Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Curr Med Sci ; 44(2): 426-434, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38561594

ABSTRACT

OBJECTIVE: Glucose-6-phosphate isomerase (GPI) deficiency is a rare hereditary nonspherocytic hemolytic anemia caused by GPI gene variants. This disorder exhibits wide heterogeneity in its clinical manifestations and molecular characteristics, often posing challenges for precise diagnoses using conventional methods. To this end, this study aimed to identify the novel variants responsible for GPI deficiency in a Chinese family. METHODS: The clinical manifestations of the patient were summarized and analyzed for GPI deficiency phenotype diagnosis. Novel compound heterozygous variants of the GPI gene, c.174C>A (p.Asn58Lys) and c.1538G>T (p.Trp513Leu), were identified using whole-exome and Sanger sequencing. The AlphaFold program and Chimera software were used to analyze the effects of compound heterozygous variants on GPI structure. RESULTS: By characterizing 53 GPI missense/nonsense variants from previous literature and two novel missense variants identified in this study, we found that most variants were located in exons 3, 4, 12, and 18, with a few localized in exons 8, 9, and 14. This study identified novel compound heterozygous variants associated with GPI deficiency. These pathogenic variants disrupt hydrogen bonds formed by highly conserved GPI amino acids. CONCLUSION: Early family-based sequencing analyses, especially for patients with congenital anemia, can help increase diagnostic accuracy for GPI deficiency, improve child healthcare, and enable genetic counseling.


Subject(s)
Anemia, Hemolytic, Congenital Nonspherocytic , Anemia, Hemolytic , Child , Humans , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/chemistry , Anemia, Hemolytic/genetics , Anemia, Hemolytic, Congenital Nonspherocytic/diagnosis , Anemia, Hemolytic, Congenital Nonspherocytic/genetics , Mutation, Missense , Exons
2.
Front Genet ; 14: 1309040, 2023.
Article in English | MEDLINE | ID: mdl-38111681

ABSTRACT

Background: Hereditary spherocytosis (HS) is a congenital haemolytic anaemia attributed to dysregulation or abnormal quantities of erythrocyte membrane proteins. Currently, the most common erythrocytic gene, spectrin ß (SPTB), variants are located in exons and give rise to mRNA defects. However, the genetic characteristics and pathogenic mechanisms of SPTB intronic variants are not completely understood. This study aimed to analyse a rare intronic inversion variant in the SPTB gene associated with HS, and explore the impact of the variant on SPTB mRNA splicing. Method: The clinical manifestations of the patient were summarised and analysed for spherocytosis phenotype diagnosis. The pathogenic variant was identified in the proband using targeted next-generation and Sanger sequencing. RNA sequencing was performed to analyse whether SPTB gene splicing and expression were affected. Results: Targeted next-generation sequencing identified a novel disease-associated intronic inversion variant of the SPTB gene in the proband. The inversion variant was located between intron 19 and 20, and contained the entire exon 20 and partial sequences of adjacent introns. Sanger sequencing confirmed that the intronic inversion variant only appeared in the genome of the proband, not in his parents. RNA sequencing revealed that the variant could result in the skipping of exon 20 and reduced expression of SPTB mRNA. Conclusion: This study identifies a rare intronic inversion variant in the SPTB gene associated with hereditary spherocytosis. The pathogenic variant can lead to exon 20 skipping and decreased SPTB gene expression. This finding has not been previously reported in any literature. This study can expand the intronic variant spectrum of the SPTB gene, deepen our understanding of HS pathogenesis, and contribute to the genetic diagnosis and clinical management of patients.

3.
Front Cell Infect Microbiol ; 12: 888398, 2022.
Article in English | MEDLINE | ID: mdl-35774403

ABSTRACT

This retrospective study aimed to determine the characteristics of infection and diagnostic efficacy of next-generation sequencing (NGS) in patients with fever after allogeneic hematopoietic stem cell transplantation (allo-HSCT). A total of 71 patients with fever after HSCT were enrolled in this study. Compared with conventional microbiological test (CMT), we found that the sensitivity of NGS versus CMT in peripheral blood samples was 91.2% vs. 41.2%, and that NGS required significantly less time to identify the pathogens in both monomicrobial infections (P=0.0185) and polymicrobial infections (P= 0.0027). The diagnostic performance of NGS was not affected by immunosuppressant use. Viruses are the most common pathogens associated with infections. These results indicated that the sensitivity, timeliness, and clinical significance of NGS are superior for the detection of infections. Although NGS has the advantage of identifying a wide range of potential pathogens, the positive rate is related closely to the sample type. Therefore, we recommend that, in the clinical application of NGS to detect pathogens in patients after allo-HSCT, an appropriate sample type and time should be selected and submitted to improve the positive rate and accuracy of NGS. NGS holds promise as a powerful technology for the diagnosis of fever after HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cell Transplantation/adverse effects , High-Throughput Nucleotide Sequencing/methods , Humans , Retrospective Studies , Transplantation, Homologous/adverse effects
4.
Front Pediatr ; 10: 831363, 2022.
Article in English | MEDLINE | ID: mdl-35712638

ABSTRACT

Cunninghamellamycosis is an unusual but often highly fatal mucormycosis caused by Cunninghamella bertholletiae, which belongs to the basal lineage order Mucorales. It is especially fatal when the central nervous system is involved. So far, there are few reported cases of surgical treatment for intracranial mucormycosis in children after allogeneic haematopoietic stem cell transplantation (HSCT). The surgical management of deep-seated basal ganglia fungal lesions remains controversial, and its clinical benefits are not yet well established. Herein, we present a rare case of disseminated mucormycosis caused by C. bertholletiae involving the lung and intracranial basal ganglia after homologous leucocytic antigen-matched sibling donor HSCT. The patient was successfully treated for intracranial cunninghamellamycosis with neuroendoscopic surgery and systemic wide-spectrum antifungal treatment and achieved pulmonary recovery without recurrent C. bertholletiae infection or neurologic sequelae. Over the follow-up period of 13 months, there were no adverse events associated with the intracranial surgical debridement, and the patient remained in good health.

5.
Front Cell Dev Biol ; 10: 829404, 2022.
Article in English | MEDLINE | ID: mdl-35309907

ABSTRACT

Chemotherapy-induced senescence promotes immunocyte aggregation in the tumor microenvironment by upregulating the surface expression of activating ligands in cancer cells. However, these senescent tumor cells cannot be completely cleared and can induce tumor recurrence. Previous studiesshowed that soluble natural killer (NK) group 2D (NKG2D) ligands impair the recognition of multiple immune cells. In this study, we established an in vitro senescence model using neuroblastoma cells subjected to low-dose Chemotherapeutic drug doxorubicin or the Aurora A inhibitor MLN8237. The results showed that different neuroblastoma cell lines showed increased secretion of the NKG2D ligand MHC class I polypeptide-related sequence A/B (MICA/B) following proteolysis after treatment, with MICA/B subsequently recruited to exosomes to downregulate NKG2D expression in NK cells. Interestingly, disintegrin and metalloproteinase domain-containing 10 (ADAM10) was upregulated in senescent tumor cells, and combined treatment with the ADAM10 inhibitor GI254023X and chemotherapeutic drugs inhibited MICA/B secretion and enhanced recognition and killing by NK cells. Additionally, we found that expression of the long noncoding RNA MALAT1 was significantly increased in senescent neuroblastoma cells, and that MALAT1 served as a sponge for microRNA (miR)-92a-3p to counteract miR-92a-3p-mediated repression of ADAM10 levels. Furthermore, administration of a MALAT1 inhibitor or an miR-92a-3p mimic reduced the MICA/B shedding and enhanced recognition and killing by NK cells. These results confirmed that low-dose chemotherapy induces senescence in neuroblastoma cells, and that senescent tumor cells promote the shedding of the NKG2D ligand MICA/B through the MALAT1/miR-92a/ADAM10 axis, thereby contributing to the formation of a suppressive immune microenvironment and promoting immune escape.

6.
Article in English | MEDLINE | ID: mdl-36843622

ABSTRACT

Sulfoconjugation is the major pathway for thyroid hormone (TH) metabolism, converting T4 to inactive metabolites, T4S, rT3S, and T3S in fetus, via sulfotransferases (SULT) and type 3 deiodinase in gestation. Consistent with high production rate of T4S and rT3S, there are high serum sulfated iodothyronine analogs, including T4S, T3S, rT3S, and 3,3'-T2S (T2S), in ovine and human fetal and preterm infants. Fetal TH metabolic pathways predict T2S as the major TH metabolite in the fetus. Since maternal T2S appears to be quantitatively derived from fetal T3 (the active TH), the amount of T2S in the maternal compartment correlates with fetal thyroid function in sheep. In humans, maternal serum contains high levels of radioimmunoassayable T2S; however, it displays as a peak adjacent to but unidentical to synthetic T2S on HLPC and we named it the W-Compound. Levels of W-Compound increase during pregnancy and peak as high as 20-fold to that of nonpregnant women. Maternal serum levels of W-Compound significantly correlate with fetal T4 and W-compound concentrations but not maternal serum T4 in euthyroid or hyperthyroid women, showing a distinct difference between fetal and maternal in TH metabolism. Fetal T2S is actively transferred to the mother via placenta and the quantity of T2S or its metabolite (W-Compound) in maternal compartment reflects fetal thyroid function. Thus, maternal serum W-Compound may be a biomarker for monitoring fetal thyroid function in utero, although more investigations are needed to determine if it can be used as an alternative strategy for screening/managing congenital hypothyroidism due to dysregulated thyroid hormone metabolism.

7.
Clin Case Rep ; 9(5): e04111, 2021 May.
Article in English | MEDLINE | ID: mdl-34084507

ABSTRACT

We report the first case of a 12-year-old boy with Wiskott-Aldrich syndrome who developed CD20-weakly expressed and CD30-highly expressed Epstein-Barr virus-related post-transplant lymphoproliferative disorder refractory to rituximab treatment. The patient was effectively and safely treated with personalized low-dose chemotherapy and subsequently remained in complete remission for 1 year.

9.
Nat Commun ; 9(1): 1739, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29712898

ABSTRACT

Chemoresistance is a major unmet clinical obstacle in ovarian cancer treatment. Epigenetics plays a pivotal role in regulating the malignant phenotype, and has the potential in developing therapeutically valuable targets that improve the dismal outcome of this disease. Here we show that a series of transcription factors, including C/EBPß, GCM1, and GATA1, could act as potential modulators of histone methylation in tumor cells. Of note, C/EBPß, an independent prognostic factor for patients with ovarian cancer, mediates an important mechanism through which epigenetic enzyme modifies groups of functionally related genes in a context-dependent manner. By recruiting the methyltransferase DOT1L, C/EBPß can maintain an open chromatin state by H3K79 methylation of multiple drug-resistance genes, thereby augmenting the chemoresistance of tumor cells. Therefore, we propose a new path against cancer epigenetics in which identifying and targeting the key regulators of epigenetics such as C/EBPß may provide more precise therapeutic options in ovarian cancer.


Subject(s)
Biomarkers, Tumor/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Chromatin/metabolism , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Histones/genetics , Ovarian Neoplasms/genetics , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Chromatin/chemistry , Cisplatin/pharmacology , DNA-Binding Proteins , Databases, Factual , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Primary Cell Culture , Prognosis , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Hematol Oncol ; 10(1): 29, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28114995

ABSTRACT

BACKGROUND: Epigenetics has been known to play a critical role in regulating the malignant phenotype. This study was designed to examine the expression of DOT1L (histone 3 lysine 79 methyltransferase) and H3K79 methylation in normal ovarian tissues and ovarian tumors and to explore the function of DOT1L and its underline mechanisms in ovarian cancer. METHODS: The expression of DOT1L and H3K79 methylation in 250 ovarian tumor samples and 24 normal ovarian samples was assessed by immunohistochemistry. The effects of DOT1L on cell proliferation in vitro were evaluated using CCK8, colony formation and flow cytometry. The DOT1L-targeted genes were determined using chromatin immune-precipitation coupled with high-throughput sequencing (ChIP-seq) and ChIP-PCR. Gene expression levels were measured by real-time PCR and immunoblotting. The effects of DOT1L on tumor growth in vivo were evaluated using an orthotopic ovarian tumor model. RESULTS: DOT1L expression and H3K79 methylation was significantly increased in malignant ovarian tumors. High DOT1L expression was associated with International Federation of Gynecology and Obstetrics (FIGO) stage, histologic grade, and lymphatic metastasis. DOT1L was an independent prognostic factor for the overall survival (OS) and progression-free survival (PFS) of ovarian cancer, and higher DOT1L expression was associated with poorer OS and PFS. Furthermore, DOT1L regulates the transcription of G1 phase genes CDK6 and CCND3 through H3K79 dimethylation; therefore, blocking DOT1L could result in G1 arrest and thereby impede the cell proliferation in vitro and tumor growth in vivo. CONCLUSIONS: Our findings first demonstrate that DOT1L over-expression has important clinical significance in ovarian cancer and also clarify that it drives cell cycle progression through transcriptional regulation of CDK6 and CCND3 through H3K79 methylation, suggesting that DOT1L might be potential target for prognostic assessment and therapeutic intervention in ovarian cancer.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Methyltransferases/genetics , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Disease-Free Survival , Female , G1 Phase , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Lymphatic Metastasis , Methyltransferases/therapeutic use , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/mortality , Prognosis , Survival Rate , Tissue Array Analysis
11.
Oncol Lett ; 12(2): 1271-1278, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27446426

ABSTRACT

Epithelial-mesenchymal transition (EMT) plays a critical role in promoting tumor invasion and metastasis. However, the key cofactors that modulate the signal transduction to induce EMT have note been fully explored to date. The present study reports that sine oculis homeobox homolog 1 (SIX1) is able to promote EMT of cervical cancer by coordinating with transforming growth factor (TGF)ß-SMAD signals. The expression of SIX1 was negatively correlated with the expression of the epithelial marker E-cadherin in two independent groups of cervical cancer specimens. SIX1 could promote the transition of mesenchymal phenotype in the presence of active TGFß signals in vitro and in vivo. TGFß-SMAD signals were required for the SIX1-mediated promotion of EMT and metastatic capacity of cervical cancer cells. Together, SIX1 and TGFß cooperated to induce more remarkable changes in the transition of phenotype than each of them alone, and coordinated to promote cell motility and tumor metastasis in cervical cancer. These results suggest that the coordination of SIX1 and TGFß signals may be crucial in the EMT program, and that SIX1/TGFß may be considered a valuable marker for evaluating the metastatic potential of cervical cancer cells, or a therapeutic target in the treatment of cervical cancer.

12.
Cancer Chemother Pharmacol ; 76(5): 897-907, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26362045

ABSTRACT

BACKGROUND: The California Cancer Consortium completed a phase I trial of E7389 (eribulin mesylate), an analog of the marine natural product halichondrin B. This trial was to determine the pharmacodynamics, pharmacokinetics, and MTD of E7389 administered by bolus injection weekly for 3 weeks out of four. METHODS: This trial included a rapid titration design. Real-time pharmacokinetics were utilized to guide dose escalation. Initially, single-patient cohorts were enrolled with intra- and inter-patient dose doubling. The second phase was a standard 3 + 3 dose escalation schedule. At the MTD, a cohort of patients was enrolled for target validation studies (separate manuscript). The starting dose was 0.125 mg/m(2), and doses were doubled within and between patients in the first phase. Blood and urine sampling for E7389 pharmacokinetics was performed on doses 1 and 3 of cycle 1. Levels were determined using a LC/MS/MS assay. RESULTS: Forty patients were entered. Thirty-eight were evaluable for toxicity and 35 for response. The rapid escalation ended with a grade 3 elevation of alkaline phosphatase at 0.5 mg/m(2)/week. The second phase ended at 2.0 mg/m(2)/week with dose-limiting toxicities of grades 3 and 4 febrile neutropenia. Other toxicities included hypoglycemia, hypophosphatemia, and fatigue. The MTD was 1.4 mg/m(2)/week. Responses included four partial responses (lung cancer [2], urothelial [1], and melanoma [1]). CONCLUSIONS: E7389 was well tolerated in this trial with the major toxicity being myelosuppression. PD shows that E7389 induces significant morphologic changes (bundle formation) in the microtubules of peripheral blood mononuclear cells and tumor cells in vivo. The data suggest that lower intra-tumoral levels of ß-tubulin III or higher intra-tumoral levels of MAP4 may correlate with response to E7389, while lower intra-tumoral levels of stathmin may be associated with progression. PK data reveal that E7389 exhibits a tri-exponential elimination from the plasma of patients receiving a rapid i.v. infusion. At sub-toxic doses, plasma concentrations of E7389 are maintained well above the levels required for activity in vitro for >72 h.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Furans/pharmacology , Ketones/pharmacology , Tubulin Modulators/pharmacology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Carcinoma/blood , Dose-Response Relationship, Drug , Drug Monitoring , Febrile Neutropenia/chemically induced , Female , Furans/adverse effects , Furans/blood , Furans/pharmacokinetics , Furans/therapeutic use , Humans , Injections, Intravenous , Ketones/adverse effects , Ketones/blood , Ketones/pharmacokinetics , Ketones/therapeutic use , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , Lymphopenia/chemically induced , Male , Melanoma/blood , Melanoma/drug therapy , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/drug therapy , Salvage Therapy , Tubulin Modulators/adverse effects , Tubulin Modulators/blood , Tubulin Modulators/pharmacokinetics , Tubulin Modulators/therapeutic use , Urologic Neoplasms/blood , Urologic Neoplasms/drug therapy
13.
Cancer Res ; 74(19): 5597-607, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25142796

ABSTRACT

Lymphatic vessels are one of the major routes for the dissemination of cancer cells. Malignant tumors release growth factors such as VEGF-C to induce lymphangiogenesis, thereby promoting lymph node metastasis. Here, we report that sine oculis homeobox homolog 1 (SIX1), expressed in tumor cells, can promote tumor lymphangiogenesis and lymph node metastasis by coordinating with TGFß to increase the expression of VEGF-C. Lymphangiogenesis and lymph node metastasis in cervical cancer were closely correlated with higher expression of SIX1 in tumor cells. By enhancing VEGF-C expression in tumor cells, SIX1 could augment the promoting effect of tumor cells on the migration and tube formation of lymphatic endothelial cells (LEC) in vitro and lymphangiogenesis in vivo. SIX1 enhanced TGFß-induced activation of SMAD2/3 and coordinated with the SMAD pathway to modulate VEGF-C expression. Together, SIX1 and TGFß induced much higher expression of VEGF-C in tumor cells than each of them alone. Despite its effect in promoting VEGF-C expression, TGFß could inhibit lymphangiogenesis by directly inhibiting tube formation by LECs. However, the increased production of VEGF-C not only directly promoted migration and tube formation of LECs but also thwarted the inhibitory effect of TGFß on LECs. That is, tumor cells that expressed high levels of SIX1 could promote lymphangiogenesis and counteract the negative effects of TGFß on lymphangiogenesis by increasing the expression of VEGF-C. These findings provide new insights into tumor lymphangiogenesis and the various roles of TGFß signaling in tumor regulation. Our results also suggest that SIX1/TGFß might be a potential therapeutic target for preventing lymph node metastasis of tumor.


Subject(s)
Homeodomain Proteins/physiology , Lymphangiogenesis/physiology , Signal Transduction , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor C/metabolism , Animals , Base Sequence , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/physiopathology
14.
Int J Oncol ; 45(3): 1232-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24970368

ABSTRACT

Malignant proliferation is the fundamental trait of tumor cells. The initiation of DNA replication represents a key process for cell proliferation, and has a marked impact on tumorigenesis and progression. Here we report that Sine oculis homeobox homolog 1 (SIX1) functions as a master regulator in DNA replication of cervical cancer cells. The expression of SIX1 was induced by the E7 oncoprotein of human papillomaviruses in cervical intraepithelial neoplasia and cervical cancer. The increase of SIX1 expression resulted in the upregulation of multiple genes related to the initiation of DNA replication, including the genes coding for the proteins in minichromosome maintenance complex (MCM2, MCM3, MCM6), DNA polymerase α-primase complex (POLA1, PRIM1, PRIM2), clamp loader (RFC3, RFC4, RFC5), DNA polymerase δ complex (POLD3) and DNA polymerase ε complex (POLE2). In line with this, the increase of SIX1 expression enhanced DNA synthesis, accelerated G1 to S phase progression, and promoted the proliferation of cervical cancer cells and the growth of cervical cancer. Consistently, knockdown of SIX1 could hamper DNA synthesis, slow down G1 to S phase progression, and suppress tumor cell proliferation and tumor growth. Importantly, SIX1 could more efficiently promote anchorage-independent cell growth. These results suggest that the increase of SIX1 expression could promote tumorigenesis, progression and invasive growth of cervical cancer by promoting DNA replication, and that targeting SIX1 may have significant therapeutic value in cervical cancer treatment.


Subject(s)
DNA Replication , Homeodomain Proteins/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Animals , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred NOD , Neoplasms, Experimental , Papillomavirus E7 Proteins/metabolism , Uterine Cervical Neoplasms/virology
15.
Biochem Biophys Res Commun ; 446(2): 549-54, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24613848

ABSTRACT

Sine oculis homeobox homolog 1 (SIX1) has been supposed to be correlated with the metastasis and poor prognosis of several malignancies. However, the effect of SIX1 on the metastatic phenotype of tumor cells and the underlying mechanisms were still unclear to date. Here we report that SIX1 can promote α5ß1-mediated metastatic capability of cervical cancer cells. SIX1 promoted the expression of α5ß1 integrin to enhance the adhesion capacity of tumor cells in vitro and tumor cell arrest in circulation in vivo. Moreover, higher expression of SIX1 in tumor cells resulted in the increased production of active MMP-2 and MMP-9, up-regulation of anti-apoptotic genes (BCL-XL and BCL2) and down-regulation of pro-apoptotic genes (BIM and BAX), thus promoting the invasive migration and anoikis-resistance of tumor cells. Importantly, blocking α5ß1 abrogated the regulatory effect of SIX1 on the expression of these genes, and also abolished the promotional effect of SIX1 on invasive capability of tumor cells. Furthermore, knock-down of α5 could abolish the promoting effect of SIX1 on the development of metastatic lesions in both experimental and spontaneous metastasis model. Therefore, by up-regulating α5ß1 expression, SIX1 not only promoted the adhesion capacity, but also augmented ECM-α5ß1-mediated regulation of gene expression to enhance the metastatic potential of cervical cancer cells. These results suggest that SIX1/α5ß1 might be considered as valuable marker for metastatic potential of cervical cancer cells, or a therapeutic target in cervical cancer treatment.


Subject(s)
Homeodomain Proteins/metabolism , Integrin alpha5beta1/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/secondary , Cell Line, Tumor , Cell Movement , Female , Humans , Neoplasm Invasiveness , Up-Regulation , Uterine Cervical Neoplasms/metabolism
16.
Free Radic Biol Med ; 57: 162-75, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23314043

ABSTRACT

Iodonium-class flavoprotein dehydrogenase inhibitors have been demonstrated to possess antiproliferative potential and to inhibit reactive oxygen production in human tumor cells, although the mechanism(s) that explains the relationship between altered cell growth and the generation of reactive oxygen species (ROS) remains an area of active investigation. Because of the ability of these compounds to inhibit the activity of flavoprotein-containing epithelial NADPH oxidases, we chose to examine the effects of several iodonium-class flavoprotein inhibitors on human colon cancer cell lines that express high, functional levels of a single such oxidase (NADPH oxidase 1, or Nox1). We found that diphenyleneiodonium (DPI), di-2-thienyliodonium (DTI), and iodonium diphenyl inhibited the growth of Caco2, HT-29, and LS-174T colon cancer cells at concentrations (10-250nM for DPI, 0.5-2.5µM for DTI, and 155nM to 10µM for iodonium diphenyl) substantially lower than needed for DU145 human prostate cancer cells, which do not possess functional NADPH oxidase activity. Drug treatment was associated with decreased H2O2 production and diminished intracellular ROS levels, lasting up to 24h, after short-term (1-h) exposure to the iodonium analogs. Decreased tumor cell proliferation was caused, in part, by a profound block in cell cycle progression at the G1/S interface in both LS-174T and HT-29 cells exposed to either DPI or DTI; and the G1 block was produced, for LS-174T cells, by upregulation of p27 and a drug concentration-related decrease in the expression of cyclins D1, A, and E that was partially prevented by exogenous H2O2. Not only did DPI and DTI decrease intracellular ROS, they both also significantly decreased the mRNA expression levels of Nox1, potentially contributing to the prolonged reduction in tumor cell reactive oxygen levels. We also found that DPI and DTI significantly decreased the growth of both HT-29 and LS-174T human tumor xenografts, at dose levels that produced peak plasma concentrations similar to those utilized for our in vitro experiments. These findings suggest that iodonium analogs have therapeutic potential for NADPH oxidase-containing human colon cancers in vivo and that at least part of their antineoplastic mechanism of action may be related to targeting Nox1.


Subject(s)
Cell Cycle/drug effects , Colonic Neoplasms/metabolism , Flavoproteins/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Reactive Oxygen Species/metabolism , Thiophenes/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclins/biosynthesis , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression , Humans , Hydrogen Peroxide/metabolism , Male , Mice , NADPH Oxidase 1 , NADPH Oxidases/genetics , Neoplasm Transplantation , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , Transplantation, Heterologous
17.
Cancer Chemother Pharmacol ; 70(6): 791-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22983015

ABSTRACT

PURPOSE: HIV protease inhibitors are associated with HIV protease inhibitor-related lipodystrophy syndrome. We hypothesized that liposarcomas would be similarly susceptible to the apoptotic effects of an HIV protease inhibitor, nelfinavir. METHODS: We conducted a phase I trial of nelfinavir for liposarcomas. There was no limit to prior chemotherapy. The starting dose was 1,250 mg twice daily (Level 1). Doses were escalated in cohorts of three to a maximally evaluated dose of 4,250 mg (Level 5). One cycle was 28 days. Steady-state pharmacokinetics (PKs) for nelfinavir and its primary active metabolite, M8, were determined at Levels 4 (3,000 mg) and 5. RESULTS: Twenty subjects (13 males) were enrolled. Median (range) age was 64 years (37-81). One subject at Level 1 experienced reversible, grade 3 pancreatitis after 1 week and was replaced. No other dose-limiting toxicities were observed. Median (range) number of cycles was 3 (0.6-13.5). Overall best responses observed were 1 partial response, 1 minor response, 4 stable disease, and 13 progressive disease. Mean peak plasma levels and AUCs for nelfinavir were higher at Level 4 (7.3 mg/L; 60.9 mg/L × h) than 5 (6.3 mg/L; 37.7 mg/L × h). The mean ratio of M8:nelfinavir AUCs for both levels was ~1:3. CONCLUSIONS: PKs demonstrate auto-induction of nelfinavir clearance at the doses studied, although the mechanism remains unclear. Peak plasma concentrations were within range where anticancer activity was demonstrated in vitro. M8 metabolite is present at ~1/3 the level of nelfinavir and may also contribute to the anticancer activity observed.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/therapeutic use , Liposarcoma/drug therapy , Nelfinavir/pharmacokinetics , Nelfinavir/therapeutic use , Adult , Aged , Aged, 80 and over , Area Under Curve , Drug Administration Schedule , Female , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV-Associated Lipodystrophy Syndrome/drug therapy , Humans , Liposarcoma/blood , Male , Middle Aged , Nelfinavir/administration & dosage , Nelfinavir/blood , Research Design , Treatment Outcome
18.
Cancer Chemother Pharmacol ; 70(4): 617-25, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22907527

ABSTRACT

PURPOSE: Pyrrole-imidazole (Py-Im) polyamides are programmable, sequence-specific DNA minor groove-binding ligands. Previous work in cell culture has shown that various polyamides can be used to modulate the transcriptional programs of oncogenic transcription factors. In this study, two hairpin polyamides with demonstrated activity against androgen receptor signaling in cell culture were administered to mice to characterize their pharmacokinetic properties. METHODS: Py-Im polyamides were administered intravenously by tail vein injection. Plasma, urine, and fecal samples were collected over a 24-h period. Liver, kidney, and lung samples were collected postmortem. Concentrations of the administered polyamide in the plasma, excretion, and tissue samples were measured using LC/MS/MS. The biodistribution data were analyzed by both non-compartmental and compartmental pharmacokinetic models. Animal toxicity experiments were also performed by monitoring weight loss after a single subcutaneous (SC) injection of either polyamide. RESULTS: The biodistribution profiles of both compounds exhibited rapid localization to the liver, kidneys, and lungs upon injection. Plasma distribution of the two compounds showed distinct differences in the rate of clearance, the volume of distribution, and the AUCs. These two compounds also have markedly different toxicities after SC injection in mice. CONCLUSIONS: The variations in pharmacokinetics and toxicity in vivo stem from a minor chemical modification that is also correlated with differing potency in cell culture. The results obtained in this study could provide a structural basis for further improvement of polyamide activity both in cell culture and in animal models.


Subject(s)
Imidazoles/pharmacokinetics , Nylons/pharmacokinetics , Pyrroles/pharmacokinetics , Animals , Female , Imidazoles/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nylons/toxicity , Pyrroles/toxicity , Tissue Distribution
19.
Clin Cancer Res ; 17(7): 1796-806, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21355074

ABSTRACT

PURPOSE: We previously reported that nelfinavir (NFV) induces G(1) cell-cycle block and apoptosis selectively in liposarcoma cell lines due to increased SREBP-1 (sterol regulatory element binding protein-1) expression in the absence of increased transcription. We postulate that NFV interferes with regulated intramembrane proteolysis of SREBP-1 and ATF6 (activating transcription factor 6). EXPERIMENTAL DESIGN: Time-lapse, confocal microscopic studies show that NFV inhibits the nuclear translocation of full-length SREBP-1-EGFP and ATF6-EGFP fusion proteins. siRNA-mediated knockdown of site-1 protease (S1P) and/or site-2 protease (S2P) leads to inhibition of SREBP-1 intracellular trafficking to the nucleus and reduces liposarcoma cell proliferation. Treatment of LiSa-2 liposarcoma cells with 3,4-dichloroisocoumarin, a serine protease inhibitor of S1P, did not affect SREBP-1 processing. In contrast, 1,10-phenanthroline, an S2P-specific inhibitor, reproduces the molecular and biological phenotypes observed in NFV-treated cells, which implicates S2P as a target of NFV. In vivo evaluation of NFV in a murine liposarcoma xenograft model leads to inhibition of tumor growth without significant toxicity. RESULTS: NFV-induced upregulation of SREBP-1 and ATF6 results from inhibition of S2P, which together with S1P mediates regulated intramembrane proteolysis from their precursor to their transcriptionally active forms. The resulting endoplasmic reticulum (ER) stress and concurrent inhibition of the unfolded protein response induce caspase-mediated apoptosis. CONCLUSIONS: These results provide new insight into the mechanism of NFV-mediated induction of ER stress and cell death in liposarcomas and are the first to report targeting S2P for cancer therapy.


Subject(s)
Activating Transcription Factor 6/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Liposarcoma/pathology , Metalloendopeptidases/antagonists & inhibitors , Nelfinavir/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Caspase 6/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Coumarins/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Enzyme Activation , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Female , Humans , Isocoumarins , Liposarcoma/drug therapy , Metalloendopeptidases/genetics , Mice , Mice, SCID , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacokinetics , Neoplasm Transplantation , Phenanthrolines/pharmacology , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Protease Inhibitors/pharmacology , Protein Transport , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transcription, Genetic , Transplantation, Heterologous , Tumor Burden/drug effects
20.
Mol Cancer Ther ; 9(6): 1669-79, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20484015

ABSTRACT

Ribonucleotide reductase (RNR) is an enzyme for the de novo conversion of ribonucleotides to deoxyribonucleotides. The two human RNR small subunits hRRM2 and hp53R2 share 83% sequence homology but show distinct expression patterns and function. Structural analyses of the oxidized form of hRRM2 and hp53R2 indicate that both proteins contain a conserved Gln127-hp53R2/Gln165-hRRM2 close to the dinuclear iron center and the essential tyrosine residue Tyr124-hp53R2/Tyr162-hRRM2 forms hydrogen bonds with the tyrosine and iron ligands, implying a critical role for the glutamine residue in assembling the dityrosyl-diiron radical cofactor. The present work also showed that Tyr221 in hRRM2, which is replaced by Phe183 in hp53R2, forms a hydrogen bond with Tyr162 to extend the hydrogen bond network from Gln165-hRRM2. Mutagenesis and spectroscopic experiments suggested that the tyrosine-to-phenylalanine switch at Phe183-hp53R2/Tyr221-hRRM2 could lead to differences in radical generation or enzymatic activity for hp53R2 and hRRM2. This study correlates the distinct catalytic mechanisms of the small subunits hp53R2 and hRRM2 with a hydrogen-bonding network and provides novel directions for designing and developing subunit-specific therapeutic agents for human RNR enzymes.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Free Radicals/metabolism , Iron Compounds/metabolism , Protein Subunits/metabolism , Ribonucleoside Diphosphate Reductase/chemistry , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Circular Dichroism , Electron Spin Resonance Spectroscopy , Humans , Molecular Sequence Data , Mutagenesis/genetics , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Subunits/chemistry , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...