Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 721
Filter
1.
Proc Natl Acad Sci U S A ; 119(33): e2206398119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35960844

ABSTRACT

During cell division, cross-linking motors determine the architecture of the spindle, a dynamic microtubule network that segregates the chromosomes in eukaryotes. It is unclear how motors with opposite directionality coordinate to drive both contractile and extensile behaviors in the spindle. Particularly, the impact of different cross-linker designs on network self-organization is not understood, limiting our understanding of self-organizing structures in cells but also our ability to engineer new active materials. Here, we use experiment and theory to examine active microtubule networks driven by mixtures of motors with opposite directionality and different cross-linker design. We find that although the kinesin-14 HSET causes network contraction when dominant, it can also assist the opposing kinesin-5 KIF11 to generate extensile networks. This bifunctionality results from HSET's asymmetric design, distinct from symmetric KIF11. These findings expand the set of rules underlying patterning of active microtubule assemblies and allow a better understanding of motor cooperation in the spindle.


Subject(s)
Kinesins , Microtubules , Oncogene Proteins , Spindle Apparatus , Cell Division , Humans , Kinesins/chemistry , Kinesins/physiology , Microtubules/chemistry , Microtubules/physiology , Oncogene Proteins/chemistry , Oncogene Proteins/physiology , Spindle Apparatus/chemistry , Spindle Apparatus/physiology
2.
Cell Rep ; 37(11): 110104, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34910916

ABSTRACT

Manipulation of energy-dissipating adipocytes has the potential to produce metabolic benefits. To this end, it is valuable to understand the mechanisms controlling the generation and function of thermogenic fat. Here, we identify Letm1 domain containing 1 (Letmd1) as a regulator of brown fat formation and function. The expression of Letmd1 is induced in brown fat by cold exposure and by ß-adrenergic activation. Letmd1-deficient mice exhibit severe cold intolerance concomitant with abnormal brown fat morphology, reduced thermogenic gene expression, and low mitochondrial content. The null mice exhibit impaired ß3-adrenoreceptor-dependent thermogenesis and are prone to diet-induced obesity and defective glucose disposal. Letmd1 was previously described as a mitochondrial protein, and we find that it also localizes to the nucleus and interacts with the transcriptional coregulator and chromatin remodeler Brg1/Smarca4, thus providing a way to impact thermogenic gene expression. Our study uncovers a role for Letmd1 as a key regulatory component of adaptive thermogenesis.


Subject(s)
Adipose Tissue, Brown/pathology , Energy Metabolism , Glucose/metabolism , Mitochondria/pathology , Oncogene Proteins/physiology , Receptors, Adrenergic, beta-3/metabolism , Receptors, Cell Surface/physiology , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Receptors, Adrenergic, beta-3/genetics
3.
FASEB J ; 35(11): e21965, 2021 11.
Article in English | MEDLINE | ID: mdl-34669999

ABSTRACT

Obesity and metabolic disorders caused by energy surplus pose an increasing concern within the global population. Brown adipose tissue (BAT) dissipates energy through mitochondrial non-shivering thermogenesis, thus representing a powerful agent against obesity. Here we explore the novel role of a mitochondrial outer membrane protein, LETM1-domain containing 1 (LETMD1), in BAT. We generated a knockout (Letmd1KO ) mouse model and analyzed BAT morphology, function and gene expression under various physiological conditions. While the Letmd1KO mice are born normally and have normal morphology and body weight, they lose multilocular brown adipocytes completely and have diminished mitochondrial abundance, DNA copy number, cristae structure, and thermogenic gene expression in the intrascapular BAT, associated with elevated reactive oxidative stress. In consequence, the Letmd1KO mice fail to maintain body temperature in response to acute cold exposure without food and become hypothermic within 4 h. Although the cold-exposed Letmd1KO mice can maintain body temperature in the presence of food, they cannot upregulate expression of uncoupling protein 1 (UCP1) and convert white to beige adipocytes, nor can they respond to adrenergic stimulation. These results demonstrate that LETMD1 is essential for mitochondrial structure and function, and thermogenesis of brown adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Mitochondria/metabolism , Oncogene Proteins/physiology , Receptors, Cell Surface/physiology , Thermogenesis , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism
4.
Int J Gynecol Cancer ; 30(8): 1224-1238, 2020 08.
Article in English | MEDLINE | ID: mdl-32571890

ABSTRACT

Elevated levels of replicative stress in gynecological cancers arising from uncontrolled oncogenic activation, loss of key tumor suppressors, and frequent defects in the DNA repair machinery are an intrinsic vulnerability for therapeutic exploitation. The presence of replication stress activates the DNA damage response and downstream checkpoint proteins including ataxia telangiectasia and Rad3 related kinase (ATR), checkpoint kinase 1 (CHK1), and WEE1-like protein kinase (WEE1), which trigger cell cycle arrest while protecting and restoring stalled replication forks. Strategies that increase replicative stress while lowering cell cycle checkpoint thresholds may allow unrepaired DNA damage to be inappropriately carried forward in replicating cells, leading to mitotic catastrophe and cell death. Moreover, the identification of fork protection as a key mechanism of resistance to chemo- and poly (ADP-ribose) polymerase inhibitor therapy in ovarian cancer further increases the priority that should be accorded to the development of strategies targeting replicative stress. Small molecule inhibitors designed to target the DNA damage sensors, such as inhibitors of ataxia telangiectasia-mutated (ATM), ATR, CHK1 and WEE1, impair smooth cell cycle modulation and disrupt efficient DNA repair, or a combination of the above, have demonstrated interesting monotherapy and combinatorial activity, including the potential to reverse drug resistance and have entered developmental pipelines. Yet unresolved challenges lie in balancing the toxicity profile of these drugs in order to achieve a suitable therapeutic index while maintaining clinical efficacy, and selective biomarkers are urgently required. Here we describe the premise for targeting of replicative stress in gynecological cancers and discuss the clinical advancement of this strategy.


Subject(s)
DNA Replication/drug effects , DNA/physiology , Genital Neoplasms, Female/drug therapy , Oncogene Proteins/physiology , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , DNA Damage , DNA Repair , Female , Genomic Instability , Humans , Molecular Targeted Therapy , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction
5.
Hum Cell ; 33(3): 790-800, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32304027

ABSTRACT

Anterior gradient 2 (AGR2) was proved to modulate cancer progression. However, the role of AGR2 on endometrial cancer was not established. Here, we investigated the effects of AGR2 expression on endometrial cancer and explored the regulation mechanism. In the study, we found that AGR2 was overexpressed in tumor tissues of 30 endometrial cancer patients. A high level of AGR2 promoted endometrial cancer cells proliferation, migration and invasion. AGR2 induced the expression of lactate dehydrogenase A (LDHA), phosphoglycerate kinase 1 (PGK1), kallikrein 2 (HK2), and enolase 1-α (ENO1), glucose uptake and lactate production. AGR2 could bind to MUC1 and induce MUC1 and hypoxia-inducible factor 1α (HIF-1α). The inhibition effects of AGR2 knockdown on cells proliferation, migration and invasion ability were abolished by the overexpression of MUC1. Besides, the overexpression of MUC1 also reversed the inhibition effects of AGR2 knockdown on the expression of LDHA, HK2, PGK1 and ENO1, glucose uptake and lactate production. AGR2 knockdown inhibited tumor growth, the levels of Ki-67, MUC1, HIF-1α and glycolysis. In conclusion, AGR2 was overexpressed in endometrial cancer and AGR2-induced glucose metabolism facilitated the progression of endometrial carcinoma via the MUC1/HIF-1α pathway. AGR2 may be an effective therapeutic target for endometrial carcinoma.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mucin-1/genetics , Mucin-1/metabolism , Mucoproteins/physiology , Oncogene Proteins/physiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Carcinoma/therapy , Cell Movement/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/therapy , Female , Gene Expression , Hexokinase/genetics , Hexokinase/metabolism , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Molecular Targeted Therapy , Mucoproteins/genetics , Mucoproteins/metabolism , Neoplasm Invasiveness/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphoglycerate Kinase/genetics , Phosphoglycerate Kinase/metabolism , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Hum Cell ; 33(3): 768-779, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32166565

ABSTRACT

Non-catalytic region of tyrosine kinase adaptor protein 1 (Nck1) is crucial for the progression of cancers. However, little is known on the role of Nck1 in the progression of ovarian carcinoma (OC). Here, we show that Nck1 expression is up-regulated in 176 OC tissues, compared with non-carcinoma ovarian tissues, and the up-regulated Nck1 expression is associated with the aggressiveness of OC and shorter overall and disease-free survival in this population. Higher Nck1 expression was an independent risk factor for poor prognosis of OC. Furthermore, Nck1 silencing by short hairpin RNA (shRNA) technology significantly inhibited the proliferation, migration and invasion of OC cells in vitro and the growth and metastasis of implanted OC tumors in vivo. Human kinase phosphorylation array indicated that Nck1 silencing significantly reduced the relative levels of 11 kinase expression and phosphorylation in OC cells, particularly for decreased levels of p70S6 kinase (p70S6K) and protein kinase B (AKT) expression in SKOV3 cells. Actually, Nck1 silencing significantly decreased PI3K and AKT expression, and reduced AKT and p70S6K phosphorylation while Nck1 over-expression had opposite effects in OC cells. Therefore, our data indicate that Nck1 promotes the progression of OC by enhancing the PI3k/AKT/p70S6K signaling in OC. Our findings suggest that Nck1 expression may be valuable for evaluating the prognosis of OC and as a target for design of new therapies for OC.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carcinoma/genetics , Oncogene Proteins/physiology , Ovarian Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/physiology , Carcinoma/therapy , Cell Line, Tumor , Disease Progression , Female , Humans , Molecular Targeted Therapy , Ovarian Neoplasms/therapy , Prognosis , Signal Transduction/genetics
7.
Life Sci ; 246: 117396, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32035130

ABSTRACT

AIMS: Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide. Decrease in NKG2D ligand levels and exhaustion of NK cells in HCC patients are major causes of immune escape, high recurrence, poor prognosis, and low overall survival. Enhancing the susceptibility of HCC to NK cells by upregulating NKG2DLs on tumor cells is an effective treatment strategy. This study aimed to identify the effect of the Anterior gradient 2 (AGR2)-derived peptide P1, which was reported to bind to HLA-A*0201 as an epitope, on both the expression of major histocompatibility complex class I-related chains A/B (MICA/B) on HCC cells and the cytotoxicity of NK cells. MAIN METHODS: The effect of P1 on MICA/B expression on HCC cells was determined by qRT-PCR, western blotting, and flow cytometry analysis. HCC cells were pre-treated with various pathway inhibitors to identify the molecular pathways associated with P1 treatment. The cytotoxicity of NK cells toward HCC was investigated by LDH cytotoxicity assay. The tumor-suppression effect of P1 was determined in vivo using a NOD/SCID mice HCC model. KEY FINDINGS: P1 significantly increased MICA/B expression on HCC cells, thereby enhancing their susceptibility to the cytotoxicity of NK cells in vitro and in vivo. Further, p38 MAPK cell signaling pathway inhibitor SB203580 significantly attenuated the effects of P1 in vivo and in vitro. SIGNIFICANCE: P1 upregulates MICA and MICB expression on HCC cells, thereby promoting their recognition and elimination by NK cells, which makes P1 an attractive novel immunotherapy agent.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Histocompatibility Antigens Class I/metabolism , Liver Neoplasms/metabolism , Mucoproteins/physiology , Oncogene Proteins/physiology , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Interferon-gamma/metabolism , Mice, Inbred NOD , Mice, SCID , Mucoproteins/metabolism , Neoplasm Transplantation , Oncogene Proteins/metabolism , Rats , Real-Time Polymerase Chain Reaction , Up-Regulation
8.
Cell Commun Signal ; 18(1): 26, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32059739

ABSTRACT

BACKGROUND: The limited options to treat obesity and its complications result from an incomplete understanding of the underlying molecular mechanisms regulating white adipose tissue development, including adipocyte hypertrophy (increase in size) and hyperplasia (increase in number through adipogenesis). We recently demonstrated that lack of the adaptor protein Nck1 in mice is associated with reduced adiposity and impaired adipocyte differentiation. In agreement, Nck1 depletion in 3 T3-L1 cells also attenuates adipocyte differentiation by enhancing PDGFRα activation and signaling. This is accompanied by higher expression of PDGF-A, a specific PDGFRα ligand, that may contribute to enhanced activation of PDGFRα signaling in the absence of Nck1 in white adipose tissue. However, whether Nck1 deficiency also impairs adipogenic differentiation in bone marrow still remains to be determined. METHODS: To address this point, Nck1-deficient derived bone marrow mesenchymal stem/stromal cells (BM-MSCs) and C3H10T1/2 mesenchymal stem cells were differentiated into adipocytes in vitro. Genes and proteins expression in these cellular models were determined using qPCR and western blotting respectively. Pharmacological approaches were used to assess a role for Nrf2 in mediating Nck1 deficiency effect on mesenchymal stem cells adipocyte differentiation. RESULTS: Nck1 deficiency in both BM-MSCs and C3H10T1/2 results in impaired adipocyte differentiation, accompanied by increased activation of the transcription factor Nrf2, as shown by increased mRNA levels of Nrf2 target genes, including PDGF-A. Using pharmacological activator and inhibitor of Nrf2, we further provide evidence that Nrf2 is an important player in PDGFRα signaling that mediates expression of PDGF-A and impaired adipogenesis in Nck1-deficient BM-MSCs and C3H10T1/2 cells. CONCLUSION: This study demonstrates that Nck1 deficiency in mesenchymal stem cells impairs adipogenesis through activation of the PDGFRα-Nrf2 anti-adipogenic signaling pathway. Video Abstract.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Mesenchymal Stem Cells , NF-E2-Related Factor 2/metabolism , Obesity , Oncogene Proteins/physiology , Platelet-Derived Growth Factor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adipogenesis , Animals , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Oncogene Proteins/genetics
9.
Exp Cell Res ; 382(1): 111442, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31128106

ABSTRACT

Oversecretion of Mucin5ac (MUC5AC), which is primarily synthesized by goblet cells and is the major gel-forming mucin, is a hallmark of various pulmonary inflammatory diseases. Hypoxia is considered a common pathophysiologic feature in various pulmonary inflammatory diseases. It has been suggested that hypoxia-inducible factor 1α (HIF-1α) acts as a key factor in hypoxia-induced MUC5AC hypersecretion; however, the exact mechanisms that maintain the stability of HIF-1α and support oversecretion by airway epithelial cells under hypoxia are still unclear. With immunohistochemistry, we found overexpression of anterior gradient 2 (AGR2) in the bronchial epithelial cells of hypoxia-treated mice. With specific shRNA transduction, AGR2 was demonstrated to be a key factor in MUC5AC hypersecretion in vitro. Additionally, co-immunoprecipitation, cell immunochemistry and confocal microscopy experiments were performed to explore the interaction between HIF-1α and AGR2 during hypoxia-induced MUC5AC hypersecretion in vitro. The results indicated increased binding and intracytoplasmic colocation of HIF-1α and AGR2. Our findings suggest that AGR2 acts as a key regulator in hypoxia-induced airway MUC5AC hypersecretion by increasing the stability of HIF-1α. Additionally, the elevated expression of AGR2 induced by hypoxia in bronchial epithelial cells likely depends on an XBP-1-associated pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mucin 5AC/metabolism , Mucoproteins/physiology , Oncogene Proteins/physiology , Signal Transduction/physiology , X-Box Binding Protein 1/physiology , Animals , Bronchi/cytology , Bronchi/metabolism , Cell Hypoxia , Cell Line , Cytoplasm/metabolism , Epithelial Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Interaction Mapping , RNA, Small Interfering/pharmacology , Random Allocation
10.
J Cell Biol ; 217(8): 2911-2929, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29921601

ABSTRACT

Septins are conserved components of the cytoskeleton that play important roles in many fundamental cellular processes including division, migration, and membrane trafficking. Septins can also inhibit bacterial infection by forming cage-like structures around pathogens such as Shigella We found that septins are recruited to vaccinia virus immediately after its fusion with the plasma membrane during viral egress. RNA interference-mediated depletion of septins increases virus release and cell-to-cell spread, as well as actin tail formation. Live cell imaging reveals that septins are displaced from the virus when it induces actin polymerization. Septin loss, however, depends on the recruitment of the SH2/SH3 adaptor Nck, but not the activity of the Arp2/3 complex. Moreover, it is the recruitment of dynamin by the third Nck SH3 domain that displaces septins from the virus in a formin-dependent fashion. Our study demonstrates that septins suppress vaccinia release by "entrapping" the virus at the plasma membrane. This antiviral effect is overcome by dynamin together with formin-mediated actin polymerization.


Subject(s)
Septins/physiology , Vaccinia/immunology , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/physiology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Cell Membrane/virology , Clathrin/analysis , Clathrin/metabolism , Dynamins/metabolism , Dynamins/physiology , HeLa Cells , Humans , Oncogene Proteins/metabolism , Oncogene Proteins/physiology , Phosphorylation , RNA Interference , Septins/analysis , Septins/antagonists & inhibitors , Signal Transduction , Virus Release/immunology
11.
EMBO J ; 37(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29724755

ABSTRACT

Helicobacter pylori infects half of the world's population, and strains that encode the cag type IV secretion system for injection of the oncoprotein CagA into host gastric epithelial cells are associated with elevated levels of cancer. CagA translocation into host cells is dependent on interactions between the H. pylori adhesin protein HopQ and human CEACAMs. Here, we present high-resolution structures of several HopQ-CEACAM complexes and CEACAMs in their monomeric and dimeric forms establishing that HopQ uses a coupled folding and binding mechanism to engage the canonical CEACAM dimerization interface for CEACAM recognition. By combining mutagenesis with biophysical and functional analyses, we show that the modes of CEACAM recognition by HopQ and CEACAMs themselves are starkly different. Our data describe precise molecular mechanisms by which microbes exploit host CEACAMs for infection and enable future development of novel oncoprotein translocation inhibitors and H. pylori-specific antimicrobial agents.


Subject(s)
Antigens, Bacterial/physiology , Antigens, CD/physiology , Bacterial Proteins/physiology , Cell Adhesion Molecules/physiology , Helicobacter pylori/physiology , Oncogene Proteins/physiology , Antigens, CD/chemistry , Bacterial Proteins/chemistry , Cell Adhesion Molecules/chemistry , HEK293 Cells , Humans , Mutagenesis , Protein Multimerization , Protein Transport
12.
Oncogene ; 37(25): 3329-3339, 2018 06.
Article in English | MEDLINE | ID: mdl-29551768

ABSTRACT

Chronic liver injury triggers liver fibrosis and hepatocellular carcinoma (HCC), the third leading cause of cancer-related mortality. Cyclin E1 (CcnE1, formerly designated Cyclin E) is a regulatory subunit of the Cyclin-dependent kinase 2 (CDK2). It is overexpressed in approximately 70% of human HCCs correlating with poor prognosis, while the relevance of its orthologue Cyclin E2 (CcnE2) is unclear. Hepatocyte-specific deletion of NF-kappa-B essential modulator (NEMOΔhepa) leads to chronic hepatitis, liver fibrosis, and HCC as well as CcnE upregulation. To this end, we generated NEMOΔhepa/CcnE1-/- and NEMOΔhepa/CcnE2-/- double knockout mice and investigated age-dependent liver disease progression in these animals. Deletion of CcnE1 in NEMOΔhepa mice decreased basal liver damage and reduced spontaneous liver inflammation in young mice. In contrast, loss of CcnE2 did not affect liver injury in NEMOΔhepa livers pointing to a unique, non-redundant function of CcnE1 in chronic hepatitis. Accordingly, basal compensatory hepatocyte proliferation in NEMOΔhepa mice was reduced by concomitant ablation of CcnE1, but not after loss of CcnE2. In aged NEMOΔhepa mice, loss of CcnE1 resulted in significant reduction of liver tumorigenesis, while deletion of CcnE2 had no effect on HCC formation. CcnE1, but not its orthologue CcnE2, substantially contributes to hepatic inflammatory response, liver disease progression, and hepatocarcinogenesis in NEMOΔhepa mice.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Cyclin E/physiology , Cyclins/physiology , Disease Models, Animal , Hepatitis/prevention & control , Intracellular Signaling Peptides and Proteins/physiology , Liver Neoplasms, Experimental/prevention & control , Liver/injuries , Liver/pathology , Oncogene Proteins/physiology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Hepatitis/etiology , Hepatitis/pathology , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29167347

ABSTRACT

The viral Bcl-2 homolog (vBcl2) of Kaposi's sarcoma-associated herpesvirus (KSHV) displays efficient antiapoptotic and antiautophagic activity through its central BH3 domain, which functions to prolong the life span of virus-infected cells and ultimately enhances virus replication and latency. Independent of its antiapoptotic and antiautophagic activity, vBcl2 also plays an essential role in KSHV lytic replication through its amino-terminal amino acids (aa) 11 to 20. Here, we report a novel molecular mechanism of vBcl2-mediated regulation of KSHV lytic replication. vBcl2 specifically bound the tegument protein open reading frame 55 (ORF55) through its amino-terminal aa 11 to 20, allowing their association with virions. Consequently, the vBcl2 peptide derived from vBcl2 aa 11 to 20 effectively disrupted the interaction between vBcl2 and ORF55, inhibiting the incorporation of the ORF55 tegument protein into virions. This study provides new insight into vBcl2's function in KSHV virion assembly that is separable from its inhibitory role in host apoptosis and autophagy.IMPORTANCE KSHV, an important human pathogen accounting for a large percentage of virally caused cancers worldwide, has evolved a variety of stratagems for evading host immune responses to establish lifelong persistent infection. Upon viral infection, infected cells can go through programmed cell death, including apoptosis and autophagy, which plays an effective role in antiviral responses. To counter the host response, KSHV vBcl2 efficiently blocks apoptosis and autophagy to persist for the life span of virus-infected cells. Besides its anti-programmed-cell-death activity, vBcl2 also interacts with the ORF55 tegument protein for virion assembly in infected cells. Interestingly, the vBcl2 peptide disrupts the vBcl2-ORF55 interaction and effectively inhibits KSHV virion assembly. This study indicates that KSHV vBcl2 harbors at least three genetically separable functions to modulate both host cell death signaling and virion production and that the vBcl2 peptide can be developed as an anti-KSHV therapeutic application.


Subject(s)
Herpesvirus 8, Human/physiology , Oncogene Proteins/physiology , Open Reading Frames , Viral Proteins/physiology , Virus Assembly , Apoptosis , Autophagy , Base Sequence , DNA Replication , DNA, Viral/genetics , Gene Expression , Gene Knockout Techniques , Genome, Viral , HEK293 Cells , Herpesvirus 8, Human/genetics , Humans , Oncogene Proteins/genetics , Viral Proteins/genetics
14.
J Biol Chem ; 292(52): 21264-21281, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29109143

ABSTRACT

The tumor microenvironment is characterized by nutrient-deprived conditions in which the cancer cells have to adapt for survival. Serum starvation resembles the growth factor deprivation characteristic of the poorly vascularized tumor microenvironment and has aided in the discovery of key growth regulatory genes and microRNAs (miRNAs) that have a role in the oncogenic transformation. We report here that miR-874 down-regulates the major G1/S phase cyclin, cyclin E1 (CCNE1), during serum starvation. Because the adaptation of cancer cells to the tumor microenvironment is vital for subsequent oncogenesis, we tested for miR-874 and CCNE1 interdependence in osteosarcoma cells. We observed that miR-874 inhibits CCNE1 expression in primary osteoblasts, but in aggressive osteosarcomas, miR-874 is down-regulated, leading to elevated CCNE1 expression and appearance of cancer-associated phenotypes. We established that loss of miR-874-mediated control of cyclin E1 is a general feature of osteosarcomas. The down-regulation of CCNE1 by miR-874 is independent of E2F transcription factors. Restoration of miR-874 expression impeded S phase progression, suppressing aggressive growth phenotypes, such as cell invasion, migration, and xenograft tumors, in nude mice. In summary, we report that miR-874 inhibits CCNE1 expression during growth factor deprivation and that miR-874 down-regulation in osteosarcomas leads to CCNE1 up-regulation and more aggressive growth phenotypes.


Subject(s)
Cyclin E/physiology , MicroRNAs/physiology , Oncogene Proteins/physiology , Osteosarcoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cyclin E/genetics , Cyclin G1/metabolism , Down-Regulation , G1 Phase Cell Cycle Checkpoints/genetics , G1 Phase Cell Cycle Checkpoints/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Oncogene Proteins/genetics , Oncogenes , Osteosarcoma/genetics , S Phase
15.
Microsc Microanal ; 23(1): 69-76, 2017 02.
Article in English | MEDLINE | ID: mdl-28162122

ABSTRACT

Cyclin E1 (CCNE1) is a core component of cell cycle regulation that drives the transition into the S phase. CCNE1 plays critical roles in cell cycle, cell proliferation, and cellular functions. However, the function of CCNE1 in early embryonic development is limited. In the present study, the function and expression of Ccne1 in porcine early parthenotes were examined. Immunostaining experiments showed that CCNE1 localized in the nucleus, starting at the four-cell stage. Knockdown of Ccne1 by double-stranded RNA resulted in the failure of blastocyst formation and induced blastocyst apoptosis. Ccne1 depletion increased expression of the pro-apoptotic gene Bax, and decreased the expression of Oct4 and the rate of inner cell mass (ICM)/trophectoderm formation. The results indicated that CCNE1 affects blastocyst formation by inducing cell apoptosis and ICM formation during porcine embryonic development.


Subject(s)
Cyclin E/pharmacology , Cyclin E/physiology , Embryonic Development/drug effects , Embryonic Development/physiology , Microscopy, Fluorescence/methods , Animals , Apoptosis/drug effects , Blastocyst/drug effects , Blastocyst Inner Cell Mass/drug effects , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Proliferation/drug effects , Cyclin E/genetics , Embryonic Stem Cells/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Octamer Transcription Factor-3/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Oncogene Proteins/physiology , Oocytes , RNA, Double-Stranded/analysis , Swine , bcl-2-Associated X Protein/metabolism
16.
Biochim Biophys Acta Bioenerg ; 1858(8): 615-627, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28087257

ABSTRACT

Until 1972, the term 'apoptosis' was used to differentiate the programmed cell death that naturally occurs in organismal development from the acute tissue death referred to as necrosis. Many studies on cell death and programmed cell death have been published and most are, at least to some degree, related to cancer. Some key proteins and molecular pathways implicated in cell death have been analyzed, whereas others are still being actively researched; therefore, an increasing number of cellular compartments and organelles are being implicated in cell death and cancer. Here, we discuss the mitochondria and subdomains of the endoplasmic reticulum (ER) that interact with mitochondria, the mitochondria-associated membranes (MAMs), which have been identified as critical hubs in the regulation of cell death and tumor growth. MAMs-dependent calcium (Ca2+) release from the ER allows selective Ca2+ uptake by the mitochondria. The perturbation of Ca2+ homeostasis in cancer cells is correlated with sustained cell proliferation and the inhibition of cell death through the modulation of Ca2+ signaling. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Subject(s)
Calcium/physiology , Mitochondria/physiology , Mitochondrial Membranes/physiology , Animals , Calcium Channels/physiology , Calcium Signaling/physiology , Cell Death , Cell Division , Cell Transformation, Neoplastic , Disease Progression , Endoplasmic Reticulum/metabolism , Homeostasis , Humans , Membrane Proteins/physiology , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins/physiology , Neoplasm Proteins/physiology , Oncogene Proteins/physiology , Signal Transduction
17.
Nat Commun ; 7: 13123, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759003

ABSTRACT

Docosahexaenoic acid (DHA, 22:6n-3) is an omega-3 fatty acid essential for proper brain development. N-docosahexaenoylethanolamine (synaptamide), an endogenous metabolite of DHA, potently promotes neurogenesis, neuritogenesis and synaptogenesis; however, the underlying molecular mechanism is not known. Here, we demonstrate orphan G-protein coupled receptor 110 (GPR110, ADGRF1) as the synaptamide receptor, mediating synaptamide-induced bioactivity in a cAMP-dependent manner. Mass spectrometry-based proteomic characterization and cellular fluorescence tracing with chemical analogues of synaptamide reveal specific binding of GPR110 to synaptamide, which triggers cAMP production with low nM potency. Disruption of this binding or GPR110 gene knockout abolishes while GPR110 overexpression enhances synaptamide-induced bioactivity. GPR110 is highly expressed in fetal brains but rapidly decreases after birth. GPR110 knockout mice show significant deficits in object recognition and spatial memory. GPR110 deorphanized as a functional synaptamide receptor provides a novel target for neurodevelopmental control and new insight into mechanisms by which DHA promotes brain development and function.


Subject(s)
Cognition/physiology , Docosahexaenoic Acids/metabolism , Endocannabinoids/physiology , Neurogenesis/physiology , Oncogene Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Brain/cytology , Cell Line , Cyclic AMP/metabolism , Endocannabinoids/metabolism , Female , Gene Knockout Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Models, Animal , Neurons/physiology , Oncogene Proteins/metabolism , Primary Cell Culture , Proteomics/methods , Rats , Receptors, G-Protein-Coupled/metabolism , Recognition, Psychology/physiology , Signal Transduction/physiology , Spatial Memory/physiology
18.
Gynecol Oncol ; 143(1): 152-158, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27461360

ABSTRACT

Cyclin E1 (CCNE1) gene amplification occurs in approximately 20% of ovarian high grade serous carcinoma (HGSC) and is associated with chemotherapy resistance and, in some studies, overall poor prognosis. The role of cyclin E1 in inducing S phase entry relies upon its interactions with cyclin dependent kinases (CDK), specifically CDK2. Therapies to target cyclin E1-related functions have centered on CDK inhibitors and proteasome inhibitors. While many studies have helped elucidate the functions and regulatory mechanisms of cyclin E1, further research utilizing cyclin E1 as a therapeutic target in ovarian cancer is warranted. This review serves to present the scientific background describing the role and function of cyclin E1 in cancer development and progression, to distinguish cyclin E1-amplified HGSC as a unique subset of ovarian cancer deserving of further therapeutic investigation, and to provide an updated overview on the studies which have utilized cyclin E1 as a target for therapy in ovarian cancer.


Subject(s)
Cyclin E/physiology , Cystadenocarcinoma, Serous/etiology , Oncogene Proteins/physiology , Ovarian Neoplasms/etiology , Cyclin E/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/physiology , Cystadenocarcinoma, Serous/therapy , Female , Humans , Oncogene Proteins/antagonists & inhibitors , Ovarian Neoplasms/therapy
19.
Neurobiol Aging ; 46: 43-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27460148

ABSTRACT

SET is a multifunctional protein, but when present in the cytoplasm, acts as a powerful inhibitor of phosphatase 2A. We previously observed that in CA1 of Down syndrome (DS) patients, the level of SET is increased, and SET is translocated to the cytoplasm and associated with the hyperphosphorylation of tau at ser202/thr205. The presence of SET in the cytoplasm in DS brains may play a role in the progression of the disease. Here, we show that in CA1 of 3-month-old Ts65Dn mice modeling DS, SET level is increased, and SET is translocated to the cytoplasm and associated with tau hyperphosphorylations at ser202/thr205 and with amyloid precursor protein caspase cleaved as observed in Alzheimer disease brains. Tau hyperphosphorylation at ser356 and activation of other phosphatase 2A targets such as the mammalian target of rapamycin and adenosine monophosphate protein kinases were also observed, suggesting deleterious mechanisms. We propose Ts65Dn mice as a model for therapeutic approaches focused on SET overexpression and its cytoplasmic translocation to slow down disease progression.


Subject(s)
CA1 Region, Hippocampal/metabolism , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/therapy , Oncogene Proteins/metabolism , Protein Transport , tau Proteins/chemistry , tau Proteins/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/cytology , Brain/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins , Gene Expression , Histone Chaperones , Male , Mice , Mice, Inbred Strains , Molecular Targeted Therapy , Oncogene Proteins/genetics , Oncogene Proteins/physiology , Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Serine , Sirolimus , Threonine
20.
Nat Rev Cancer ; 16(7): 413-30, 2016 07.
Article in English | MEDLINE | ID: mdl-27282250

ABSTRACT

The recent genomic characterization of cancers has revealed recurrent somatic point mutations and copy number changes affecting genes encoding RNA splicing factors. Initial studies of these 'spliceosomal mutations' suggest that the proteins bearing these mutations exhibit altered splice site and/or exon recognition preferences relative to their wild-type counterparts, resulting in cancer-specific mis-splicing. Such changes in the splicing machinery may create novel vulnerabilities in cancer cells that can be therapeutically exploited using compounds that can influence the splicing process. Further studies to dissect the biochemical, genomic and biological effects of spliceosomal mutations are crucial for the development of cancer therapies targeted at these mutations.


Subject(s)
Genes, Tumor Suppressor , Oncogene Proteins/physiology , RNA Splicing Factors/physiology , Catalysis , Humans , Neoplasms/drug therapy , Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...