Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
2.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(3): 455-461, 2023 May.
Article in Chinese | MEDLINE | ID: mdl-37248568

ABSTRACT

CD47 is an immunoglobulin that is overexpressed on the surface of a variety of cancer cells. CD47 forms a signaling complex with signal regulatory protein alpha (SIRPα), prompting the escape of cancer cells from macrophage-mediated phagocytosis. In recent years, CD47 has been shown to be highly expressed in many types of solid tumors and is associated with poor prognosis in patients. More and more studies have shown that inhibition of the CD47-SIRPα signaling pathway can promote adaptive immune responses and enhance the phagocytosis of tumor cells by macrophages. Humanized anti-CD47 IgG4 monoclonal antibody has been studied in clinical trials for the treatment of a variety of advanced solid tumors and lymphomas, demonstrating a sound safety profile and achieving partial remission in some patients. In this review we discuss the structure and function of CD47 and the mechanism of CD47 regulation in tumors, summarize the research progress in therapeutic antibody drugs targeting CD47 and a bottleneck in research that targeted drugs are more prone to result in serious adverse effects, and evaluated the potential of the applying CD47-SIRPα signaling pathway in anti-cancer therapy.


Subject(s)
Antineoplastic Agents , CD47 Antigen , Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , CD47 Antigen/metabolism , Immunotherapy , Macrophages/metabolism , Neoplasms/drug therapy , Phagocytosis , Tumor Escape
3.
Cancer Biol Med ; 19(8)2021 12 01.
Article in English | MEDLINE | ID: mdl-34846107

ABSTRACT

OBJECTIVE: We aimed to investigate the radiosensitizing efficacy of the poly-ADP-ribose polymerase (PARP) inhibitor, olaparib, and the Bloom syndrome protein (BLM) helicase inhibitor, ML216, in non-small cell lung cancer (NSCLC) cells. METHODS: Radiosensitization of NSCLC cells was assessed by colony formation and tumor growth assays. Mechanistically, the effects of ML216, olaparib, and radiation on cell and tumor proliferation, DNA damage, cell cycle, apoptosis, homologous recombination (HR) repair, and non-homologous end joining (NHEJ) repair activity were determined. RESULTS: Both olaparib and ML216 enhanced the radiosensitivities of olaparib-sensitive H460 and H1299 cells, which was seen as decreased surviving fractions and Rad51 foci, increased total DNA damage, and γH2AX and 53BP1 foci (P < 0.05). The expressions of HR repair proteins were remarkably decreased in olaparib-treated H460 and H1299 cells after irradiation (P < 0.05), while olaparib combined with ML216 exerted a synergistic radiosensitization effect on olaparib-resistant A549 cells. In addition to increases of double strand break (DSB) damage and decreases of Rad51 foci, olaparib combined with ML216 also increased pDNA-PKcs (S2056) foci, abrogated G2 cell cycle arrest, and induced apoptosis in A549 lung cancer after irradiation in vitro and in vivo (P < 0.05). Moreover, Western blot showed that olaparib combined with ML216 and irradiation inhibited HR repair, promoted NHEJ repair, and inactivated cell cycle checkpoint signals both in vitro and in vivo (P < 0.05). CONCLUSIONS: Taken together, these results showed the efficacy of PARP and BLM helicase inhibitors for radiosensitizing NSCLC cells, and supported the model that BLM inhibition sensitizes cells to PARP inhibitor-mediated radiosensitization, as well as providing the basis for the potential clinical development of this combination for tumors intrinsically resistant to PARP inhibitors and radiotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Radiation-Sensitizing Agents , Adenosine Diphosphate Ribose/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Phthalazines , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , RecQ Helicases/genetics , Recombinational DNA Repair
4.
Nucleic Acids Res ; 48(16): 9109-9123, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32729622

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (NRF2) is a well-characterized transcription factor that protects cells against oxidative and electrophilic stresses. Emerging evidence has suggested that NRF2 protects cells against DNA damage by mechanisms other than antioxidation, yet the mechanism remains poorly understood. Here, we demonstrate that knockout of NRF2 in cells results in hypersensitivity to ionizing radiation (IR) in the presence or absence of reactive oxygen species (ROS). Under ROS scavenging conditions, induction of DNA double-strand breaks (DSBs) increases the NRF2 protein level and recruits NRF2 to DNA damage sites where it interacts with ATR, resulting in activation of the ATR-CHK1-CDC2 signaling pathway. In turn, this leads to G2 cell cycle arrest and the promotion of homologous recombination repair of DSBs, thereby preserving genome stability. The inhibition of NRF2 by brusatol increased the radiosensitivity of tumor cells in xenografts by perturbing ATR and CHK1 activation. Collectively, our results reveal a novel function of NRF2 as an ATR activator in the regulation of the cellular response to DSBs. This shift in perspective should help furnish a more complete understanding of the function of NRF2 and the DNA damage response.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , NF-E2-Related Factor 2/genetics , Recombinational DNA Repair/genetics , A549 Cells , Animals , CDC2 Protein Kinase/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Checkpoint Kinase 1/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockout Techniques , Heterografts , Humans , Mice , Quassins/pharmacology , Radiation Tolerance/drug effects , Radiation, Ionizing , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/radiation effects , Signal Transduction/drug effects
5.
Int J Clin Exp Pathol ; 13(2): 192-202, 2020.
Article in English | MEDLINE | ID: mdl-32211099

ABSTRACT

BACKGROUND: Hepatorenal and hepatopulmonary syndrome are common clinical diseases; however, their mechanisms have not been fully elucidated. Our aim was to determine whether liver injury by bile duct ligation (BDL) causes modifications in kidney and lung tissue in mice, and to explore the possible mechanism of these changes. METHODS: BDL in mice was used as a research model. Pathologic changes of liver, kidney, and lung tissue were observed by hematoxylin-eosin (H&E) staining. The expression of IGFBPrP1, NF-κB, TNF-α, and IL-6 were investigated in liver, kidney, and lung tissue by immunohistochemical staining and western blot. The correlation between IGFBPrP1 and NF-κB, TNF-α, and IL-6 protein expression in liver, kidney, and lung tissues of each group was analyzed by the Pearson method. RESULTS: H&E staining showed, after BDL administration in mice, different degrees of inflammatory change in liver, kidney, and lung tissues of mice in each group. The results of immunohistochemical staining and western blot analysis showed increased expressions of IGFBPrP1, NF-κB, TNF-α, and IL-6 after BDL. Pearson correlation analysis showed that IGFBPrP1 positively correlated with the expressions of NF-κB, TNF-α, and IL-6. CONCLUSION: Liver injury caused by bile duct ligation can lead to kidney and lung tissue injury in mice. The mechanism of injury may be related to the high expression of liver injury factor IGFBPrP1, transcription factor NF-κB, proinflammatory cytokine TNF-α, and IL-6 in kidney and lung tissue. Moreover, an increased expression level of IGFBPrP1 may be accompanied by the activation of the NF-κB inflammatory pathway.

6.
Dig Dis Sci ; 65(2): 509-523, 2020 02.
Article in English | MEDLINE | ID: mdl-31468266

ABSTRACT

BACKGROUND: Autophagy is a self-degrading process. Previously, we showed that insulin-like growth factor binding protein-related protein 1 (IGFBPrP1) is a novel transforming growth factor ß1 (TGFß1)-interacting factor in liver fibrosis; the role of TGFß1-mediated autophagy in hepatic stellate cells (HSCs) activation has been investigated. However, whether autophagy is regulated by IGFBPrP1 remains unknown. AIMS: We investigated the interactions among IGFBPrP1, autophagy, and activation of primary rat HSCs. METHODS: Primary HSCs were separated from Sprague Dawley rats by two-step enzymatic digestion, and then, we overexpressed or inhibited IGFBPrP1 expression in HSCs under serum-starved condition. Autophagy inducer rapamycin or inhibitor 3-methyladenine (3MA) was used to assess the relationship between autophagy and HSCs activation. RESULTS: We observed the expression of activation marker α-SMA and autophagy markers such as LC3B and Beclin1, which were significantly increased in HSCs treated with adenovirus vector harboring the IGFBPrP1 gene (AdIGFBPrP1) compared to cells cultured under serum-starved. In comparison, HSCs treated with shIGFBPrP1 showed opposite results. Furthermore, HSCs activation and autophagy increased when cells were treated with rapamycin, whereas opposite results were obtained when cells were treated with 3MA. AdIGFBPrP1 treatment downregulated the phosphorylation of Akt and mTOR. CONCLUSION: Autophagy was induced in IGFBPrP1-treated primary HSCs, and IGFBPrP1-induced autophagy promoted the activation of HSCs and extracellular matrix expression, the underlying mechanism of which may involve the phosphatidylinositide 3-kinase/Akt/mTOR signaling pathway.


Subject(s)
Autophagy/genetics , Hepatic Stellate Cells/metabolism , Insulin-Like Growth Factor Binding Proteins/genetics , Actins/genetics , Actins/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Beclin-1/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/ultrastructure , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transfection , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
7.
Life Sci ; 237: 116902, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31610195

ABSTRACT

AIMS: Insulin-like growth factor binding protein-related protein 1 (IGFBPrP1) promotes hepatic stellate cell (HSC) autophagy and activation. However, the underlying mechanism remains unknown. Noncoding RNAs (ncRNAs) including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), have received increasing attention. We aimed to investigate the roles of the lncRNA nuclear enriched abundant transcript 1 (NEAT1), miR-29b, and autophagy related protein 9a (Atg9a), and their relationships with each other during IGFBPrP1-induced HSC autophagy and activation. MAIN METHODS: Levels of NEAT1, miR-29b, Atg9a, and autophagy were detected in adenovirus-mediated IGFBPrP1 (AdIGFBPrP1)-treated mouse liver tissue and immortalized mouse hepatic stellate cell line JS1 transfected with either AdIGFBPrP1 or siIGFBPrP1. In AdIGFBPrP1-treated JS1 cells, autophagy and activation were detected after altering NEAT1, miR-29b, or Atg9a levels. In AdIGFBPrP1-treated JS1 cells, relationships among NEAT1, miR-29b, and Atg9a were explored using dual-luciferase reporter assays, Western blot, qRT-PCR, and immunofluorescence. KEY FINDINGS: IGFBPrP1 increased levels of NEAT1, Atg9a, and autophagy while decreasing the level of miR-29b in mouse liver tissues and mouse HSCs. Moreover, NEAT1 increased HSC autophagy and activation while miR-29b decreased both processes. Atg9a also participated in IGFBPrP1-induced HSC autophagy and activation. Importantly, NEAT1, miR-29b, and Atg9a formed a NEAT1/miR-29b/Atg9a regulatory axis for IGFBPrP1-induced HSC autophagy and activation. SIGNIFICANCE: Our study unveiled the new NEAT1/miR-29b/Atg9a regulatory axis involved in IGFBPrP1-induced mouse HSC autophagy and activation. The study thus provides new insights in the pathogenesis and potential therapeutic strategies of liver fibrosis.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy , Hepatic Stellate Cells/pathology , Insulin-Like Growth Factor Binding Proteins/metabolism , Liver Cirrhosis/pathology , Membrane Proteins/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Vesicular Transport Proteins/metabolism , Adenoviridae/genetics , Animals , Autophagy-Related Proteins/genetics , Cells, Cultured , Hepatic Stellate Cells/metabolism , Insulin-Like Growth Factor Binding Proteins/genetics , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Vesicular Transport Proteins/genetics
8.
Biomed Pharmacother ; 116: 109034, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31152924

ABSTRACT

BACKGROUND: Our previous study found that insulin-like growth factor binding protein-associated protein (IGFBPrP1) drives hepatic stellate cells (HSCs) activation, and IGFBPrP1 and transforming growth factor ß1 (TGFß1) likely interact with each other to promote HSCs activation. TGFß1 reportedly promotes autophagy and contributes to HSCs activation; however, the mechanism between IGFBPrP1 and autophagy in liver fibrogenesis is yet unknown. Moreover, long noncoding RNA (lncRNA) H19 participates in autophagy regulation and plays a crucial function in liver fibrosis. AIMS: To define the relationship between IGFBPrP1 and autophagy and the role of H19 in IGFBPrP1-induced hepatic fibrosis. METHODS: IGFBPrP1 and autophagy were detected in bile duct ligation (BDL)-induced hepatic fibrosis. Adenovirus-mediated IGFBPrP1 was transfected into mouse liver and JS-1 cells with or without LY294002 or rapamycin to examine the effects of IGFBPrP1 on HSCs activation and autophagy as well as the PI3K/AKT/mTOR pathway. lncRNA H19 in liver fibrosis tissues and JS-1 cells induced by IGFBPrP1 were detected, then autophagy and HSCs activation level were detected in JS-1 cells by IGFBPrP1 with H19 overexpression or knowdown. RESULTS: IGFBPrP1 expression and autophagy level were concomitantly increased in liver tissue with BDL-induced hepatic fibrosis. Furthermore, we found that IGFBPrP1 stimulated autophagy and HSCs activation in vivo and in vitro, and PI3K/AKT/mTOR signaling pathway was involved in the regulation of autophagy by IGFBPrP1. In addition, H19 promoted autophagy by interacting with the PI3K/AKT/mTOR pathway in IGFBPrP1-induced HSCs activation. CONCLUSIONS: IGFBPrP1 promoted autophagy and contributed to HSCs activation via mutual regulation between H19 and the PI3K/AKT/mTOR pathway.


Subject(s)
Autophagy , Hepatic Stellate Cells/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Bile Ducts/pathology , Cell Line , Fatty Liver/pathology , Hepatic Stellate Cells/pathology , Hepatic Stellate Cells/ultrastructure , Ligation , Liver/pathology , Male , Mice, Inbred C57BL
9.
Nanotechnology ; 30(33): 335703, 2019 Aug 16.
Article in English | MEDLINE | ID: mdl-30995621

ABSTRACT

The mechanical properties of two-dimensional (2D) materials are critical for their applications in functional devices as well as for strain engineering. Here, we report the Young's modulus and breaking strength of multilayered InSe, an emerging 2D semiconductor of the layered group III chalcogenide. Few-layer InSe flaks were exfoliated from bulk InSe crystal onto Si/SiO2 substrate with micro-fabricated holes, and indentation tests were carried out using an atomic force microscopy probe. In combination with both continuum analysis and finite element simulation, we measured the Young's modulus of multilayer 2D InSe (>5 L) to be 101.37 ± 17.93 GPa, much higher than its bulk counterpart, while its breaking strength is determined to be 8.68 GPa, approaching the theoretical limit of 10.1 GPa. Density functional theory calculations were also carried out to explain the insensitivity of Young's modulus to the layer count. It is found that 2D InSe is softer than most 2D materials, and exhibits breaking strength higher than that of carbon fiber, yet remaining more compliant, making it ideal for flexible electronics applications. The reliability of our method is also validated by measurement of graphene.

10.
Cell Death Dis ; 9(10): 1026, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297887

ABSTRACT

The acquisition of radioresistance by breast cancer cells during radiotherapy may lead to cancer recurrence and poor survival. Signal transducer and activator of transcription 3 (Stat3) is activated in breast cancer cells and, therefore, may be an effective target for overcoming therapeutic resistance. Mesenchymal stem cells (MSCs) have been investigated for use in cancer treatment. Here, we investigated the potential of MSC conditioned medium (MSC-CM) in sensitizing breast cancer to radiotherapy. It was found that MSC-CM could inhibit the level of activated Stat3, suppress cancer growth, and exhibit synergetic effects with radiation treatment in vitro and in vivo. Furthermore, MSC-CM reduced the ALDH-positive cancer stem cells (CSCs) population, modulated several potential stem cell markers, and decreased tumor migration, as well as metastasis. These results demonstrate that MSC-CM suppresses breast cancer cells growth and sensitizes cancer cells to radiotherapy through inhibition of the Stat3 signaling pathway, thus, providing a novel strategy for breast cancer therapy by overcoming radioresistance.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Down-Regulation/physiology , Mesenchymal Stem Cells/physiology , Radiation Tolerance/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Cell Transformation, Neoplastic/pathology , Culture Media, Conditioned/metabolism , Female , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/pathology
11.
Int J Lab Hematol ; 40(6): 630-636, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29989348

ABSTRACT

INTRODUCTION: Fanconi anemia (FA), as one of the congenital bone marrow failure syndromes, is characterized by severe bone marrow hypocellularity and pancytopenia which is similar with acquired aplastic anemia (AAA). However, patients with FA or AAA need an accurate diagnose, as the two syndromes differ significantly in both treatment and prognosis. FA results from gene mutations of the FA pathway genes specifically required for DNA repair, and the mutation of these genes contributes to the genome instability of FA cells. Based on this feature, chromosome aberration (CA) has been used as a "golden standard" to the auxiliary diagnosis of FA from AAA. However, CA diagnose calls for more technical requirements and a long time for the subsequent statistical analysis. METHODS: In our study, another two genome instability examination tools, cytokinesis-block micronucleus (CBMN) and single-cell gel electrophoresis (SCGE), were used to distinguish FA patients from AAA patients, compared with CA. RESULTS: The results suggested that significant differences were observed in the FA patients compared with the AAA patients and the controls using all of the three genomic instability examination tools. However, CBMN is the most cost-effective method to distinguish FA patients from AAA patients among the three genome instability examination tools, when the time costs, instrument costs, and technical costs were compared. CONCLUSION: In areas with economic and technical limitations, CBMN is an alternative assay to help distinguish FA patients from AAA patients.


Subject(s)
Comet Assay/methods , Fanconi Anemia/diagnosis , Genomic Instability , Micronuclei, Chromosome-Defective , Adolescent , Child , Child, Preschool , China , Female , Humans , Male
12.
Nanotechnology ; 29(28): 285402, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-29671406

ABSTRACT

Semiconductor phase transitions and plasma noble metal quantum dots (QDs) for visible-light-driven photocatalysts have attracted significant research interest. In this study, novel microwave hydrothermal and photo-reduction methods are proposed to synthesise a visible-light-driven plasma photocatalytic 1T@2H-MoS2/Ag composite. Photoelectrochemical results show that the introduction of the 1T phase and Ag significantly enhances the light response range and charge separation. The 1T phase can act as a co-catalyst to provide a high electron concentration. Ag QDs can effectively improve the light absorption and catalytic effect. The synergistic effect between the 1T@2H-MoS2 microspheres and localised surface plasmon resonance of the Ag QDs can effectively enhance the photocatalytic activity of 1T@2H-MoS2/Ag. The developed 1T@2H-MoS2/Ag composite is superior, not only with respect to a visible-light photocatalytic degradation of conventional dyes, but also in the photocatalytic reduction of Cr(VI). Compared with 2H-MoS2, the catalytic efficiency of 1T@2H-MoS2/Ag for Cr(VI) and MB is increased by 81% and 41%, respectively. This study demonstrates that the introduction of 1T-MoS2 and Ag QDs can significantly enhance the catalytic properties of 2H-MoS2. The microwave and photo-reduction technologies can be employed as green, safe, simple, and rapid methods for the synthesis of noble metal plasma composites.

13.
Org Biomol Chem ; 16(11): 1921-1931, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29488528

ABSTRACT

A series of primary aminomethyl derivatives of kaempferol were synthesized by a combination strategy involving two steps of the Mannich reaction and SN2 nucleophilic substitution. The structures of the products show that the preferential aminomethylations are in the position C-6 or C-8 of the A-ring of kaempferol, especially the latter. Interestingly, the experimental data indicate that the intermolecular hydrogen bonding plays a key role in the formation of primary aminomethyl products of kaempferol. The formation of appropriate hydrogen bonds between strong nucleophilic amino acids and phenol is essential for the smooth reaction of the SN2 nucleophilic substitution. The SN2 mechanism hypothesis involving a hydrogen bond-assisted process was also supported by the density functional theory (DFT) analysis. An antiproliferative test of synthetic compounds shows the moderate to potent cytotoxic activity against three human cancer cell lines (HeLa, HCC1954, and SK-OV-3) by the CCK-8 assay. Compound 4e shows selective antiproliferative activity against HeLa cells with a low IC50 value (4.27 µm) and is worthy of further development. Another interesting result is that the maximum emission bands for most metal complexes are located at about 480 nm, but the ones for Tm and Yb complexes appear at about 533 nm.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Kaempferols/chemistry , Kaempferols/pharmacology , Amination , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic/methods , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Hydrogen Bonding , Kaempferols/chemical synthesis , Methylation , Models, Molecular , Neoplasms/drug therapy
14.
Biochem Biophys Res Commun ; 494(1-2): 158-164, 2017 12 09.
Article in English | MEDLINE | ID: mdl-29042194

ABSTRACT

RMI1 (RecQ-mediated genome instability protein 1) forms a conserved BTR complex with BLM, Topo IIIα, and RMI2, and its absence causes genome instability. It has been revealed that RMI1 localizes to nuclear foci with BLM and Topo IIIα in response to replication stress, and that RMI1 functions downstream of BLM in promoting replication elongation. However, the precise functions of RMI1 during replication stress are not completely understood. Here we report that RMI1 knockdown cells are hypersensitive to hydroxyurea (HU). Using comet assay, we show that RMI1 knockdown cells exhibit accumulation of broken DNAs after being released from HU treatment. Moreover, we demonstrate that RMI1 facilitates the recovery from activated checkpoint and resuming the cell cycle after replicative stress. Surprisingly, loss of RMI1 results in a failure of RAD51 loading onto DNA damage sites. These findings reveal the importance of RMI1 in response to replication stress, which could explain the molecular basis for its function in maintaining genome integrity.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , DNA Repair/genetics , DNA Replication/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Checkpoints , Cell Survival/drug effects , DNA Damage , DNA Repair/physiology , DNA Replication/physiology , DNA-Binding Proteins , Gene Knockdown Techniques , Genomic Instability/drug effects , HeLa Cells , Humans , Hydroxyurea/toxicity , Nuclear Proteins/metabolism , Rad51 Recombinase/metabolism , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...