Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Neurooncol ; 166(3): 419-430, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38277015

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common primary brain tumor in adults. Despite extensive research and clinical trials, median survival post-treatment remains at 15 months. Thus, all opportunities to optimize current treatments and improve patient outcomes should be considered. A recent retrospective clinical study found that taking TMZ in the morning compared to the evening was associated with a 6-month increase in median survival in patients with MGMT-methylated GBM. Here, we hypothesized that TMZ efficacy depends on time-of-day and O6-Methylguanine-DNA Methyltransferase (MGMT) activity in murine and human models of GBM. METHODS AND RESULTS: In vitro recordings using real-time bioluminescence reporters revealed that GBM cells have intrinsic circadian rhythms in the expression of the core circadian clock genes Bmal1 and Per2, as well as in the DNA repair enzyme, MGMT. Independent measures of MGMT transcript levels and promoter methylation also showed daily rhythms intrinsic to GBM cells. These cells were more susceptible to TMZ when delivered at the daily peak of Bmal1 transcription. We found that in vivo morning administration of TMZ also decreased tumor size and increased body weight compared to evening drug delivery in mice bearing GBM xenografts. Finally, inhibition of MGMT activity with O6-Benzylguanine abrogated the daily rhythm in sensitivity to TMZ in vitro by increasing sensitivity at both the peak and trough of Bmal1 expression. CONCLUSION: We conclude that chemotherapy with TMZ can be dramatically enhanced by delivering at the daily maximum of tumor Bmal1 expression and minimum of MGMT activity and that scoring MGMT methylation status requires controlling for time of day of biopsy.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Animals , Mice , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Dacarbazine/therapeutic use , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , O(6)-Methylguanine-DNA Methyltransferase/genetics , Retrospective Studies , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Methylation , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Methylation , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
2.
Altern Ther Health Med ; 29(5): 278-283, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37083649

ABSTRACT

Background and Objective: miR-22-3p functions as a tumor suppressor by targeting a variety of downstream genes, while its role and downstream targets in gastric cancer (GC) remain to be determined. We aimed to explore the role of miR-22-3p in gastric cancer and the potential mechanism. Methods: miR-22-3p mimic and inhibitor were used to overexpress or knockdown the expression of miR-22-3p separately. Quantitative real-time PCR (RT-qPCR) and Western blot were used to analyse the abundance of mRNA or protein level respectively. CCK-8 assay, cell colony formation assay, and flow cytometry were implemented to investigate the effect of miR-22-3p on gastric cancer cell proliferation and apoptosis. Luciferase assay was used to evaluate the role of miR-22-3p on the expression of glycolytic enzyme enolase 1 (ENO1). Results: In this study, we found that miR-22-3p was downregulated in GC cells. By transfecting the cells with miR-22-3p inhibitors or mimics, we showed that miR-22-3p suppressed GC cell proliferation and migration, as well as triggered cell death. In addition, we discovered that miR-22-3p was engaged in glycolysis by controlling the generation of lactate as well as the consumption of glucose. TargetScan database suggested that the ENO1 may be a target of the miR-22-3p, and the luciferase experiment verified this hypothesis. Recovery assays showed that the proliferation and migration of GC cells suppressed by miR-22-3p could be rescued by overexpression of ENO1. Conclusion: Collectively, we identified a new axis of miR-22-3p/ENO1 for GC development, which could be investigated as a therapeutic target for GC.


Subject(s)
MicroRNAs , Stomach Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Phosphopyruvate Hydratase/genetics , DNA-Binding Proteins , Biomarkers, Tumor , Tumor Suppressor Proteins/genetics
3.
BMC Endocr Disord ; 22(1): 259, 2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36284286

ABSTRACT

BACKGROUND: Parathyroid carcinoma is an uncommon cause of PTH-dependent hypercalcemia. Only a handful of cases have been reported of parathyroid carcinoma during pregnancy. CASE PRESENTATION: Twenty-four - Year - old female presented with proximal myopathy was found to have hypercalcemia. Her serum corrected total calcium was - 15 mg/dl (8.5 - 10.3), serum phosphate - 2.3 mg/dl (2.5 - 4.5), intact PTH - 118 pg/ml (20 - 80), Vitamin D - 15 ng/ml and Urine Ca/Cr ratio - 2.1 (0.1 - 0.2). Her CECT-neck revealed a well-defined mass lesion posterior to the right lobe of the thyroid - 2.6 cm × 2.5 cm × 2.9 cm in size. She was started on vitamin D supplementation, and she underwent right lower focal parathyroidectomy. Her PTH levels normalized following surgery. Her histology revealed an atypical parathyroid adenoma. She was treated with calcium and vitamin D. Her follow up was uneventful. One year following initial surgery the patient became pregnant and at 16 weeks of POA, the patient presented with a rapidly enhancing neck mass for one week duration. Her biochemical investigations were suggestive of a recurrence of primary hyperparathyroidism. Her ultrasound scan of the neck revealed a well-defined discreate hypoechoic nodule, superior to the thyroid isthmus which was confirmed by a non-contrast MRI scan of the neck. She underwent an uncomplicated second trimester parathyroid tumour excision with normalization of post op PTH. Her histology revealed a parathyroid carcinoma with vascular and capsular invasion. Her genetic studies revealed a novel frameshift mutation of the CDC73 gene. She was treated with calcium and vitamin D supplementation and closely followed up with ionized calcium and PTH levels which were normal throughout the pregnancy. She had an uncomplicated caesarean section at a POA of 37 weeks. Currently she is twelve weeks post-partum, in remission of disease. CONCLUSION: This case shows the importance of stringent follow up of atypical parathyroid adenoma patients, the benefit of second trimester surgery in management of hypercalcemia due to parathyroid carcinoma during pregnancy and the importance of identifying the novel CDC73 gene mutation.


Subject(s)
Adenoma , Hypercalcemia , Parathyroid Neoplasms , Humans , Female , Pregnancy , Parathyroid Neoplasms/complications , Parathyroid Neoplasms/genetics , Parathyroid Neoplasms/pathology , Hypercalcemia/etiology , Calcium , Cesarean Section/adverse effects , Parathyroid Hormone , Adenoma/complications , Adenoma/genetics , Adenoma/pathology , Vitamin D , Phosphates , Mutation , Tumor Suppressor Proteins/genetics
4.
Am J Hum Genet ; 109(10): 1932-1943, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36206744

ABSTRACT

Proteins containing the FERM (four-point-one, ezrin, radixin, and moesin) domain link the plasma membrane with cytoskeletal structures at specific cellular locations and have been implicated in the localization of cell-membrane-associated proteins and/or phosphoinositides. FERM domain-containing protein 5 (FRMD5) localizes at cell adherens junctions and stabilizes cell-cell contacts. To date, variants in FRMD5 have not been associated with a Mendelian disease in OMIM. Here, we describe eight probands with rare heterozygous missense variants in FRMD5 who present with developmental delay, intellectual disability, ataxia, seizures, and abnormalities of eye movement. The variants are de novo in all for whom parental testing was available (six out of eight probands), and human genetic datasets suggest that FRMD5 is intolerant to loss of function (LoF). We found that the fly ortholog of FRMD5, CG5022 (dFrmd), is expressed in the larval and adult central nervous systems where it is present in neurons but not in glia. dFrmd LoF mutant flies are viable but are extremely sensitive to heat shock, which induces severe seizures. The mutants also exhibit defective responses to light. The human FRMD5 reference (Ref) cDNA rescues the fly dFrmd LoF phenotypes. In contrast, all the FRMD5 variants tested in this study (c.340T>C, c.1051A>G, c.1053C>G, c.1054T>C, c.1045A>C, and c.1637A>G) behave as partial LoF variants. In addition, our results indicate that two variants that were tested have dominant-negative effects. In summary, the evidence supports that the observed variants in FRMD5 cause neurological symptoms in humans.


Subject(s)
Intellectual Disability , Animals , Ataxia/genetics , DNA, Complementary , Developmental Disabilities/genetics , Eye Movements , Humans , Intellectual Disability/genetics , Membrane Proteins , Phosphatidylinositols , Seizures , Tumor Suppressor Proteins/genetics
5.
J Neurosci ; 42(42): 7885-7899, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36028316

ABSTRACT

Anterolateral system (AS) neurons transmit pain signals from the spinal cord to the brain. Their morphology, anatomy, and physiological properties have been extensively characterized and suggest that specific AS neurons and their brain targets are concerned with the discriminatory aspects of noxious stimuli, such as their location or intensity, and their motivational/emotive dimension. Among the recently unraveled molecular markers of AS neurons is the developmentally expressed transcription factor Phox2a, providing us with the opportunity to selectively disrupt the embryonic wiring of AS neurons to gain insights into the logic of their adult function. As mice with a spinal-cord-specific loss of the netrin-1 receptor deleted in colorectal carcinoma (DCC) have increased AS neuron innervation of ipsilateral brain targets and defective noxious stimulus localization or topognosis, we generated mice of either sex carrying a deletion of Dcc in Phox2a neurons. Such DccPhox2a mice displayed impaired topognosis along the rostrocaudal axis but with little effect on left-right discrimination and normal aversive responses. Anatomical tracing experiments in DccPhox2a mice revealed defective targeting of cervical and lumbar AS axons within the thalamus. Furthermore, genetic labeling of AS axons revealed their expression of DCC on their arrival in the brain, at a time when many of their target neurons are being born and express Ntn1 Our experiments suggest a postcommissural crossing function for netrin-1:DCC signaling during the formation of somatotopically ordered maps and are consistent with a discriminatory function of some of the Phox2a AS neurons.SIGNIFICANCE STATEMENT How nociceptive (pain) signals are relayed from the body to the brain remains an important question relevant to our understanding of the basic physiology of pain perception. Previous studies have demonstrated that the AS is a main effector of this function. It is composed of AS neurons located in the spinal cord that receive signals from nociceptive sensory neurons that detect noxious stimuli. In this study, we generate a genetic miswiring of mouse AS neurons that results in a decreased ability to perceive the location of a painful stimulus. The precise nature of this defect sheds light on the function of different kinds of AS neurons and how pain information may be organized.


Subject(s)
Colorectal Neoplasms , Nerve Growth Factors , Animals , Mice , Colorectal Neoplasms/metabolism , DCC Receptor/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nerve Growth Factors/metabolism , Netrin Receptors/metabolism , Netrin-1 , Neurons/physiology , Pain/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Thalamus
6.
Nature ; 607(7918): 313-320, 2022 07.
Article in English | MEDLINE | ID: mdl-35768506

ABSTRACT

The grey wolf (Canis lupus) was the first species to give rise to a domestic population, and they remained widespread throughout the last Ice Age when many other large mammal species went extinct. Little is known, however, about the history and possible extinction of past wolf populations or when and where the wolf progenitors of the present-day dog lineage (Canis familiaris) lived1-8. Here we analysed 72 ancient wolf genomes spanning the last 100,000 years from Europe, Siberia and North America. We found that wolf populations were highly connected throughout the Late Pleistocene, with levels of differentiation an order of magnitude lower than they are today. This population connectivity allowed us to detect natural selection across the time series, including rapid fixation of mutations in the gene IFT88 40,000-30,000 years ago. We show that dogs are overall more closely related to ancient wolves from eastern Eurasia than to those from western Eurasia, suggesting a domestication process in the east. However, we also found that dogs in the Near East and Africa derive up to half of their ancestry from a distinct population related to modern southwest Eurasian wolves, reflecting either an independent domestication process or admixture from local wolves. None of the analysed ancient wolf genomes is a direct match for either of these dog ancestries, meaning that the exact progenitor populations remain to be located.


Subject(s)
Dogs , Genome , Genomics , Phylogeny , Wolves , Africa , Animals , DNA, Ancient/analysis , Dogs/genetics , Domestication , Europe , Genome/genetics , History, Ancient , Middle East , Mutation , North America , Selection, Genetic , Siberia , Tumor Suppressor Proteins/genetics , Wolves/classification , Wolves/genetics
7.
Cell Mol Gastroenterol Hepatol ; 13(4): 1201-1221, 2022.
Article in English | MEDLINE | ID: mdl-34920145

ABSTRACT

BACKGROUND & AIMS: TOB1 is an anti-proliferative protein of Tob/BTG family and typically involved in the tumorigenesis and T cell activation. Although TOB1 is associated with T helper 17 cell-related autoimmunity, its role in modulating T cell-mediated immune responses in IBD remains poorly understood. Here, we explored its expression and the underlying mechanisms involved in the pathogenesis of inflammatory bowel disease (IBD). METHODS: TOB1 and ID2 expression in IBD patients was examined by quantitative real time polymerase chain reaction and immunohistochemistry. IBD CD4+ T cells were transfected with lentivirus expressing TOB1, ID2, TOB1 short hairpin RNA and ID2 short hairpin RNA, respectively, and Tob1-/-CD4+ T cells were transfected with lentivirus expressing Id2. Experimental colitis was established in Tob1-/- mice by trinitrobenzene sulfonic acid enema and in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells to further explore the role of Tob1 in intestinal mucosal inflammation. Splenic CD4+ T cells of Tob1-/- mice were sorted to determine transcriptome differences by RNA sequencing. RESULTS: TOB1 expression was decreased in inflamed mucosa and peripheral blood CD4+ T cells of IBD patients compared with healthy subjects. Overexpression of TOB1 downregulated IBD CD4+ T cells to differentiate into Th1/Th17 cells compared with control subjects. Severe colitis was observed in Tob1-/- mice through trinitrobenzene sulfonic acid enema or in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells, compared with control animals. RNA sequencing analysis revealed ID2 as functional target of TOB1 to inhibit IBD CD4+ T cell differentiation into Th1/Th17 cells. Mechanistically, TOB1 was associated with Smad4/5 to induce ID2 expression and restrain Th1/Th17 cell differentiation. CONCLUSIONS: TOB1 restrains intestinal mucosal inflammation through suppressing Th1/Th17 cell-mediated immune responses via the Smad4/5-ID2 pathway. It may serve as a novel therapeutic target for treatment of human IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Homeodomain Proteins/metabolism , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Intestinal Mucosa/metabolism , Lymphocyte Activation , Mice , RNA, Small Interfering/metabolism , Sulfonic Acids/metabolism , Sulfonic Acids/therapeutic use , Th1 Cells , Th17 Cells/metabolism , Th17 Cells/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
JAMA Netw Open ; 4(11): e2132615, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34767027

ABSTRACT

Importance: In BAP1 tumor predisposition syndrome, clear cell renal cell carcinoma (RCC) is frequently associated with melanoma and/or mesothelioma, while germline MITF p.E318K alterations are being increasingly reported in melanoma/RCC. Limited data exist on the co-occurrence of melanoma and/or mesothelioma with renal neoplasia and the prevalence of associated germline alterations. Objective: To assess the frequency of melanoma and/or mesothelioma co-occurring with renal neoplasia using our institutional nephrectomy registry and to determine the prevalence of BAP1 and MITF alterations within this cohort. Design, Setting, and Participants: In this genetic association study, medical records from 8295 patients from 1970 to 2018, renal neoplasia co-occurring with melanoma and/or mesothelioma within a single institutional nephrectomy registry was reevaluated based on contemporary histopathologic criteria and the medical records were reviewed. Data were analyzed from September 2019 to May 2021. Main Outcomes and Measures: Identified cases were screened for BAP1 loss using immunohistochemistry; while patients with melanoma and clear cell RCC were screened for MITF p.E318K alterations. Tumors from patients with potential germline alterations were analyzed with comprehensive molecular profiling using a 514-gene next generation sequencing panel. Results: Of a total of 8295 patients, 93 (1.1%; 95% CI, 0.9%-1.4%) had melanoma and/or mesothelioma co-occurring with renal neoplasia (cutaneous melanoma, n = 76; uveal melanoma, n = 11; mesothelioma, n = 6). A total of 69 (74.2%) were male; 24 (25.8%) were female; median age at diagnosis of renal neoplasia was 63 years (IQR, 58-70 years) and the median duration of follow-up was 8.5 years (IQR, 5.0-14.6 years). Two patients with clear cell RCC had germline BAP1 alterations in the setting of cutaneous melanoma and mesothelioma. Two patients with hybrid oncocytic tumors had biallelic inactivation of FLCN in a setting of Birt-Hogg-Dubé (BHD) syndrome associated with uveal melanoma and mesothelioma. Tumor-only screening of clear cell RCC associated with cutaneous (n = 53) and uveal melanoma (n = 6) led to the identification of 1 patient with a likely germline MITF p.E318K alteration. After excluding benign renal neoplasia (such as oncocytoma and angiomyolipoma), alterations of BAP1, FLCN, and MITF were identified in 5 of 81 patients (6.2%) with melanoma and/or mesothelioma and renal neoplasia. In contrast to hybrid oncocytic tumors in BHD, no unique genotype-phenotype correlations were seen for clear cell RCC with pathogenic BAP1/ MITF alterations and VHL loss of function variants. Four of 5 cases (80%) met current National Comprehensive Cancer Network criteria for germline testing based on a combination of age, multifocality, histologic findings, and family history. Conclusions and Relevance: In this genetic association study, findings support the continued use of these National Comprehensive Cancer Network criteria and suggest more stringent screening may be warranted in this patient population.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Kidney Neoplasms/genetics , Melanoma/genetics , Mesothelioma/genetics , Microphthalmia-Associated Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Adult , Aged , Aged, 80 and over , Female , Germ-Line Mutation , Humans , Kidney Neoplasms/complications , Kidney Neoplasms/epidemiology , Kidney Neoplasms/pathology , Male , Melanoma/complications , Melanoma/epidemiology , Melanoma/pathology , Mesothelioma/complications , Mesothelioma/epidemiology , Mesothelioma/pathology , Middle Aged , Minnesota/epidemiology , Proto-Oncogene Proteins , Registries
9.
J Biol Chem ; 297(5): 101223, 2021 11.
Article in English | MEDLINE | ID: mdl-34597666

ABSTRACT

Malignant pleural mesothelioma (MPM) is a rare, aggressive, and incurable cancer arising from the mesothelial lining of the pleura, with few available treatment options. We recently reported that loss of function of the nuclear deubiquitinase BRCA1-associated protein 1 (BAP1), a frequent event in MPM, is associated with sensitivity to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis. As a potential underlying mechanism, here we report that BAP1 negatively regulates the expression of TRAIL receptors: death receptor 4 (DR4) and death receptor 5 (DR5). Using tissue microarrays of tumor samples from MPM patients, we found a strong inverse correlation between BAP1 and TRAIL receptor expression. BAP1 knockdown increased DR4 and DR5 expression, whereas overexpression of BAP1 had the opposite effect. Reporter assays confirmed wt-BAP1, but not catalytically inactive BAP1 mutant, reduced promoter activities of DR4 and DR5, suggesting deubiquitinase activity is required for the regulation of gene expression. Co-immunoprecipitation studies demonstrated direct binding of BAP1 to the transcription factor Ying Yang 1 (YY1), and chromatin immunoprecipitation assays revealed BAP1 and YY1 to be enriched in the promoter regions of DR4 and DR5. Knockdown of YY1 also increased DR4 and DR5 expression and sensitivity to TRAIL. These results suggest that BAP1 and YY1 cooperatively repress transcription of TRAIL receptors. Our finding that BAP1 directly regulates the extrinsic apoptotic pathway will provide new insights into the role of BAP1 in the development of MPM and other cancers with frequent BAP1 mutations.


Subject(s)
Mesothelioma, Malignant/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , YY1 Transcription Factor/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mesothelioma, Malignant/genetics , Mutation , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , YY1 Transcription Factor/genetics
10.
Pharmacol Res ; 173: 105889, 2021 11.
Article in English | MEDLINE | ID: mdl-34536548

ABSTRACT

Iron is an indispensable requirement for essential biological processes in cancer cells. Due to the greater proliferation of neoplastic cells, their demand for iron is considerably higher relative to normal cells, making them highly susceptible to iron depletion. Understanding this sensitive relationship led to research exploring the effect of iron chelation therapy for cancer treatment. The classical iron-binding ligand, desferrioxamine (DFO), has demonstrated effective anti-proliferative activity against many cancer-types, particularly neuroblastoma tumors, and has the surprising activity of down-regulating the potent oncogene, N-myc, which is a major oncogenic driver in neuroblastoma. Even more significant is the ability of DFO to simultaneously up-regulate the potent metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), which plays a plethora of roles in suppressing a variety of oncogenic signaling pathways. However, DFO suffers the disadvantage of demonstrating poor membrane permeability and short plasma half-life, requiring administration by prolonged subcutaneous or intravenous infusions. Considering this, the specifically designed di-2-pyridylketone thiosemicarbazone (DpT) series of metal-binding ligands was developed in our laboratory. The lead agent from the first generation DpT series, di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT), showed exceptional anti-cancer properties compared to DFO. However, it exhibited cardiotoxicity in mouse models at higher dosages. Therefore, a second generation of agents was developed with the lead compound being di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) that progressed to Phase I clinical trials. Importantly, DpC showed better anti-proliferative activity than Dp44mT and no cardiotoxicity, demonstrating effective anti-cancer activity against neuroblastoma tumors in vivo.


Subject(s)
Iron Chelating Agents/therapeutic use , Neuroblastoma/drug therapy , Animals , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc , Humans , Iron Chelating Agents/pharmacology , Neuroblastoma/genetics , Neuroblastoma/pathology , Oncogenes , Therapies, Investigational , Tumor Suppressor Proteins/genetics , Up-Regulation/drug effects
11.
Nutrients ; 13(8)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34444824

ABSTRACT

Glioblastoma (GBM) is an aggressive tumor with a dismal prognosis. Neural stem-like cells contribute to GBM's poor prognosis by driving drug resistance and maintaining cellular heterogeneity. GBM neural stem-like cells express high levels of brain fatty acid-binding protein (FABP7), which binds to polyunsaturated fatty acids (PUFAs) ω-6 arachidonic acid (AA) and ω-3 docosahexaenoic acid (DHA). Similar to brain, GBM tissue is enriched in AA and DHA. However, DHA levels are considerably lower in GBM tissue compared to adult brain. Therefore, it is possible that increasing DHA content in GBM, particularly in neural stem-like cells, might have therapeutic value. Here, we examine the fatty acid composition of patient-derived GBM neural stem-like cells grown as neurosphere cultures. We also investigate the effect of AA and DHA treatment on the fatty acid profiles of GBM neural stem-like cells with or without FABP7 knockdown. We show that DHA treatment increases DHA levels and the DHA:AA ratio in GBM neural stem-like cells, with FABP7 facilitating the DHA uptake. We also found that an increased uptake of DHA inhibits the migration of GBM neural stem-like cells. Our results suggest that increasing DHA content in the GBM microenvironment may reduce the migration/infiltration of FABP7-expressing neural stem-like cancer cells.


Subject(s)
Brain Neoplasms/metabolism , Docosahexaenoic Acids/metabolism , Fatty Acid-Binding Protein 7/metabolism , Glioblastoma/metabolism , Tumor Suppressor Proteins/metabolism , Arachidonic Acid/metabolism , Biological Transport , Brain/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Docosahexaenoic Acids/pharmacology , Fatty Acid-Binding Protein 7/genetics , Fatty Acid-Binding Proteins/metabolism , Fatty Acids , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Phospholipids/metabolism , Prognosis , Tumor Microenvironment/drug effects , Tumor Suppressor Proteins/genetics
12.
Curr Treat Options Oncol ; 22(10): 85, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34424438

ABSTRACT

OPINION STATEMENT: Since the 2013 Supreme Court declaration, panel testing for hereditary cancer syndromes has evolved into the gold standard for oncology germline genetic testing. With the advent of next-generation sequencing, competitive pricing, and developing therapeutic options, panel testing is now well integrated into breast cancer management and surveillance. Although many established syndromes have well-defined cancer risks and management strategies, several breast cancer genes are currently classified as limited-evidence genes by the National Comprehensive Cancer Network (NCCN). Follow-up for individuals with mutations in these genes is a point of contention due to conflicting information in the literature. The most recent NCCN guidelines have stratified management based on gene-specific cancer risks indicating that expanding data will allow for better recommendations as research progresses. The evolving management for these genes emphasizes the clinicians' need for evidence-based understanding of low penetrance breast cancer genes and their implications for patient care. This article reviews current literature for limited evidence genes, detailing cancer risks, association with triple-negative breast cancer, and recommendations for surveillance. A brief review of the challenges and future directions is outlined to discuss the evolving nature of cancer genetics and the exciting opportunities that can impact management.


Subject(s)
Breast Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Population Surveillance , Cell Cycle Proteins/genetics , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Female , Genetic Predisposition to Disease , Genetic Testing , Germ-Line Mutation , Humans , Nuclear Proteins/genetics , Penetrance , RNA Helicases/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
13.
Int J Mol Med ; 48(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34278452

ABSTRACT

Banxia xiexin decoction (BXXX) is a classic preparation used to treat gastrointestinal diseases, and also has certain therapeutic effects on gastrointestinal tumors. BXXX has been reported to regulate the expression of proteins associated with drug resistance and sensitivity in tumors, and thus, the aim of the present study was to investigate the mechanisms of BXXX drug sensitivity in gastric cancer (GC). The expression levels of programmed cell death 1 ligand 1 (PD­L1), 6­O­methylguanine­DNA methyltransferase (MGMT) and STAT3 were immunohistochemically detected in the cancer and adjacent non­cancer tissues of patients with GC, and in vitro experimentation was conducted using drug­resistant and ­sensitive GC cells. The expression levels of PD­L1, MGMT and STAT3 were determined using reverse transcription­quantitative PCR. Different concentrations of BXXX drug serum were used to treat the cells and the cellular inhibition rate was assessed using a Cell Counting Kit­8 assay. Flow cytometry was used to detect apoptosis, and western blot analysis was used to detect the expression levels of IL­6, IFN­Î³, JAK/STAT3 pathway proteins, PD­L1 and MGMT. The association between PD­L1 and MGMT protein expression levels was subsequently assessed via co­immunoprecipitation. Furthermore, in vivo studies were conducted following the establishment of a drug­resistant tumor­bearing mouse model, where GC tumor size was assessed under different treatment conditions, and western blot analysis was used to detect the expression of related pathway proteins. The expression levels of PD­L1, MGMT and STAT3 were significantly increased in GC tissues, GC cells and cisplatin­resistant cells. Furthermore, BXXX inhibited the proliferation of drug­resistant cells and promoted the inhibitory effects of chemotherapeutic drugs on drug­resistant cells. BXXX also inhibited the expression levels of IL­6, IFN­Î³ and JAK/STAT3 pathway proteins, as well as the expression levels of PD­L1 and MGMT. Colivelin, an activator of STAT3, reversed the effects of BXXX on drug­resistant GC cells, and significantly reversed the effect of BXXX on PD­L1 expression. In conclusion, BXXX was found to influence the drug sensitivity of GC cells by regulating the expression of MGMT. This process functions viaPD­L1, which was itself mediated by IL­6/JAK/STAT3 signaling.


Subject(s)
B7-H1 Antigen/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drugs, Chinese Herbal/pharmacology , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Animals , B7-H1 Antigen/metabolism , Cell Line, Tumor , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/metabolism , Janus Kinases/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays/methods
14.
Mol Med Rep ; 23(6)2021 06.
Article in English | MEDLINE | ID: mdl-33786612

ABSTRACT

Dioscin, an extract from traditional Chinese herbal plants, displays various biological and pharmacological effects on tumors, including inhibition of cell proliferation and induction of DNA damage. However, the effects of dioscin on oral squamous cell carcinoma (OSCC) cells are not completely understood. The present study aimed to evaluate the impact of dioscin on OSCC cell proliferation. Cell Counting Kit­8 and 5­ethynyl­2'­deoxyuridine incorporation assays were performed to assess cell proliferation. Flow cytometry was conducted to detect alterations in the cell cycle and cell apoptosis. Western blotting and coimmunoprecipitation were performed to determine protein expression levels. In SCC15 cells, dioscin treatment significantly induced cell cycle arrest, increased apoptosis and inhibited proliferation compared with the control group. Mechanistically, the tumor suppressor protein Ras association domain­containing protein 1A (RASSF1A) was activated and oncoprotein yes­associated protein (YAP) was phosphorylated by dioscin. Furthermore, YAP overexpression and knockdown reduced and enhanced the inhibitory effects of dioscin on SCC15 cells, respectively. In summary, the results demonstrated that, compared with the control group, dioscin upregulated RASSF1A expression in OSCC cells, which resulted in YAP phosphorylation, thus weakening its transcriptional coactivation function, enhancing cell cycle arrest and apoptosis, and inhibiting cell proliferation. The present study indicated that dioscin may serve as a therapeutic agent for OSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/drug effects , Diosgenin/analogs & derivatives , Mouth Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Cell Cycle Checkpoints , Cell Line, Tumor , Diosgenin/pharmacology , Humans , Protein Serine-Threonine Kinases/metabolism , Serine-Threonine Kinase 3 , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
15.
J Mol Neurosci ; 71(8): 1598-1604, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33523385

ABSTRACT

The 2016 World Health Organization classification of central nervous system tumor firstly introduces molecular diagnosis to glioma, while the molecular features of adult thalamic gliomas (ATGs) in a relatively large sample have not been reported. We aimed at exploring molecular characteristics in ATGs. The data of 97 and 575 newly diagnosed ATGs and superficial gliomas (SGs) patients were collected, and we performed a comparative analysis of molecular characteristics between them. We analyzed expressions of molecules as follow: H3 K27M, isocitrate dehydrogenase1 (IDH1), Ki-67, O6-Methylguanine-DNA methyltransferase (MGMT) promoter, EGFR, p53, ATRX, GFAP, Oligo2, PTEN, MGMT, and MMP9 by immunohistochemistry. Direct gene sequencing was performed to test the H3 K27M, IDH1, and TERT promoter mutation. The median age at diagnosis of ATGs was 36.0 years, and majority of them were high-grade glioma. We found a significant difference in H3 K27M mutation (P = 0.003), IDH1 mutation (P < 0.001), MGMT promoter methylation (P = 0.005), and Ki67 > 0.1 (P < 0.001) between ATGs and SGs. The statuses of IDH1 (P < 0.001), MGMT promoter (P < 0.001), and Ki67 (P < 0.001) were significantly different between these two groups in lower-grade gliomas. And statuses of IDH1 (P < 0.001), Ki67 (P < 0.001), and EGFR (P = 0.032) were different between these two groups in high-grade gliomas. Only Ki67 > 0.1 was differentially expressed between lower- and high-grade gliomas in ATGs (P = 0.014). The high occurrence of H3 K27M mutation and Ki67 > 0.1, rare occurrence of IDH1 mutation, and MGMT promoter methylation in ATGs suggested that ATGs may be a distinct type of glioma entity.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Thalamus/metabolism , Adult , Aged , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glioma/genetics , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Middle Aged , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
16.
Int J Mol Sci ; 21(15)2020 Aug 04.
Article in English | MEDLINE | ID: mdl-32759702

ABSTRACT

Vitamin A is an important regulator of immune protection, but it is often overlooked in studies of infectious disease. Vitamin A binds an array of nuclear receptors (e.g., retinoic acid receptor, peroxisome proliferator-activated receptor, retinoid X receptor) and influences the barrier and immune cells responsible for pathogen control. Children and adults in developed and developing countries are often vitamin A-deficient or insufficient, characteristics associated with poor health outcomes. To gain a better understanding of the protective mechanisms influenced by vitamin A, we examined immune factors and epithelial barriers in vitamin A deficient (VAD) mice, vitamin D deficient (VDD) mice, double deficient (VAD+VDD) mice, and mice on a vitamin-replete diet (controls). Some mice received insults, including intraperitoneal injections with complete and incomplete Freund's adjuvant (emulsified with PBS alone or with DNA + Fus-1 peptide) or intranasal inoculations with Sendai virus (SeV). Both before and after insults, the VAD and VAD+VDD mice exhibited abnormal serum immunoglobulin isotypes (e.g., elevated IgG2b levels, particularly in males) and cytokine/chemokine patterns (e.g., elevated eotaxin). Even without insult, when the VAD and VAD+VDD mice reached 3-6 months of age, they frequently exhibited opportunistic ascending bacterial urinary tract infections. There were high frequencies of nephropathy (squamous cell hyperplasia of the renal urothelium, renal scarring, and ascending pyelonephritis) and death in the VAD and VAD+VDD mice. When younger VAD mice were infected with SeV, the predominant lesion was squamous cell metaplasia of respiratory epithelium in lungs and bronchioles. Results highlight a critical role for vitamin A in the maintenance of healthy immune responses, epithelial cell integrity, and pathogen control.


Subject(s)
Vitamin A Deficiency/genetics , Vitamin A/genetics , Vitamin D Deficiency/genetics , Vitamin D/genetics , Animals , Communicable Diseases/genetics , Communicable Diseases/immunology , Communicable Diseases/metabolism , Death , Disease Models, Animal , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Mice , Mice, Knockout , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/immunology , Neoplasms, Squamous Cell/metabolism , Tumor Suppressor Proteins/genetics , Vitamin A/metabolism , Vitamin A Deficiency/immunology , Vitamin A Deficiency/metabolism , Vitamin D/metabolism , Vitamin D Deficiency/immunology , Vitamin D Deficiency/metabolism
17.
Cancer Biomark ; 28(2): 201-211, 2020.
Article in English | MEDLINE | ID: mdl-32224527

ABSTRACT

BACKGROUND: Zinc finger MYND (Myeloid, Nervy and DEAF-1)-type containing 8 (ZMYND8) is closely correlated with tumor proliferation and invasiveness. However, its prognostic value has not been estimated in colorectal cancer (CRC). OBJECTIVE: We aimed to elucidate the prognostic significance of ZMYND8 expression and the pN and pM classification supplemented by its expression in CRCs. METHODS: The candidate gene ZMYND8 is identified by TCGA database and GEO database, and then we retrospectively evaluated the status and prognostic significance of ZMYND8 expression of 174 patients with CRC. RESULTS: Online data showed high expression of ZMYND8 is closely correlated with worse overall survival. Our study revealed high expression of ZMYND8 in CRC patients was significantly associated with worse overall and disease-free survival (P< 0.05), and was an independently adverse prognostic factor for overall survival (P< 0.001) and disease-free survival (P= 0.001) by univariate and multivariate analysis. C-index to combined prognostic model containing the pN, pM classification supplemented by the status of ZMYND8 expression showed improved predictive ability comparing with the pN and pM classification model (C-index of 0.597 vs. 0.545, respectively). CONCLUSION: The combined prognostic model could improve the ability to determine the clinical outcome of patients with CRC.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Cell Line, Tumor , Colon/pathology , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Computational Biology , Datasets as Topic , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Models, Biological , Neoplasm Staging , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Assessment/methods , Up-Regulation
18.
J Cell Mol Med ; 24(8): 4819-4829, 2020 04.
Article in English | MEDLINE | ID: mdl-32168425

ABSTRACT

Psoriasis is a chronic immune-mediated inflammatory dermatosis. Recently, ozone therapy has been applicated to psoriasis treatment; however, the mechanism by which ozone therapy improves psoriasis remains unclear. The excessive proliferation and the differentiation of basal keratinocytes have been considered critical issues during pathological psoriasis process, in which keratin 6 (KRT6) and KRT10 might be involved. In the present study, KRT6, IL-17 and IL-22 protein within psoriasis lesions was decreased, while KRT10 and Tp63 protein in psoriasis lesions was increased by ozone treatment in both patient and IMQ mice psoriatic tissues. In the meantime, ozone treatment down-regulated KRT6 mRNA and protein expression while up-regulated KRT10 mRNA and protein expression within IL-22 treated primary KCs; the cell viability of KCs was suppressed by ozone treatment. Moreover, Tp63 bound to KRT10 promoter region to activate its transcription in basal keratinocytes; the promotive effects of ozone on Tp63 and KRT10 were significantly reversed by Tp63 silence. Both TP63 and KRT10 mRNA expression were significantly increased by ozone treatment in psoriasis lesions; there was a positive correlation between Tp63 and KRT10 expression within tissue samples, suggesting that ozone induces the expression of Tp63 to enhance the expression of KRT10 and the differentiation of keratinocytes, therefore improving the psoriasis. In conclusion, the application of ozonated oil could be an efficient and safe treatment for psoriasis; ozone promotes the differentiation of keratinocytes via increasing Tp63-mediated transcription of KRT10, therefore improving psoriasis.


Subject(s)
Keratin-10/genetics , Keratin-6/genetics , Ozone/pharmacology , Psoriasis/therapy , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adult , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dermatitis/genetics , Dermatitis/pathology , Dermatitis/therapy , Disease Models, Animal , Female , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Male , Mice , Ozone/therapeutic use , Primary Cell Culture , Psoriasis/genetics , Psoriasis/pathology , Skin/drug effects , Skin/pathology
19.
Integr Cancer Ther ; 19: 1534735419900804, 2020.
Article in English | MEDLINE | ID: mdl-32046536

ABSTRACT

The traditional Chinese medicine formula Jianpi-Huayu (JPHY) has been reported to be effective in the treatment of hepatocellular carcinoma (HCC). However, its underlying mechanism remains unclear. In this article, we employed an orthotopic transplantation model in nude mice to explore whether JPHY could inhibit the development of HCC by regulating miR-602, which targets the Ras association domain-containing protein 1A (RASSF1A) pathway. HCC SMMC-7721 cells were treated with JPHY to test whether the RASSF1A gene as mediated by miR-602 affected the proliferation and apoptosis of tumor cells. We subsequently detected miR-602, RASSF1A, and tumor cell apoptosis-related markers in cells and liver tumor tissues. We observed that mice treated with JPHY had smaller tumors and higher survival rates than untreated ones. Similarly, JPHY-treated SMMC-7721 cells exhibited alterations in morphology and higher cytotoxicity compared with the control group. Furthermore, we found that JPHY decreased overexpression of miR-602 and increased protein expression levels of the RASS1A gene, which in turn altered protein expression levels of tumor cell apoptosis-related genes in the cells and liver tumor tissues of drug-treated mice. These results indicated that JPHY could potentially be used to treat HCC by targeting miR-602, which targets the RASSF1A gene, which in turn plays a major role in HCC pathogenesis.


Subject(s)
Carcinoma, Hepatocellular , Drugs, Chinese Herbal/pharmacology , MicroRNAs/genetics , Signal Transduction/drug effects , Tumor Suppressor Proteins/genetics , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Mice, Nude
20.
Amino Acids ; 52(1): 73-85, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31853708

ABSTRACT

Statin-induced myopathy affects more than 10 million people worldwide. But discontinuation of statin treatment increases mortality and cardiovascular events. Recently, L-arginine:glycine amidinotransferase (AGAT) gene was associated with statin-induced myopathy in two populations, but the causal link is still unclear. AGAT is responsible for the synthesis of L-homoarginine (hArg) and guanidinoacetate (GAA). GAA is further methylated to creatine (Cr) by guanidinoacetate methyltransferase (GAMT). In cerebrovascular patients treated with statin, lower hArg and GAA plasma concentrations were found than in non-statin patients, indicating suppressed AGAT expression and/or activity (n = 272, P = 0.033 and P = 0.039, respectively). This observation suggests that statin-induced myopathy may be associated with AGAT expression and/or activity in muscle cells. To address this, we studied simvastatin-induced myopathy in AGAT- and GAMT-deficient mice. We found that simvastatin induced muscle damage and reduced AGAT expression in wildtype mice (myocyte diameter: 34.1 ± 1.3 µm vs 21.5 ± 1.3 µm, P = 0.026; AGAT expression: 1.0 ± 0.3 vs 0.48 ± 0.05, P = 0.017). Increasing AGAT expression levels of transgenic mouse models resulted in rising plasma levels of hArg and GAA (P < 0.01 and P < 0.001, respectively). Simvastatin-induced motor impairment was exacerbated in AGAT-deficient mice compared with AGAT-overexpressing GAMT-/- mice and therefore revealed an effect independent of Cr. But Cr supplementation itself improved muscle strength independent of AGAT expression (normalized grip strength: 55.8 ± 2.9% vs 72.5% ± 3.0%, P < 0.01). Homoarginine supplementation did not affect statin-induced myopathy in AGAT-deficient mice. Our results from clinical and animal studies suggest that AGAT expression/activity and its product Cr influence statin-induced myopathy independent of each other. The interplay between simvastatin treatment, AGAT expression and activity, and Cr seems to be complex. Further clinical pharmacological studies are needed to elucidate the underlying mechanism(s) and to evaluate whether supplementation with Cr, or possibly GAA, in patients under statin medication may reduce the risk of muscular side effects.


Subject(s)
DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Guanidinoacetate N-Methyltransferase/genetics , Muscle, Skeletal/drug effects , Simvastatin/pharmacology , Tumor Suppressor Proteins/genetics , Animals , Arginine/metabolism , Creatine/metabolism , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Gene Expression Regulation/drug effects , Guanidinoacetate N-Methyltransferase/deficiency , Homoarginine/metabolism , Humans , Mice , Muscle, Skeletal/metabolism , Phenotype , Tumor Suppressor Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL