene Mas , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tosyl Compounds/administration & dosage , Tosyl Compounds/chemistry , Transcription, Genetic/drug effects
18.
Transl Oncol ; 12(6): 801-809, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30953928

ABSTRACT

As a heterogeneous group of clonal disorders, acute myeloid leukemia with internal tandem duplication of fms-like tyrosine kinase 3 (FLT3-ITD) mutation usually shows an inferior prognosis. In the present study, we found that homoharringtonine (HHT), a protein translation inhibitor of plant alkaloid in China, exhibited potent cytotoxic effect against FLT3-ITD (+) cell lines and primary leukemia cells, and a remarkable synergistic anti-leukemia action was demonstrated in vitro and in vivo in xenograft mouse models when co-treated with the heat shock protein 90 inhibitor IPI504. Mechanistically, HHT combined with IPI504 synergistically inhibited the growth of leukemia cells by inducing apoptosis and G1 phase arrest. This synergistic action resulted in a prominent reduction of total and phosphorylated FLT3 (p-FLT3) as well as inhibition of its downstream signaling molecules such as STAT5, AKT, ERK and 4E-BP1. Furthermore, co-treatment of HHT and IPI504 led to a synergistic or additive effect on 55.56%(10/18) of acute myeloid leukemia cases tested, including three relapsed/refractory patients. In conclusion, our findings indicate that the combination of HHT and HSP90 inhibitor provides an alternative way for the treatment of FLT3-ITD positive acute myeloid leukemia, especially for relapsed/refractory AML.

19.
Exp Cell Res ; 371(2): 409-416, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30179602

ABSTRACT

Identifying and targeting oncogenic fusion genes have revolutionized the treatment of leukemia, such as PML-RARα fusion gene in acute promyelocytic leukemia. Here we identified an intrachromosomal fusion gene located on chromosome 19q.13 between UBA2 and WTIP gene in a case of acute myeloid leukemia. The UBA2-WTIP fusion gene contains the N-terminal E1_enzyme_family, VAE_Ubl domains of UBA2, and the C-terminal LIM domains of WTIP. The UBA2-WTIP fusion was detected by reverse transcriptase polymerase chain reaction and Sanger sequencing in 19 of 56 acute myeloid leukemia samples (33.9%). Ectopic expression of the UBA2-WTIP fusion in human acute myeloid leukemia KG-1a cells showed enhanced cell proliferation both in vitro and in vivo. The UBA2-WTIP fusion induced phosphorylation of STAT3, STAT5 and ERK1/2, and abrogates WTIP-mediated mammalian processing body formation. Finally, triptolide displayed selective cytotoxicity against KG-1a cells harboring the UBA2-WTIP fusion. Collectively, our findings suggest that the UBA2-WTIP fusion is an oncogenic fusion gene, as well as a promising therapeutic target for the treatment of acute myeloid leukemia.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Leukocytes/metabolism , Oncogene Proteins, Fusion/genetics , Ubiquitin-Activating Enzymes/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Co-Repressor Proteins , Cytoskeletal Proteins , Diterpenes/pharmacology , Epoxy Compounds/pharmacology , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Leukocytes/drug effects , Leukocytes/pathology , Mice , Mice, Inbred NOD , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Proteins, Fusion/metabolism , Phenanthrenes/pharmacology , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Survival Analysis , Ubiquitin-Activating Enzymes/metabolism , Xenograft Model Antitumor Assays
20.
Ann Hematol ; 97(12): 2353-2362, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30099568

ABSTRACT

C-Myc expression is associated with poor prognosis and aggressive progression of diffuse large B cell lymphoma (DLBCL), and the development of drug-like c-Myc inhibitors remains challenging. In this study, we report a novel berbamine derivative termed 4-chlorobenzoyl berbamine (CBBM) that potently induced the apoptosis of c-Myc-overexpressing DLBCL cells but spared normal blood cells. The compound showed IC50 values ranging from 1.93 to 3.89 µmol/L in DLCBL cells and exhibited a 4.75- to 9.64-fold increase in anti-tumor activity compared to berbamine. Additionally, CBBM inhibited the proliferation of the DLBCL line OCI-Ly3 cells through G0/G1 cell-cycle arrest and induced apoptosis. Further studies have shown that CBBM treatment leads to the proteasome-dependent degradation of c-Myc protein in OCI-Ly3 cells. Interestingly, we found that the inhibitory effect of CBBM was positively correlated with basal levels of CaMKIIγ, which is a key inducer of c-Myc expression in DLBCL cells. We also observed that CBBM inhibits the JAK2/STAT3 pathway, leading to reduced c-Myc transcription. Collectively, these findings suggest that CBBM could be a promising lead compound for treatment of c-Myc-driven DLBCL.


Subject(s)
Apoptosis/drug effects , Benzylisoquinolines , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma, Large B-Cell, Diffuse , Proto-Oncogene Proteins c-myc/biosynthesis , Signal Transduction/drug effects , Benzylisoquinolines/chemical synthesis , Benzylisoquinolines/chemistry , Benzylisoquinolines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Janus Kinase 2/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...