Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Pathol ; 260(1): 17-31, 2023 05.
Article in English | MEDLINE | ID: mdl-36715683

ABSTRACT

Macropinocytosis is an effective strategy to mitigate nutrient starvation. It can fuel cancer cell growth in nutrient-limited conditions. However, whether and how macropinocytosis contributes to the rapid proliferation of hepatocellular carcinoma cells, which frequently experience an inadequate nutrient supply, remains unclear. Here, we demonstrated that nutrient starvation strongly induced macropinocytosis in some hepatocellular carcinoma cells. It allowed the cells to acquire extracellular nutrients and supported their energy supply to maintain rapid proliferation. Furthermore, we found that the phospholipid flippase ATP9A was critical for regulating macropinocytosis in hepatocellular carcinoma cells and that high ATP9A levels predicted a poor outcome for patients with hepatocellular carcinoma. ATP9A interacted with ATP6V1A and facilitated its transport to the plasma membrane, which promoted plasma membrane cholesterol accumulation and drove RAC1-dependent macropinocytosis. Macropinocytosis inhibitors significantly suppressed the energy supply and proliferation of hepatocellular carcinoma cells characterised by high ATP9A expression under nutrient-limited conditions. These results have revealed a novel mechanism that overcomes nutrient starvation in hepatocellular carcinoma cells and have identified the key regulator of macropinocytosis in hepatocellular carcinoma. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , Cell Membrane , Liver Neoplasms/metabolism , Nutrients , Phospholipids/metabolism
2.
J Phys Chem A ; 125(39): 8626-8634, 2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34585571

ABSTRACT

This paper presents the reaction mechanism, cross sections, and product energy partitioning for the O + CO2 reaction, calculated using Born-Oppenheimer molecular dynamics simulations with the quasiclassical trajectory (BOMD-QCT) method. At collision energies up to 9.5 eV, three reactions, oxygen exchange (above ∼1.5 eV), abstraction (above ∼5.5 eV), and dissociation (above ∼7.5 eV) occur, with abstraction and dissociation involving either an insertion-elimination mechanism or a stripping mechanism. The insertion-elimination mechanism involves the formation of a planar CO3 intermediate which lies 0.52 eV above the ground-state CO2; the energetic barrier for oxygen abstraction via this mechanism is 3.52 eV. Interestingly, the insertion-elimination mechanism predominately contributes to the cross sections at collision energies just above the effective energetic threshold for the abstraction and dissociation reactions; at higher collision energies, the contribution from the stripping mechanism increases and eventually dominates. At a collision energy of 9.5 eV, the cross sections for oxygen exchange, abstraction, and dissociation are 4.17 a02, 1.58 a02, and 0.68 a02, respectively. The lower reaction cross sections, higher effective reaction barrier, and product energy distribution of the stripping mechanism were attributed to the short lifetime (28 fs) of the OCOO species compared with that of the CO3 species (45 fs) that arises in the insertion-elimination mechanism. For the exchange reaction, it is found that roughly 40% of the reactant translational energy ends up in CO2 vibration, which provides a single-collision mechanism to produce highly excited CO2. We also studied intersystem crossing effects using trajectory surface hopping calculations and find no changes compared to single surface (triplet) calculations at energies below 7.5 eV; however, at 7.5 eV and higher the abstraction cross sections are changed by as much as 20%, and the (very small) dissociation cross sections are changed by factors of four or more.

3.
Nano Lett ; 19(3): 2106-2113, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30763517

ABSTRACT

Electrochemical atomic force microscopy tip-enhanced Raman spectroscopy (EC-AFM-TERS) was used for the first time to spatially resolve local heterogeneity in redox behavior on an electrode surface in situ and at the nanoscale. A structurally well-defined Au(111) nanoplate located on a polycrystalline ITO substrate was studied to examine nanoscale redox contrast across the two electrode materials. By monitoring the TERS intensity of adsorbed Nile Blue (NB) molecules on the electrode surface, TERS maps were acquired with different applied potentials. The EC-TERS maps showed a spatial contrast in TERS intensity between Au and ITO. TERS line scans near the edge of a 20 nm-thick Au nanoplate demonstrated a spatial resolution of 81 nm under an applied potential of -0.1 V vs Ag/AgCl. The intensities from the TERS maps at various applied potentials followed Nernstian behavior, and a formal potential ( E0') map was constructed by fitting the TERS intensity at each pixel to the Nernst equation. Clear nanoscale spatial contrast between the Au and ITO regions was observed in the E0' map. In addition, statistical analysis of the E0' map identified a statistically significant 4 mV difference in E0' on Au vs ITO. Electrochemical heterogeneity was also evident in the E0' distribution, as a bimodal distribution was observed in E0' on polycrystalline ITO, but not on gold. A direct comparison between an AFM friction image and the E0' map resolved the electrochemical behavior of individual ITO grains with a spatial resolution of ∼40 nm. The variation in E0' was attributed to different local surface charges on the ITO grains. Such site-specific electrochemical information with nanoscale spatial and few mV voltage resolutions is not available using ensemble spectroelectrochemical methods. We expect that in situ redox mapping at the nanoscale using EC-AFM-TERS will have a crucial impact on understanding the role of nanoscale surface features in applications such as electrocatalysis.

4.
Nano Lett ; 19(10): 7309-7316, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31518135

ABSTRACT

The plasmonic properties of tip-substrate composite systems are of vital importance to near-field optical spectroscopy, in particular tip-enhanced Raman spectroscopy (TERS), which enables operando studies of nanoscale chemistry at a single molecule level. The nanocavities formed in the tip-substrate junction also offer a highly tunable platform for studying field-matter interactions at the nanoscale. While the coupled nanoparticle dimer model offers a correct qualitative description of gap-mode plasmon effects, it ignores the full spectrum of multipolar tip plasmon modes and their interaction with surface plasmon polariton (SPP) excitation in the substrate. Herein, we perform the first tip-enhanced Raman excitation spectroscopy (TERES) experiment and use the results, both in ambient and aqueous media, in combination with electrodynamics simulations, to explore the plasmonic response of a Au tip-Au substrate composite system. The gap-mode plasmon features a wide spectral window corresponding to a host of tip plasmon modes interacting with the plasmonic substrate. Simulations of the electric field confinement demonstrate that optimal spatial resolution is achieved when a hybrid plasmon mode that combines a multipolar tip plasmon and a substrate SPP is excited. Nevertheless, a wide spectral window over 1000 nm is available for exciting the tip plasmon with high spatial resolution, which enables the simultaneous resonant detection of different molecular species. This window is robust as a function of tip-substrate distance and tip radius of curvature, indicating that many choices of tips will work, but it is restricted to wavelengths longer than ∼600 nm for the Au tip-Au substrate combination. Other combinations, such as Ag tip-Ag substrate, can access wavelengths as low as 350 nm.

5.
Angew Chem Int Ed Engl ; 59(31): 13037-13043, 2020 Jul 27.
Article in English | MEDLINE | ID: mdl-32347593

ABSTRACT

We investigate a family of dinuclear dysprosium metallocene single-molecule magnets (SMMs) bridged by methyl and halogen groups [Cp'2 Dy(µ-X)]2 (Cp'=cyclopentadienyltrimethylsilane anion; 1: X=CH3 - ; 2: X=Cl- ; 3: X=Br- ; 4: X=I- ). For the first time, the magnetic easy axes of dysprosium metallocene SMMs are experimentally determined, confirming that the orientation of them are perpendicular to the equatorial plane which is made up of dysprosium and bridging atoms. The orientation of the magnetic easy axis for 1 deviates from the normal direction (by 10.3°) due to the stronger equatorial interactions between DyIII and methyl groups. Moreover, its magnetic axes show a temperature-dependent shifting, which is caused by the competition between exchange interactions and Zeeman interactions. Studies of fluorescence and specific heat as well as ab initio calculations reveal the significant influences of the bridging ligands on their low-lying exchange-based energy levels and, consequently, low-temperature magnetic properties.

6.
J Am Chem Soc ; 139(1): 373-380, 2017 01 11.
Article in English | MEDLINE | ID: mdl-27936686

ABSTRACT

The pursuit of single-molecule magnets (SMMs) with better performance urges new molecular design that can endow SMMs larger magnetic anisotropy. Here we report that two-coordinate cobalt imido complexes featuring highly covalent Co═N cores exhibit slow relaxation of magnetization under zero direct-current field with a high effective relaxation barrier up to 413 cm-1, a new record for transition metal based SMMs. Two theoretical models were carried out to investigate the anisotropy of these complexes: single-ion model and Co-N coupling model. The former indicates that the pseudo linear ligand field helps to preserve the first-order orbital momentum, while the latter suggests that the strong ferromagnetic interaction between Co and N makes the [CoN]+ fragment a pseudo single paramagnetic ion, and that the excellent performance of these cobalt imido SMMs is attributed to the inherent large magnetic anisotropy of the [CoN]+ core with |MJ = ± 7/2⟩ ground Kramers doublet.

7.
J Clin Invest ; 133(14)2023 07 17.
Article in English | MEDLINE | ID: mdl-37463446

ABSTRACT

Human epidermal growth factor receptor 2-targeted (HER2-targeted) therapy is the mainstay of treatment for HER2+ breast cancer. However, the proteolytic cleavage of HER2, or HER2 shedding, induces the release of the target epitope at the ectodomain (ECD) and the generation of a constitutively active intracellular fragment (p95HER2), impeding the effectiveness of anti-HER2 therapy. Therefore, identifying key regulators in HER2 shedding might provide promising targetable vulnerabilities against resistance. In the current study, we found that upregulation of dolichyl-phosphate N-acetylglucosaminyltransferase (DPAGT1) sustained high-level HER2 shedding to confer trastuzumab resistance, which was associated with poor clinical outcomes. Upon trastuzumab treatment, the membrane-bound DPAGT1 protein was endocytosed via the caveolae pathway and retrogradely transported to the ER, where DPAGT1 induced N-glycosylation of the sheddase - ADAM metallopeptidase domain 10 (ADAM10) - to ensure its expression, maturation, and activation. N-glycosylation of ADAM10 at N267 protected itself from ER-associated protein degradation and was essential for DPAGT1-mediated HER2 shedding and trastuzumab resistance. Importantly, inhibition of DPAGT1 with tunicamycin acted synergistically with trastuzumab treatment to block HER2 signaling and reverse resistance. These findings reveal a prominent mechanism for HER2 shedding and suggest that targeting DPAGT1 might be a promising strategy against trastuzumab-resistant breast cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction , Membrane Proteins/metabolism , Drug Resistance, Neoplasm , Cell Line, Tumor , Antineoplastic Agents/pharmacology
8.
Theranostics ; 13(1): 339-354, 2023.
Article in English | MEDLINE | ID: mdl-36593950

ABSTRACT

Rationale: Chemoresistance is a major challenge in the clinical management of patients with breast cancer. Mutant p53 proteins tend to form aggregates that promote tumorigenesis in cancers. We here aimed to explore the mechanism for the generation of mutant p53 aggregates in breast cancer and assess its role in inducing chemoresistance. Methods: Expression of BCL2-associated athanogene 2 (BAG2) was evaluated by qRT-PCR, western blotting, and immunohistochemistry in breast cancer patient specimens. The significance of BAG2 expression in prognosis was assessed by Kaplan-Meier survival analysis and the Cox regression model. The roles of BAG2 in facilitating the formation of mutant p53 aggregates were analyzed by co-immunoprecipitation, immunofluorescence, and semi-denaturing detergent-agarose gel electrophoresis assays. The effects of BAG2 on the chemoresistance of breast cancer were demonstrated by cell function assays and mice tumor models. Results: In the present study, we found that BAG2 was significantly upregulated in relapse breast cancer patient tissues and high BAG2 was associated with a worse prognosis. BAG2 localized in mutant p53 aggregates and interacted with misfolded p53 mutants. BAG2 exacerbated the formation of the aggregates and recruited HSP90 to promote the propagation and maintenance of the aggregates. Consequently, BAG2-mediated mutant p53 aggregation inhibited the mitochondrial apoptosis pathway, leading to chemoresistance in breast cancer. Importantly, silencing of BAG2 or pharmacological targeting of HSP90 substantially reduced the aggregates and increased the sensitivity of chemotherapy in breast cancer. Conclusion: These findings reveal a significant role of BAG2 in the chemoresistance of breast cancer via exacerbating mutant p53 aggregates and suggest that BAG2 may serve as a potential therapeutic target for breast cancer patients with drug resistance.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Molecular Chaperones , Tumor Suppressor Protein p53 , Animals , Mice , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , HSP90 Heat-Shock Proteins/metabolism , Neoplasm Recurrence, Local , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Breast Neoplasms/genetics , Humans , Female
9.
Discov Oncol ; 13(1): 81, 2022 Aug 24.
Article in English | MEDLINE | ID: mdl-36002694

ABSTRACT

BACKGROUND: Distant metastasis is the prominent factor for cancer-induced death of gastric cancer in which peritoneum is one of the dominating targets of gastric cancer metastasis. However, there is still a lack of effective predictive indicators and treatment methods for gastric cancer patients with peritoneal metastasis. METHODS: A clustering assay was used to investigate the cell aggregates formation ability. While the soft agar assay and anoikis assay were performed to detect the anchorage-independent growth and anoikis-resistant ability respectively. Luciferase activity assay, western blotting and immunofluorescence were used to explore the effect of HMMR on AKT signaling activity. The peritoneal implantation model was examined to explore the role of HMMR in vivo. RESULTS: Silencing of HMMR expression markedly reduced the peritoneal metastasis of gastric cancer cells through reducing cell-cell interactions. Mechanistically, HA-HMMR could activate Akt signaling, thus succeeding in distant colonization and metastatic outgrowth. Importantly, inducible depletion of HMMR significantly abrogates peritoneal implantation of gastric cancer in vitro and in vivo. CONCLUSION: Our study highlights that HMMR promotes peritoneal implantation of gastric cancer. A better understanding of HMMR's functions and mechanism might provide a novel therapeutic target and prognostic marker for metastatic gastric cancer.

10.
Cancer Res ; 82(22): 4191-4205, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36112698

ABSTRACT

Tumor metastasis is one of the major causes of high mortality in patients with hepatocellular carcinoma (HCC). Sustained activation of STAT3 signaling plays a critical role in HCC metastasis. RNA binding protein (RBP)-mediated posttranscriptional regulation is involved in the precise control of signal transduction, including STAT3 signaling. In this study, we investigated whether RBPs are important regulators of HCC metastasis. The RBP MEX3C was found to be significantly upregulated in highly metastatic HCC and correlated with poor prognosis in HCC. Mechanistically, MEX3C increased JAK2/STAT3 pathway activity by downregulating SOCS3, a major negative regulator of JAK2/STAT3 signaling. MEX3C interacted with the 3'UTR of SOCS3 and recruited CNOT7 to ubiquitinate and accelerate decay of SOCS3 mRNA. Treatment with MEX3C-specific antisense oligonucleotide significantly inhibited JAK2/STAT3 pathway activation, suppressing HCC migration in vitro and metastasis in vivo. These findings highlight a novel mRNA decay-mediated mechanism for the disruption of SOCS3-driven negative regulation of JAK2/STAT3 signaling, suggesting MEX3C may be a potential prognostic biomarker and promising therapeutic target in HCC. SIGNIFICANCE: This study reveals that RNA-binding protein MEX3C induces SOCS3 mRNA decay to promote JAK2/STAT3 activation and tumor metastasis in hepatocellular carcinoma, identifying MEX3C targeting as a potential approach for treating metastatic disease.


Subject(s)
Carcinoma, Hepatocellular , Janus Kinase 2 , Liver Neoplasms , RNA Stability , RNA-Binding Proteins , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Humans , Carcinoma, Hepatocellular/pathology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Liver Neoplasms/pathology , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling Proteins/genetics
11.
Clin Transl Med ; 12(1): e725, 2022 01.
Article in English | MEDLINE | ID: mdl-35090098

ABSTRACT

Triple-negative breast cancer (TNBC) is fast-growing and highly metastatic with the poorest prognosis among the breast cancer subtypes. Inactivation of glycogen synthase kinase 3 beta (GSK3ß) plays a vital role in the aggressiveness of TNBC; however, the underlying mechanism for sustained GSK3ß inhibition remains largely unknown. Here, we find that protein phosphatase 1 regulatory inhibitor subunit 14C (PPP1R14C) is upregulated in TNBC and relevant to poor prognosis in patients. Overexpression of PPP1R14C facilitates cell proliferation and the aggressive phenotype of TNBC cells, whereas the depletion of PPP1R14C elicits opposite effects. Moreover, PPP1R14C is phosphorylated and activated by protein kinase C iota (PRKCI) at Thr73. p-PPP1R14C then represses Ser/Thr protein phosphatase type 1 (PP1) to retain GSK3ß phosphorylation at high levels. Furthermore, p-PPP1R14C recruits E3 ligase, TRIM25, toward the ubiquitylation and degradation of non-phosphorylated GSK3ß. Importantly, the blockade of PPP1R14C phosphorylation inhibits xenograft tumorigenesis and lung metastasis of TNBC cells. These findings provide a novel mechanism for sustained GSK3ß inactivation in TNBC and suggest that PPP1R14C might be a potential therapeutic target.


Subject(s)
Glycogen Synthase Kinase 3 beta/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Triple Negative Breast Neoplasms/genetics , Disease Progression , Female , Glycogen Synthase Kinase 3 beta/genetics , Humans , Intracellular Signaling Peptides and Proteins/adverse effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
12.
Chem Catal ; 1(1): 106-116, 2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34337591

ABSTRACT

The use of visible-light photosensitizers to power [2+2] photocycloadditions that produce complex tetrasubstituted cyclobutanes is a true success of photochemistry, but the scope of this reaction has been limited to activated α, ß-unsaturated carbonyls. This paper describes selective intermolecular homo- and hetero-[2+2] photocycloadditions of terminal and internal aryl conjugated dienes - substrates historically unsuited for this reaction because of their multiple possible reaction pathways and product configurations - through triplet-triplet energy transfer from CdSe nanocrystal photocatalysts, to generate valuable and elusive syn-trans aryl vinylcyclobutanes. The negligible singlet-triplet splitting of nanocrystals' excited states allows them to drive the [2+2] pathway over the competing [4+2] photoredox pathway, a chemoselectivity not achievable with any known molecular photosensitizer. Reversible tethering of the cyclobutane product to the nanocrystal surface results in near quantitative yield of the syn-trans product. Flat colloidal CdSe nanoplatelets produce cyclobutanes coupled at the terminal alkenes of component dienes with up to 89% regioselectivity.

13.
Cancer Lett ; 503: 43-53, 2021 04 10.
Article in English | MEDLINE | ID: mdl-33352248

ABSTRACT

Chemotherapy regimens containing cisplatin remain the first-line treatments for patients with oral squamous cell cancer (OSCC); however, the treatment effect is often transient because of chemoresistance and recurrence. Understanding the mechanisms of chemoresistance in OSCC might provide novel targetable vulnerabilities. In the present study, we revealed that Forkhead box D1 (FOXD1) is upregulated in OSCC and predicted poor prognosis. Moreover, ectopic expression of FOXD1 promoted, while silencing of FOXD1 inhibited, the epithelial-mesenchymal transition (EMT) and chemoresistance of OSCC, both in vitro and in vivo. Mechanistically, FOXD1 binds to the promoter of long non-coding RNA Cytoskeleton Regulator RNA (CYTOR) and activates its transcription. CYTOR then acts as a competing endogenous RNA to inhibit miR-1252-5p and miR-3148, thus upregulating lipoma preferred partner (LPP) expression. Importantly, the CYTOR/LPP axis was proven to be essential for FOXD1-induced EMT and chemoresistance in OSCC. These findings reveal a novel mechanism for the chemotherapy resistance of OSCC, suggesting that FOXD1 might be a potential prognostic marker and anti-resistance therapeutic target.


Subject(s)
Carcinoma, Squamous Cell/pathology , Drug Resistance, Neoplasm , Forkhead Transcription Factors/metabolism , Mouth Neoplasms/pathology , RNA, Long Noncoding/genetics , Up-Regulation , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , LIM Domain Proteins/genetics , Mice , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Neoplasm Transplantation , Prognosis , Promoter Regions, Genetic
14.
J Phys Chem Lett ; 12(50): 12135-12141, 2021 Dec 23.
Article in English | MEDLINE | ID: mdl-34913699

ABSTRACT

This paper describes a photocatalytic hydrogen evolution system that is dynamically and reversibly responsive to the pH of the surrounding solution through the actuation of a microhydrogel (microgel) matrix that hosts the photocatalysts (CdSe/CdS nanorods). The reversible actuation occurs within 0.58 (swelling) and 1.7 s (contraction). ΔpH = 0.01 relative to the pKa of the tertiary amine on the microgel polymer (7.27) results in a reversible change in the average diameter of the microgel hosts by a factor of 2.4 and a change in the photocatalytic turnover frequency (TOF) by a factor of 5. Kinetic isotope effect and photoluminescence quenching experiments reveal that the scavenging of the photoexcited hole by sulfite ions is the rate-limiting step and leads to the observed response of the TOF to pH through the actuation of the microgel. Molecular dynamics simulations quantify a greater local concentration of sulfite hole scavengers for pH < pKa.


Subject(s)
Hydrogels/chemistry , Light , Cadmium Compounds/chemistry , Catalysis , Deuterium Oxide/chemistry , Hydrogen-Ion Concentration , Kinetics , Molecular Dynamics Simulation , Nanotubes/chemistry , Selenium Compounds/chemistry , Sulfides/chemistry
15.
J Hematol Oncol ; 14(1): 6, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33407765

ABSTRACT

BACKGROUND: HOMER family scaffolding proteins (HOMER1-3) play critical roles in the development and progression of human disease by regulating the assembly of signal transduction complexes in response to extrinsic stimuli. However, the role of HOMER protein in breast cancer remains unclear. METHODS: HOMER3 expression was examined by immunohistochemistry in breast cancer patient specimens, and its significance in prognosis was assessed by Kaplan-Meier survival analysis. The effects of HOMER3 in growth factor-induced ß-Catenin activation were analyzed by assays such as TOP/FOP flash reporter, tyrosine phosphorylation assay and reciprocal immunoprecipitation (IP) assay. Role of HOMER3 in breast cancer metastasis was determined by cell function assays and mice tumor models. RESULTS: Herein, we find that, among the three HOMER proteins, HOMER3 is selectively overexpressed in the most aggressive triple negative breast cancer (TNBC) subtype, and significantly correlates with earlier tumor metastasis and shorter patient survival. Mechanismly, HOMER3 interacts with both c-Src and ß-Catenin, thus providing a scaffolding platform to facilitate c-Src-induced ß-Catenin tyrosine phosphorylation under growth factor stimulation. HOMER3 promotes ß-Catenin nuclear translocation and activation, and this axis is clinically relevant. HOMER3 promotes and is essential for EGF-induced aggressiveness and metastasis of TNBC cells both in vitro and in vivo. CONCLUSION: These findings identify a novel role of HOMER3 in the transduction of growth factor-mediated ß-Catenin activation and suggest that HOMER3 might be a targetable vulnerability of TNBC.


Subject(s)
Homer Scaffolding Proteins/metabolism , Lung Neoplasms/secondary , Triple Negative Breast Neoplasms/pathology , beta Catenin/metabolism , Animals , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Triple Negative Breast Neoplasms/metabolism , Tyrosine/metabolism
16.
Nat Commun ; 12(1): 7006, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853315

ABSTRACT

Nicotine addiction and the occurrence of lymph node spread are two major significant factors associated with esophageal cancer's poor prognosis; however, nicotine's role in inducing lymphatic metastasis of esophageal cancer remains unclear. Here we show that OTU domain-containing protein 3 (OTUD3) is downregulated by nicotine and correlates with poor prognosis in heavy-smoking esophageal cancer patients. OTUD3 directly interacts with ZFP36 ring finger protein (ZFP36) and stabilizes it by inhibiting FBXW7-mediated K48-linked polyubiquitination. ZFP36 binds with the VEGF-C 3-'UTR and recruits the RNA degrading complex to induce its rapid mRNA decay. Downregulation of OTUD3 and ZFP36 is essential for nicotine-induced VEGF-C production and lymphatic metastasis in esophageal cancer. This study establishes that the OTUD3/ZFP36/VEGF-C axis plays a vital role in nicotine addiction-induced lymphatic metastasis, suggesting that OTUD3 may serve as a prognostic marker, and induction of the VEGF-C mRNA decay might be a potential therapeutic strategy against human esophageal cancer.


Subject(s)
Down-Regulation/drug effects , Esophageal Neoplasms/metabolism , Lymphatic Metastasis , Nicotine/pharmacology , RNA Stability/physiology , Ubiquitin-Specific Proteases/metabolism , Vascular Endothelial Growth Factor C/metabolism , Angiogenesis Inducing Agents/pharmacology , Animals , Cell Line, Tumor , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/metabolism , Tristetraprolin/metabolism , Ubiquitin-Specific Proteases/genetics , Vascular Endothelial Growth Factor C/genetics
17.
Cancer Res ; 81(13): 3525-3538, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33975879

ABSTRACT

Balancing mRNA nuclear export kinetics with its nuclear decay is critical for mRNA homeostasis control. How this equilibrium is aberrantly disrupted in esophageal cancer to acquire cancer stem cell properties remains unclear. Here we find that the RNA-binding protein interleukin enhancer binding factor 2 (ILF2) is robustly upregulated by nicotine, a major chemical component of tobacco smoke, via activation of JAK2/STAT3 signaling and significantly correlates with poor prognosis in heavy-smoking patients with esophageal cancer. ILF2 bound the THO complex protein THOC4 as a regulatory cofactor to induce selective interactions with pluripotency transcription factor mRNAs to promote their assembly into export-competent messenger ribonucleoprotein complexes. ILF2 facilitated nuclear mRNA export and inhibited hMTR4-mediated exosomal degradation to promote stabilization and expression of SOX2, NANOG, and SALL4, resulting in enhanced stemness and tumor-initiating capacity of esophageal cancer cells. Importantly, inducible depletion of ILF2 significantly increased the therapeutic efficiency of cisplatin and abrogated nicotine-induced chemoresistance in vitro and in vivo. These findings reveal a novel role of ILF2 in nuclear mRNA export and maintenance of cancer stem cells and open new avenues to overcome smoking-mediated chemoresistance in esophageal cancer. SIGNIFICANCE: This study defines a previously uncharacterized role of nicotine-regulated ILF2 in facilitating nuclear mRNA export to promote cancer stemness, suggesting a potential therapeutic strategy against nicotine-induced chemoresistance in esophageal cancer.


Subject(s)
Drug Resistance, Neoplasm , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Neoplastic Stem Cells/pathology , Nicotine/pharmacology , Nuclear Factor 45 Protein/metabolism , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nicotinic Agonists/pharmacology , Nuclear Factor 45 Protein/genetics , Prognosis , RNA, Messenger/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Survival Rate , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Adv Mater ; 32(8): e1906600, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31944429

ABSTRACT

A novel method for synthesizing and photopatterning colloidal crystals via light-responsive DNA is developed. These crystals are composed of 10-30 nm gold nanoparticles interconnected with azobenzene-modified DNA strands. The photoisomerization of the azobenzene molecules leads to reversible assembly and disassembly of the base-centered cubic (bcc) and face-centered cubic (fcc) crystalline nanoparticle lattices. In addition, UV light is used as a trigger to selectively remove nanoparticles on centimeter-scale thin films of colloidal crystals, allowing them to be photopatterned into preconceived shapes. The design of the azobenzene-modified linking DNA is critical and involves complementary strands, with azobenzene moieties deliberately staggered between the bases that define the complementary code. This results in a tunable wavelength-dependent melting temperature (Tm ) window (4.5-15 °C) and one suitable for affecting the desired transformations. In addition to the isomeric state of the azobenzene groups, the size of the particles can be used to modulate the Tm window over which these structures are light-responsive.


Subject(s)
DNA/chemistry , Metal Nanoparticles/chemistry , Ultraviolet Rays , Azo Compounds/chemistry , Gold/chemistry , Scattering, Small Angle , Transition Temperature , X-Ray Diffraction
19.
Ann Surg Treat Res ; 97(1): 1-6, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31297346

ABSTRACT

PURPOSE: The combination of indocyanine green and methylene blue (ICG + MB) was reported to be an efficient tracer method in sentinel lymph node biopsy (SLNB). However, whether this method is superior to MB only or carbon nanoparticles (CN) is controversial. This study was to evaluate the efficacy of the three methods in SLNB for breast cancer, and to analyze its influencing factors. METHODS: One hundred eighty patients with early breast cancer were recruited and randomly divided into 3 groups. Each group comprising of 60 patients with SLNB using ICG + MB, MB, and CN, respectively. Then the 3 groups were compared in detection rate, mean number of SLNs, and the detection rates and number of metastatic sentinel lymph nodes (SLNs). RESULTS: The detection rate of SLNs was 100% (60 of 60) in ICG + MB group, 96.7% (58 of 60), and 98.3% (59 of 60) in MB and CN group, respectively, with no significant difference (P = 0.362). Totally, 204 SLNs (mean ± standard deviation [SD] [range], 3.4 ± 1.4 [2-8]) were detected in ICG + MB group, 102 (1.7 ± 0.7 [0-3]) and 145 (2.4 ± 0.7 [0-6]) in MB and CN group, indicating significant difference (P < 0.001). The detection rate of metastatic SLN was 23.3% (14 of 60) in ICG + MB group, which was higher than 18.3% (11 of 60) and 20% (11 of 60) in MB and CN group, respectively, but showed no statistical significance (P = 0.788). CONCLUSION: ICG + MB method was superior to MB only and CN only methods in the mean number of SLNs, thus predicting axillary lymph node metastasis more accurately. Therefore, in areas where the standard method is not available, ICG + MB may be more suitable as an alternative tracer for SLNB.

20.
Curr Gene Ther ; 19(1): 54-65, 2019.
Article in English | MEDLINE | ID: mdl-30848201

ABSTRACT

BACKGROUND: Both Drosophila melanogaster deoxyribonucleoside kinase (Dm-dNK) suicide gene therapy and exogenous CD40 ligand (CD40L)-CD40 interaction in cancer via conditionally replicating adenovirus can selectively kill tumors without damaging normal tissues. OBJECTIVE: To further improve the cancer killing effect, we investigated the therapeutic effect of combined cancer gene therapy based on a selective oncolytic adenovirus vector containing Dm-dNK suicide gene and exogenous CD40L on breast carcinoma cells in vitro and in vivo. METHODS: A series of conditionally replicating adenoviruses using adenovirus vector P74 were generated: P74-dNK, P74-CD40L (expressing Dm-dNK or CD40L respectively), and P74-dNK-CD40L (expressing combined Dm-dNK and CD40L). Breast cancer cell lines (MDA-MB-231, MCF-7) and non-tumor cell line (MRC5) were treated with adenovirus and cytotoxicity determined by MTT assay, and apoptosis assessed by flow cytometry after 72h. We also assessed in vivo cell killing efficiency using a mouse xenograft model with MDA-MB-231 cells. RESULTS AND DISCUSSION: Co-expression of Dm-dNK and CD40L reduced cell proliferation of MDAMB- 231 or MCF7 cancer cells, and induced more apoptosis in TERT and CD40 positive cancer cells, but not normal MRC5 cells. Significant reduction in tumor volume was also seen in combined treatment arms as compared to any single treatment. CONCLUSION: Our data suggest enhanced, selective tumor cell killing using combined gene therapy with conditionally replicating adenovirus containing Dm-dNK suicide gene and exogenous CD40 ligation (CD40L-CD40).


Subject(s)
CD40 Ligand/genetics , Drosophila melanogaster/enzymology , Genetic Therapy , Genetic Vectors/administration & dosage , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Proliferation , Female , Genes, Transgenic, Suicide , Genetic Vectors/genetics , Humans , Mice , Mice, Nude , Phosphorylation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL