1
|
Wang X, Zhao J, Zhang Y, Liu Y, Wang J, Shi R, Yuan J, Meng K. Molecular mechanism of Wilms' tumor (Wt1) (+/-KTS) variants promoting proliferation and migration of ovarian epithelial cells by bioinformatics analysis. J Ovarian Res 2023; 16:46. [PMID: 36829196 PMCID: PMC9951437 DOI: 10.1186/s13048-023-01124-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/20/2023] [Indexed: 02/26/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is a gynecological disease with the highest mortality. With the lack of understanding of its pathogenesis, no accurate early diagnosis and screening method has been established for EOC. Studies revealed the multi-faceted function of Wilms' tumor (Wt1) genes in cancer, which may be related to the existence of multiple alternative splices. Our results show that Wt1 (+KTS) or Wt1 (-KTS) overexpression can significantly promote the proliferation and migration of human ovarian epithelial cells HOSEpiC, and Wt1 (+KTS) effects were more evident. To explore the Wt1 (+/-KTS) variant mechanism in HOSEpiC proliferation and migration and ovarian cancer (OC) occurrence and development, this study explored the differential regulation of Wt1 (+/-KTS) in HOSEpiC proliferation and migration by transcriptome sequencing. OC-related hub genes were screened by bioinformatics analysis to further explore the differential molecular mechanism of Wt1 (+/-KTS) in the occurrence of OC. Finally, we found that the regulation of Wt1 (+/-KTS) variants on the proliferation and migration of HOSEpiC may act through different genes and signaling pathways and screened out key genes and differentially regulated genes that regulate the malignant transformation of ovarian epithelial cells. The implementation of this study will provide new clues for the early diagnosis and precise treatment of OC.
Collapse
Affiliation(s)
- Xiaomei Wang
- grid.449428.70000 0004 1797 7280College of Basic Medicine, Jining Medical University, Jining, China
| | - Jingyu Zhao
- grid.449428.70000 0004 1797 7280Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China ,grid.449428.70000 0004 1797 7280College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Yixin Zhang
- grid.449428.70000 0004 1797 7280Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China ,grid.449428.70000 0004 1797 7280College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Yuxin Liu
- grid.449428.70000 0004 1797 7280Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China ,grid.449428.70000 0004 1797 7280College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Jinzheng Wang
- grid.449428.70000 0004 1797 7280Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China ,grid.449428.70000 0004 1797 7280College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Ruoxi Shi
- grid.449428.70000 0004 1797 7280Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China ,grid.449428.70000 0004 1797 7280College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Jinxiang Yuan
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China. .,Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China.
| | - Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China. .,Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China.
| |
Collapse
|
2
|
Araújo Galdino O, de Souza Gomes I, Ferreira de Almeida Júnior R, Conceição Ferreira de Carvalho MI, Abreu BJ, Abbott Galvão Ururahy M, Cabral B, Zucolotto Langassner SM, Costa de Souza KS, Augusto de Rezende A. The nephroprotective action of Passiflora edulis in streptozotocin-induced diabetes. Sci Rep 2022; 12:17546. [PMID: 36266308 PMCID: PMC9584925 DOI: 10.1038/s41598-022-21826-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/04/2022] [Indexed: 01/13/2023] Open
Abstract
In the present study, we aimed to evaluate the therapeutic effect of Passiflora edulis fruit peel aqueous (AFA) extract as an adjuvant to insulin to confer nephroprotection against streptozotocin-induced diabetes. Male Wistar rats were divided into four groups based on treatment received for 60 days: diabetic (DB), control (CTL), insulin (INS), and insulin + AFA extract (INS + AFA). mRNA and protein expression levels of podocyte (nephrin, podocin, and WT1) and tubular (megalin) proteins were measured in kidney tissue specimens and urine. Biochemical parameters and kidney histopathology were also examined. Herein, the INS + AFA group showed superior glycemic control, which resulted in the reduction of urinary albumin/creatinine ratio, maintenance of baseline levels of Nphs1, Nphs2, Wt1, and Lrp2 mRNA expression, prevention of protein loss from the kidney tissue into the urinary space, along with the maintenance of glomerular basement membrane thickness, hyalinization, glomerular and tubulointerstitial fibrosis at values approximating those of the CTL group and significantly lower than those in the DB group. Therefore, these results suggest that, as an anti-diabetic agent, the AFA extract adjuvant to insulin could reduce and potentially prevent diabetic kidney disease.
Collapse
Affiliation(s)
- Ony Araújo Galdino
- grid.411233.60000 0000 9687 399XDepartment of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Av. General Gustavo Cordeiro de Farias, S/N, Faculty of Pharmacy, Petrópolis, Natal, RN CEP: 59012-570 Brazil
| | - Iago de Souza Gomes
- grid.411233.60000 0000 9687 399XDepartment of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Av. General Gustavo Cordeiro de Farias, S/N, Faculty of Pharmacy, Petrópolis, Natal, RN CEP: 59012-570 Brazil
| | - Renato Ferreira de Almeida Júnior
- grid.411233.60000 0000 9687 399XDepartment of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Av. General Gustavo Cordeiro de Farias, S/N, Faculty of Pharmacy, Petrópolis, Natal, RN CEP: 59012-570 Brazil
| | - Maria Imaculada Conceição Ferreira de Carvalho
- grid.411233.60000 0000 9687 399XDepartment of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Av. General Gustavo Cordeiro de Farias, S/N, Faculty of Pharmacy, Petrópolis, Natal, RN CEP: 59012-570 Brazil
| | - Bento João Abreu
- grid.411233.60000 0000 9687 399XDepartment of Morphology, Federal University of Rio Grande do Norte, Natal, RN Brazil
| | - Marcela Abbott Galvão Ururahy
- grid.411233.60000 0000 9687 399XDepartment of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Av. General Gustavo Cordeiro de Farias, S/N, Faculty of Pharmacy, Petrópolis, Natal, RN CEP: 59012-570 Brazil
| | - Barbara Cabral
- grid.411233.60000 0000 9687 399XDepartment of Pharmacy, Federal University of Rio Grande do Norte, Natal, RN Brazil
| | | | - Karla Simone Costa de Souza
- grid.411233.60000 0000 9687 399XDepartment of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Av. General Gustavo Cordeiro de Farias, S/N, Faculty of Pharmacy, Petrópolis, Natal, RN CEP: 59012-570 Brazil
| | - Adriana Augusto de Rezende
- grid.411233.60000 0000 9687 399XDepartment of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Av. General Gustavo Cordeiro de Farias, S/N, Faculty of Pharmacy, Petrópolis, Natal, RN CEP: 59012-570 Brazil
| |
Collapse
|
3
|
Dratwa M, Wysoczańska B, Łacina P, Kubik T, Bogunia-Kubik K. TERT-Regulation and Roles in Cancer Formation. Front Immunol 2020; 11:589929. [PMID: 33329574 PMCID: PMC7717964 DOI: 10.3389/fimmu.2020.589929] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is a catalytic subunit of telomerase. Telomerase complex plays a key role in cancer formation by telomere dependent or independent mechanisms. Telomere maintenance mechanisms include complex TERT changes such as gene amplifications, TERT structural variants, TERT promoter germline and somatic mutations, TERT epigenetic changes, and alternative lengthening of telomere. All of them are cancer specific at tissue histotype and at single cell level. TERT expression is regulated in tumors via multiple genetic and epigenetic alterations which affect telomerase activity. Telomerase activity via TERT expression has an impact on telomere length and can be a useful marker in diagnosis and prognosis of various cancers and a new therapy approach. In this review we want to highlight the main roles of TERT in different mechanisms of cancer development and regulation.
Collapse
Affiliation(s)
- Marta Dratwa
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Barbara Wysoczańska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Tomasz Kubik
- Department of Computer Engineering, Faculty of Electronics, Wrocław University of Science and Technology, Wroclaw, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
4
|
Goel H, Rahul E, Gupta AK, Meena JP, Chopra A, Ranjan A, Hussain S, Rath GK, Tanwar P. Molecular update on biology of Wilms Tumor 1 gene and its applications in acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2020; 10:151-160. [PMID: 33224559 PMCID: PMC7675129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/16/2020] [Indexed: 06/11/2023]
Abstract
Wilms tumor gene 1 (WT1) is an important gene which is involved in growth and development of many organs. It is identified as a tumor suppressor gene in nephroblastoma. However, its role as a tumor oncogene has been highlighted by many studies in haematological as well as non haematological malignant neoplasm. The expression of WT1 on leukemic blast cells sensitised us to explore its impact on neoplastic phenomenon. WT1 is has been found both mutated as well as over expressed in different subsets of acute myeloid leukemia (AML). WT1 is a gene has been used as a biomarker for diagnosis, monitoring of minimal residual disease (MRD) and detection of relapse for molecular remission in AML. It also has potential of being a predictive molecular predictive biomarker for the treatment of leukemic cases after allogeneic transplantation. The WT1 specific expression on blast cells and its interaction with cytotoxic T cell has also been explored for its potential usage WT1 based immunotherapy. Here, we are reviewing molecular updates of WT1 gene and discuss its potential clinical applications as a predictive molecular biomarker for diagnosis, as MRD detection and as immunotherapy in AML.
Collapse
Affiliation(s)
- Harsh Goel
- Laboratory Oncology Unit, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ekta Rahul
- Laboratory Oncology Unit, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Aditya Kumar Gupta
- Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Jagdish Prasad Meena
- Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Amar Ranjan
- Laboratory Oncology Unit, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Showket Hussain
- Division of Molecular Oncology, National Institute of Cancer Prevention & Research I-7Sector-39, NOIDA-201301, India
| | - GK Rath
- All India Institute of Medical ScincesNew Delhi, India
| | - Pranay Tanwar
- Laboratory Oncology Unit, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi 110029, India
| |
Collapse
|
5
|
Křivohlavá R, Grobárová V, Neuhöferová E, Fišerová A, Benson V. Interaction of colon cancer cells with glycoconjugates triggers complex changes in gene expression, glucose transporters and cell invasion. Mol Med Rep 2018; 17:5508-5517. [PMID: 29393416 DOI: 10.3892/mmr.2018.8490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/15/2017] [Indexed: 11/06/2022] Open
Abstract
Glycan metabolism balance is critical for cell prosperity, and macromolecule glycosylation is essential for cell communication, signaling and survival. Thus, glycotherapy may be a potential cancer treatment. The aim of the present study was to determine whether combined synthetic glycoconjugates (GCs) induce changes in gene expression that alter the survival of colon cancer cells. The current study evaluated the effect of the GCs N‑acetyl‑D‑glucosamine modified polyamidoamine dendrimer and calix[4]arene scaffold on cancer cell proliferation, apoptosis, invasion and sensitivity to immune cell‑mediated killing. Using reverse transcription‑quantitative polymerase chain reaction, the expression of genes involved in the aforementioned processes was measured. It was determined that GCs reduce the expression of the glucosaminyltransferases Mgat3 and Mgat5 responsible for surface glycosylation and employed components of the Wnt signaling pathway Wnt2B and Wnt9B. In addition, the calix[4]arene‑based GC reduced cell colony formation; this was accompanied by the downregulation of the metalloproteinase Mmp3. By contrast, the dendrimer‑based GC affected the expression of the glucose transporter components Sglt1 and Egfr1. Therefore, to the best of our knowledge, the present study is the first to reveal that N‑acetyl‑D‑glucosamine‑dendrimer/calix[4]arene GCs alter mRNA expression in a comprehensive way, resulting in the reduced malignant phenotype of the colon cancer cell line HT‑29.
Collapse
Affiliation(s)
- Romana Křivohlavá
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Valika Grobárová
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Eva Neuhöferová
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Anna Fišerová
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Veronika Benson
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| |
Collapse
|
6
|
RNA binding protein QKI contributes to WT1 mRNA and suppresses apoptosis in ST cells. Genes Genomics 2017. [DOI: 10.1007/s13258-017-0560-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
7
|
Blocking the binding of WT1 to bcl-2 promoter by G-quadruplex ligand SYUIQ-FM05. Biochem Biophys Rep 2016; 5:346-352. [PMID: 28955841 PMCID: PMC5600358 DOI: 10.1016/j.bbrep.2015.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 11/23/2022] Open
Abstract
At present, wt1, a Wilms’ tumor suppressor gene, is recognized as a critical regulator of tumorigenesis and a potential therapeutic target. WT1 shows the ability to regulate the transcription of bcl-2 by binding to a GC-rich region in the promoter, which can then fold into a special DNA secondary structure called the G-quadruplex. This function merits the exploration of the effect of a G-quadruplex ligand on the binding and subsequent regulation of WT1 on the bcl-2 promoter. In the present study, WT1 was found to bind to the double strand containing the G-quadruplex-forming sequence of the bcl-2 promoter. However, the G-quadruplex ligand SYUIQ-FM05 effectively blocked this binding by interacting with the GC-rich sequence. Our new findings are significant in the exploration of new strategies to block WT1's transcriptional regulation for cancer-cell treatment. WT1 bound to double-stranded but not G-quadruplex structure in bcl-2′s promoter. G-quadruplexes ligand SYUIQ-FM05 blocked the binding of WT1 to bcl-2. SYUIQ-FM05's regulation effects depends on its interaction with GC-rich sequence.
Collapse
|
8
|
The DRY box and C-terminal domain of the human cytomegalovirus US27 gene product play a role in promoting cell growth and survival. PLoS One 2014; 9:e113427. [PMID: 25409008 PMCID: PMC4237426 DOI: 10.1371/journal.pone.0113427] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/28/2014] [Indexed: 01/24/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a widespread pathogen that can lay dormant in healthy individuals and establish lifelong latent infection. This successful co-existence is facilitated by a number of viral gene products that manipulate host cellular functions and immune responses. Among these immunomodulatory genes are four G-protein coupled receptors (GPCRs) encoded by HCMV, designated US27, US28, UL33, and UL78. Studies have shown the US28 gene product to be a functional chemokine receptor that signals both constitutively and in a ligand-dependent manner, resulting in a wide range of cellular effects. In previous work, we have found that US27 expression results in at least two biological effects: enhanced CXCR4 signaling and increased in cellular proliferation in HEK293 cells. Here, we examined the involvement of two protein domains, the DRY box and the C-terminal intracellular domain (CTD) of US27, in mediating both cell proliferation and survival. While both domains were required for a proliferative effect, loss of either domain only moderately impacted cell survival, suggesting that US27 may interact with cell survival pathways through protein regions other than the DRY box and CTD. Quantitative RT-PCR arrays were used to profile changes in cellular gene expression in the HEK293-US27 cell line, and down-regulation of cell cycle regulators CDKN1A/p21/CIP1 (cyclin dependent kinase inhibitor 1A) and SESN (Sestrin2 or Hi95) was observed. These results indicate that increased cell proliferation due to US27 may be linked to suppression of negative growth regulators, and that these interactions require the DRY box and CTD.
Collapse
|
9
|
Massaoka MH, Matsuo AL, Figueiredo CR, Girola N, Faria CF, Azevedo RA, Travassos LR. A novel cell-penetrating peptide derived from WT1 enhances p53 activity, induces cell senescence and displays antimelanoma activity in xeno- and syngeneic systems. FEBS Open Bio 2014; 4:153-61. [PMID: 24490140 PMCID: PMC3907745 DOI: 10.1016/j.fob.2014.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/07/2014] [Accepted: 01/14/2014] [Indexed: 01/24/2023] Open
Abstract
The Wilms tumor protein 1 (WT1) transcription factor has been associated in malignant melanoma with cell survival and metastasis, thus emerging as a candidate for targeted therapy. A lysine-arginine rich peptide, WT1-pTj, derived from the ZF domain of WT1 was evaluated as an antitumor agent against A2058 human melanoma cells and B16F10-Nex2 syngeneic murine melanoma. Peptide WT1-pTj quickly penetrated human melanoma cells and induced senescence, recognized by increased SA-β-galactosidase activity, enhanced transcriptional activity of p53, and induction of the cell cycle inhibitors p21 and p27. Moreover, the peptide bound to p53 and competed with WT1 protein for binding to p53. WT1-pTj treatment led to sustained cell growth suppression, abrogation of clonogenicity and G2/M cell cycle arrest. Notably, in vivo studies showed that WT1-pTj inhibited both the metastases and subcutaneous growth of murine melanoma in syngeneic mice, and prolonged the survival of nude mice challenged with human melanoma cells. The 27-amino acid cell-penetrating WT1-derived peptide, depends on C(3) and H(16) for effective antimelanoma activity, inhibits proliferation of WT1-expressing human tumor cell lines, and may have an effective role in the treatment of WT1-expressing malignancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Luiz R. Travassos
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, SP 04023-062, Brazil
| |
Collapse
|
10
|
Heger M, van Golen RF, Broekgaarden M, Michel MC. The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol Rev 2013; 66:222-307. [PMID: 24368738 DOI: 10.1124/pr.110.004044] [Citation(s) in RCA: 363] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review addresses the oncopharmacological properties of curcumin at the molecular level. First, the interactions between curcumin and its molecular targets are addressed on the basis of curcumin's distinct chemical properties, which include H-bond donating and accepting capacity of the β-dicarbonyl moiety and the phenylic hydroxyl groups, H-bond accepting capacity of the methoxy ethers, multivalent metal and nonmetal cation binding properties, high partition coefficient, rotamerization around multiple C-C bonds, and the ability to act as a Michael acceptor. Next, the in vitro chemical stability of curcumin is elaborated in the context of its susceptibility to photochemical and chemical modification and degradation (e.g., alkaline hydrolysis). Specific modification and degradatory pathways are provided, which mainly entail radical-based intermediates, and the in vitro catabolites are identified. The implications of curcumin's (photo)chemical instability are addressed in light of pharmaceutical curcumin preparations, the use of curcumin analogues, and implementation of nanoparticulate drug delivery systems. Furthermore, the pharmacokinetics of curcumin and its most important degradation products are detailed in light of curcumin's poor bioavailability. Particular emphasis is placed on xenobiotic phase I and II metabolism as well as excretion of curcumin in the intestines (first pass), the liver (second pass), and other organs in addition to the pharmacokinetics of curcumin metabolites and their systemic clearance. Lastly, a summary is provided of the clinical pharmacodynamics of curcumin followed by a detailed account of curcumin's direct molecular targets, whereby the phenotypical/biological changes induced in cancer cells upon completion of the curcumin-triggered signaling cascade(s) are addressed in the framework of the hallmarks of cancer. The direct molecular targets include the ErbB family of receptors, protein kinase C, enzymes involved in prostaglandin synthesis, vitamin D receptor, and DNA.
Collapse
Affiliation(s)
- Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
11
|
Han Y, Cui J, Lu Y, Sue S, Arpaia E, Mak TW, Minden MD. FCHSD2 predicts response to chemotherapy in acute myeloid leukemia patients. Leuk Res 2012; 36:1339-46. [PMID: 22902056 DOI: 10.1016/j.leukres.2012.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 06/12/2012] [Accepted: 06/16/2012] [Indexed: 11/25/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by the growth and accumulation of cells blocked in their ability to differentiate, and blocks production of normal blood cells. The response to induction chemotherapy is heterogeneous, therefore biomarkers that predict for the outcome of such treatment are of potential value. FCHSD2 in a sensitized screen was identified as a potential chemo-protector (TW Mak, unpublished). In the present study, we found that FCHSD2 knockdown by shRNA could enhance chemosensitivity of U937 cells. This coincided with the increased expression of p21 and PUMA as well as the decreased expression of Bcl-2, c-myc and hTERT. In contrast, over-expression of FCHSD2 significantly increased cellular chemoresistance. To see if there was potential clinical relevance of FCHSD2 expression in leukemia we used qRT-PCR to assess FCHSD2 expression levels in peripheral blood or bone marrow blasts of 71 AML patients. There was an inverse correlation between the level of FCHSD2 with overall survival time (r=-0.7647, p<0.0001) and relapse free time (r=-0.8165, p<0.0001). By dividing patients into high and low expression groups using a FCHSD2 expression threshold value of 0.001, the median survival of the high expression group (72 days) was shorter than in the low expression group (2472 days). The average FCHSD2 expression level in 41 patients with complete remission was significantly lower than that in 30 non-responder patients (p<0.0001). Moreover, in 32 de novo AML patients receiving initial remission-induction chemotherapy, we confirmed that the patients with lower FCHSD2 expression prior to the treatment had an increased chance of attaining remission compared to patients with high level FCHSD2. In conclusion, our study, for the first time, demonstrates FCHSD2 as a predictor of outcome for AML patient. The determination of FCHSD2 expression at the time of diagnosis could help to identify the responses of AML patients to chemotherapy.
Collapse
Affiliation(s)
- Youqi Han
- Division of Stem Cell and Developmental Biology, Ontario Cancer Institute, Princess Margaret Hospital, Toronto, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
12
|
Real-time PCR quantification of major Wilms’ tumor gene 1 (WT1) isoforms in acute myeloid leukemia, their characteristic expression patterns and possible functional consequences. Leukemia 2012; 26:2086-95. [DOI: 10.1038/leu.2012.76] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Sakairi T, Abe Y, Kopp JB. TGF-beta1 reduces Wilms' tumor suppressor gene expression in podocytes. Nephrol Dial Transplant 2011; 26:2746-52. [PMID: 21378152 DOI: 10.1093/ndt/gfr061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Wilms' tumor suppressor gene (WT1) is essential for normal podocyte function, and transforming growth factor (TGF)-beta contributes to focal segmental glomerulosclerosis (FSGS). We aimed to address whether TGF-beta affects WT1 expression in podocytes. METHODS A human podocyte cell line treated with TGF-beta1 and kidneys in Alb/TGF-beta1-transgenic mice were analyzed for WT1 expression. RESULTS In cultured podocytes, TGF-beta1 reduced WT1 protein expression determined by western blotting beginning at 8 h and decreased WT1 messenger RNA (mRNA) expression measured by quantitative reverse transcription-polymerase chain reaction beginning at 3 h. Knockdown of Smad4 by small hairpin (sh) RNA partially rescued the TGF-beta1-induced reduction of both WT1 protein and mRNA expressions in the cultured podocytes. TGF-beta1 did not alter luciferase activity of the reporter construct for a human WT1 promoter but reduced that for a human WT1 5' enhancer construct, suggesting that TGF-beta1 may regulate WT1 expression by altering the 5' enhancer activity. In the transgenic mice, WT1 protein expression in podocytes was decreased at 1 and 3 weeks of age, while glomeruloclerosis developed after 3 weeks. CONCLUSION TGF-beta1 reduces WT1 expression in cultured human podocytes and podocytes in mice before overt glomerulosclerosis begins. The effects are at least partially Smad4 dependent. Our findings identify a novel pathway linking TGF-beta1 to podocyte injury and FSGS. The WT1 reduction may be a useful marker for early podocyte injury.
Collapse
Affiliation(s)
- Toru Sakairi
- Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
14
|
Benson V, Grobarova V, Richter J, Fiserova A. Glycosylation regulates NK cell-mediated effector function through PI3K pathway. Int Immunol 2010; 22:167-77. [DOI: 10.1093/intimm/dxp123] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|