1
|
Dissanayake UC, Roy A, Maghsoud Y, Polara S, Debnath T, Cisneros GA. Computational studies on the functional and structural impact of pathogenic mutations in enzymes. Protein Sci 2025; 34:e70081. [PMID: 40116283 PMCID: PMC11926659 DOI: 10.1002/pro.70081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Enzymes are critical biological catalysts involved in maintaining the intricate balance of metabolic processes within living organisms. Mutations in enzymes can result in disruptions to their functionality that may lead to a range of diseases. This review focuses on computational studies that investigate the effects of disease-associated mutations in various enzymes. Through molecular dynamics simulations, multiscale calculations, and machine learning approaches, computational studies provide detailed insights into how mutations impact enzyme structure, dynamics, and catalytic activity. This review emphasizes the increasing impact of computational simulations in understanding molecular mechanisms behind enzyme (dis)function by highlighting the application of key computational methodologies to selected enzyme examples, aiding in the prediction of mutation effects and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Upeksha C. Dissanayake
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Arkanil Roy
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Yazdan Maghsoud
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
- Present address:
Department of Biochemistry and Molecular PharmacologyBaylor College of MedicineHoustonTexasUSA
| | - Sarthi Polara
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Tanay Debnath
- Department of PhysicsThe University of Texas at DallasRichardsonTexasUSA
- Present address:
Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
| | - G. Andrés Cisneros
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
- Department of PhysicsThe University of Texas at DallasRichardsonTexasUSA
| |
Collapse
|
2
|
Liang F, Rai R, Sodeinde T, Chang S. TRF2-RAP1 represses RAD51-dependent homology-directed telomere repair by promoting BLM-mediated D-loop unwinding and inhibiting BLM-DNA2-dependent 5'-end resection. Nucleic Acids Res 2024; 52:9695-9709. [PMID: 39082275 PMCID: PMC11381343 DOI: 10.1093/nar/gkae642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 09/10/2024] Open
Abstract
Inappropriate homology-directed repair (HDR) of telomeres results in catastrophic telomere loss and aberrant chromosome fusions, leading to genome instability. We have previously shown that the TRF2-RAP1 heterodimer protects telomeres from engaging in aberrant telomere HDR. Cells lacking the basic domain of TRF2 and functional RAP1 display HDR-mediated telomere clustering, resulting in the formation of ultrabright telomeres (UTs) and massive chromosome fusions. Using purified proteins, we uncover three distinct molecular pathways that the TRF2-RAP1 heterodimer utilizes to protect telomeres from engaging in aberrant HDR. We show mechanistically that TRF2-RAP1 inhibits RAD51-initiated telomeric D-loop formation. Both the TRF2 basic domain and RAP1-binding to TRF2 are required to block RAD51-mediated homology search. TRF2 recruits the BLM helicase to telomeres through its TRFH domain to promote BLM-mediated unwinding of telomere D-loops. In addition, TRF2-RAP1 inhibits BLM-DNA2-mediated 5' telomere end resection, preventing the generation of 3' single-stranded telomere overhangs necessary for RAD51-dependent HDR. Importantly, cells expressing BLM mutants unable to interact with TRF2 accumulate telomere D-loops and UTs. Our findings uncover distinct molecular mechanisms coordinated by TRF2-RAP1 to protect telomeres from engaging in aberrant HDR.
Collapse
Affiliation(s)
- Fengshan Liang
- Departments of Laboratory Medicine, Yale University School of Medicine, 330 Cedar St., New Haven, CT 06520, USA
| | - Rekha Rai
- Departments of Laboratory Medicine, Yale University School of Medicine, 330 Cedar St., New Haven, CT 06520, USA
| | - Tori Sodeinde
- Departments of Laboratory Medicine, Yale University School of Medicine, 330 Cedar St., New Haven, CT 06520, USA
| | - Sandy Chang
- Departments of Laboratory Medicine, Yale University School of Medicine, 330 Cedar St., New Haven, CT 06520, USA
- Pathology, Yale University School of Medicine, 330 Cedar St., New Haven, CT 06520, USA
- Molecular Biophysics and Biochemistry, Yale University School of Medicine, 330 Cedar St., New Haven, CT 06520, USA
| |
Collapse
|
3
|
An D, Chen X, Li Z, Dai L, Huang J, Xiao M, Liu H, Xu J, Ruan Y. Genetic variation in the BLM gene and its expression in the ovaries is closely related to kidding number in goats. Theriogenology 2024; 218:254-266. [PMID: 38367334 DOI: 10.1016/j.theriogenology.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 02/19/2024]
Abstract
Bloom (BLM) helicase plays an important role in DNA replication and the maintenance of genome integrity. BLM protein deficiency, which plays a vital role in the sperm-egg union and germ-cell development during reproduction, can lead to severe DNA damage in goats. However, the effect of BLM protein deficiency on goat litter size has not been reported. Herein, we studied the association between the genetic variation in the BLM gene and the number of kids per litter in Guizhou white goats. We explored differences in the expression of the BLM protein in the follicles of single and multi-kid nanny goats. We also analyzed the effects of dysregulated BLM gene expression on the proliferation and apoptosis of ovarian granulosa cells and the expression of genes related to follicle development in goats. Five single nucleotide polymorphism (SNP) loci, including the non-synonymous mutations g.38179 A > G, g.40626 G > C and g.89621 T > G; the intron synonymous mutation g.56961 G > A and the exon synonymous mutation g.65796 C > T were found in the BLM gene. All SNPs loci were in Hardy-Weinberg equilibrium, and correlation analysis showed that the g.65796 C > T and g.89621 T > G loci polymorphism was strongly associated with litter size in the first three litters (P < 0.05). The diplogenotype Hap 2/2 (AAGGAACCTT) showed no significant difference in litter size between different births, indicating that the diploid genotype is stable in different litter sizes. Bioinformatics analysis showed that three non-synonymous mutation loci (p.T488A, p.A662S, and p.S1373A) could affect BLM protein stability, and mutations in p.T488A and p.S1373A led to changes in amino acid polarity and associated interactions. qPCR results showed that the expression level of the BLM gene in the uterus and ovaries of TT genotype nanny goats was significantly higher than that of GG genotype nanny goats. Indirect immunofluorescence assay (IF) showed that the BLM protein was significantly overexpressed in both the primordial and growing follicles of nanny goats with multiple kids (P < 0.01). Disrupting BLM gene expression in the ovarian granulosa cells down-regulated the expression of the Cyp19A1 gene. It also significantly inhibited the proliferation of follicles and induces early apoptosis of the granulosa cells. These findings confirm that polymorphism in the BLM gene is closely related to the littering traits of Guizhou white goats, and it affects the reproductive performance of nanny goats by regulating the development of the oocytes and granulosa cells. This work provides new evidence on the regulatory effect of the BLM gene on the litter size of nanny goats.
Collapse
Affiliation(s)
- Dongwei An
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Xiang Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Ziyang Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Lingang Dai
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Jiajin Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Meimei Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Huan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Jiali Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China
| | - Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, PR China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, 550025, PR China; College of Animal Science, Guizhou University, Guiyang, 550025, PR China.
| |
Collapse
|
4
|
Haggag HS, Aboukhatwa SM, Nafie MS, Paul A, Sharafeldin N, Oliver AW, El-Hamamsy MH. Design and synthesis of quinazolin-4-one derivatives as potential anticancer agents and investigation of their interaction with RecQ helicases. Bioorg Chem 2024; 144:107086. [PMID: 38219478 DOI: 10.1016/j.bioorg.2023.107086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2024]
Abstract
The upregulation of RecQ helicases has been associated with cancer cell survival and resistance to chemotherapy, making them appealing targets for therapeutic intervention. In this study, twenty-nine novel quinazolinone derivatives were designed and synthesized. The anti-proliferative activity of all compounds was evaluated against 60 cancer cell lines at the National Cancer Institute Developmental Therapeutic Program, with six compounds (11f, 11g, 11k, 11n, 11p, and 11q) being promoted to a five-dose screen. Compound 11g demonstrated high cytotoxic activity against all examined cell lines. The compounds were further assayed for Bloom syndrome (BLM) helicase inhibition, where 11g, 11q, and 11u showed moderate activity. These compounds were counter-screened against WRN and RECQ1 helicases, where 11g moderately inhibited both enzymes. An ATP competition assay confirmed that the compounds bound to the ATP site of RecQ helicases, and molecular docking simulations were used to study the binding mode within the active site of BLM, WRN, and RECQ1 helicases. Compound 11g induced apoptosis in both HCT-116 and MDA-MB-231 cell lines, but also caused an G2/M phase cell cycle arrest in HCT-116 cells. This data revealed the potential of 11g as a modulator of cell cycle dynamics and supports its interaction with RecQ helicases. In addition, compound 11g displayed non-significant toxicity against FCH normal colon cells at doses up to 100 µM, which confirming its high safety margin and selectivity on cancer cells. Overall, these findings suggest compound 11g as a potential pan RecQ helicase inhibitor with high anticancer potency and a favorable safety margin and selectivity.
Collapse
Affiliation(s)
- Hanan S Haggag
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Shaimaa M Aboukhatwa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago 60608, IL, USA
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, (P. O. Box 27272), Sharjah, United Arab Emirates; Chemistry Department (Biochemistry Program), Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Anju Paul
- Genome Damage and Stability Centre, Science Park Road, University of Sussex Falmer, Brighton BN1 9RQ, UK
| | - Nabaweya Sharafeldin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Antony W Oliver
- Genome Damage and Stability Centre, Science Park Road, University of Sussex Falmer, Brighton BN1 9RQ, UK
| | - Mervat H El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
5
|
Jiang H, Liu Y, Zhou R, Feng Y, Yan L. Circulating interleukins and risk of colorectal cancer: a Mendelian randomization study. Scand J Gastroenterol 2023; 58:1466-1473. [PMID: 37525405 DOI: 10.1080/00365521.2023.2240928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/11/2023] [Accepted: 07/20/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Recent studies have suggested a potential causal association between Interleukins (ILs) and Colorectal Cancer (CRC), and thus, it is important to examine the causal relationship between them using a Mendelian randomization (MR) approach. METHODS The instrumental variables were extracted for IL-1ra, IL-6, IL-6ra, IL-8, IL-16, IL-18, IL-27 from genome-wide association studies of European ancestry. Summary statistics of CRC were also retrieved. An inverse variance-weighted MR approach was implemented as the primary method to compute overall effects from multiple instruments. Additional MR approaches and sensitivity and heterogeneity pleiotropy analyses were also conducted respectively. RESULTS Our analysis suggested a causal effect between an increase of IL-8 and a reduced risk of CRC (odds ratio 0.65; 95% confidence interval, 0.43-0.98; p = 0.041) and did not provide evidence for causal effects of IL-1ra, IL-6, IL-6ra, IL-16, IL-18, IL-27. Sensitivity analyses suggested the robustness of MR results and that they were unlikely to be affected by unbalanced pleiotropy or significant heterogeneity. CONCLUSIONS This study investigated the role of ILs in the development of CRC and we found a causal effect between an increase of IL-8 and a reduced risk of CRC but not found evidence for causal effects of IL-1ra, IL-6, IL-6ra, IL-16, IL-18, IL-27. Sensitivity analyses suggested the robustness of MR results and that they were unlikely to be affected by unbalanced pleiotropy or significant heterogeneity.
Collapse
Affiliation(s)
- Haifeng Jiang
- Department of General Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongming Liu
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ru Zhou
- Department of General Surgery, RuiJin Hospital LuWan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Feng
- Department of General Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Yan
- Department of General Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Wojnicki K, Kaczmarczyk A, Wojtas B, Kaminska B. BLM helicase overexpressed in human gliomas contributes to diverse responses of human glioma cells to chemotherapy. Cell Death Discov 2023; 9:157. [PMID: 37169803 PMCID: PMC10175545 DOI: 10.1038/s41420-023-01451-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023] Open
Abstract
Most of anti-tumour therapies eliminate neoplastic cells by introducing DNA damage which ultimately triggers cell death. These effects are counteracted by activated DNA repair pathways to sustain tumour proliferation capacity. RECQL helicases family, including BLM, participate in DNA damage and repair, and prevent the replication stress. Glioblastoma (GBM) is a common, malignant brain tumour that inevitably recurs despite surgical resection, radiotherapy, and chemotherapy with temozolomide (TMZ). Expression and functions of the BLM helicase in GBM therapy resistance have not been elucidated. We analysed expression and localisation of BLM in human gliomas and several glioma cell lines using TCGA datasets, immunostaining and Western blotting. BLM depleted human glioma cells were generated with CRISPR/Cas9 system. Effects of chemotherapeutics on cell proliferation, DNA damage and apoptosis were determined with flow cytometry, immunofluorescence, Western blotting and RNA sequencing. We found upregulated BLM mRNA levels in malignant gliomas, increased cytosolic localisation and poor survival of GBM patients with high BLM expression. BLM deficiency in LN18 and LN229 glioma cells resulted in profound transcriptomic alterations, reduced cell proliferation, and altered cell responses to chemotherapeutics. BLM-deficient glioma cells were resistant to the TMZ and PARP inhibitor treatment and underwent polyploidy or senescence depending on the TP53 activity. Our findings of high BLM expression in GBMs and its roles in responses to chemotherapeutics provide a rationale for targeting BLM helicase in brain tumours. BLM deficiency affects responses of glioma cells to chemotherapeutics targeting PARP1 dependent pathways.
Collapse
Affiliation(s)
- Kamil Wojnicki
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Agnieszka Kaczmarczyk
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
7
|
Ovejero S, Viziteu E, Dutrieux L, Devin J, Lin YL, Alaterre E, Jourdan M, Basbous J, Requirand G, Robert N, de Boussac H, Seckinger A, Hose D, Vincent L, Herbaux C, Constantinou A, Pasero P, Moreaux J. The BLM helicase is a new therapeutic target in multiple myeloma involved in replication stress survival and drug resistance. Front Immunol 2022; 13:983181. [PMID: 36569948 PMCID: PMC9780552 DOI: 10.3389/fimmu.2022.983181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is a hematologic cancer characterized by accumulation of malignant plasma cells in the bone marrow. To date, no definitive cure exists for MM and resistance to current treatments is one of the major challenges of this disease. The DNA helicase BLM, whose depletion or mutation causes the cancer-prone Bloom's syndrome (BS), is a central factor of DNA damage repair by homologous recombination (HR) and genomic stability maintenance. Using independent cohorts of MM patients, we identified that high expression of BLM is associated with a poor outcome with a significant enrichment in replication stress signature. We provide evidence that chemical inhibition of BLM by the small molecule ML216 in HMCLs (human myeloma cell lines) leads to cell cycle arrest and increases apoptosis, likely by accumulation of DNA damage. BLM inhibition synergizes with the alkylating agent melphalan to efficiently inhibit growth and promote cell death in HMCLs. Moreover, ML216 treatment re-sensitizes melphalan-resistant cell lines to this conventional therapeutic agent. Altogether, these data suggest that inhibition of BLM in combination with DNA damaging agents could be of therapeutic interest in the treatment of MM, especially in those patients with high BLM expression and/or resistance to melphalan.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Elena Viziteu
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Laure Dutrieux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Julie Devin
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Yea-Lih Lin
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Elina Alaterre
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Michel Jourdan
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Jihane Basbous
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Guilhem Requirand
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | | | | | - Dirk Hose
- Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Laure Vincent
- Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Charles Herbaux
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France,Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - Philippe Pasero
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Jérôme Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France,Department of Clinical Hematology, CHU Montpellier, Montpellier, France,*Correspondence: Jérôme Moreaux,
| |
Collapse
|
8
|
Ahmed F, Khan AA, Ansari HR, Haque A. A Systems Biology and LASSO-Based Approach to Decipher the Transcriptome-Interactome Signature for Predicting Non-Small Cell Lung Cancer. BIOLOGY 2022; 11:biology11121752. [PMID: 36552262 PMCID: PMC9774707 DOI: 10.3390/biology11121752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
The lack of precise molecular signatures limits the early diagnosis of non-small cell lung cancer (NSCLC). The present study used gene expression data and interaction networks to develop a highly accurate model with the least absolute shrinkage and selection operator (LASSO) for predicting NSCLC. The differentially expressed genes (DEGs) were identified in NSCLC compared with normal tissues using TCGA and GTEx data. A biological network was constructed using DEGs, and the top 20 upregulated and 20 downregulated hub genes were identified. These hub genes were used to identify signature genes with penalized logistic regression using the LASSO to predict NSCLC. Our model’s development involved the following steps: (i) the dataset was divided into 80% for training (TR) and 20% for testing (TD1); (ii) a LASSO logistic regression analysis was performed on the TR with 10-fold cross-validation and identified a combination of 17 genes as NSCLC predictors, which were used further for development of the LASSO model. The model’s performance was assessed on the TD1 dataset and achieved an accuracy and an area under the curve of the receiver operating characteristics (AUC-ROC) of 0.986 and 0.998, respectively. Furthermore, the performance of the LASSO model was evaluated using three independent NSCLC test datasets (GSE18842, GSE27262, GSE19804) and achieved high accuracy, with an AUC-ROC of >0.99, >0.99, and 0.95, respectively. Based on this study, a web application called NSCLCpred was developed to predict NSCLC.
Collapse
Affiliation(s)
- Firoz Ahmed
- Department of Biochemistry, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Saudi Arabia
- Correspondence:
| | - Abdul Arif Khan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hifzur Rahman Ansari
- King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 9515, Jeddah 21423, Saudi Arabia
| | - Absarul Haque
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| |
Collapse
|
9
|
Discovery of a Novel Bloom's Syndrome Protein (BLM) Inhibitor Suppressing Growth and Metastasis of Prostate Cancer. Int J Mol Sci 2022; 23:ijms232314798. [PMID: 36499126 PMCID: PMC9736344 DOI: 10.3390/ijms232314798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Prostate cancer (PCa) is a common cancer and a major cause of cancer-related death worldwide in men, necessitating novel targets for cancer therapy. High expression of Bloom's syndrome protein (BLM) helicase is associated with the occurrence and development of PCa. Therefore, the identification and development of new BLM inhibitors may be a new direction for the treatment of PCa. Here, we identified a novel inhibitor by molecular docking and put it to systematic evaluation via various experiments, AO/854, which acted as a competitive inhibitor that blocked the BLM-DNA interaction. Cellular evaluation indicated that AO/854-suppressed tumor growth and metastasis in PC3 cells by enhancing DNA damage, phosphorylating Chk1/Chk2, and altering the p53 signaling pathway. Collectively, the study highlights the potential of BLM as a therapeutic target in PCa and reveals a distinct mechanism by which AO/854 competitively inhibits the function of BLM.
Collapse
|
10
|
Identification and Characterization of Novel Mutations in Chronic Kidney Disease (CKD) and Autosomal Dominant Polycystic Kidney Disease (ADPKD) in Saudi Subjects by Whole-Exome Sequencing. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58111657. [PMID: 36422197 PMCID: PMC9692281 DOI: 10.3390/medicina58111657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Background: Autosomal dominant polycystic kidney disease (ADPKD) is a condition usually caused by a single gene mutation and manifested by both renal and extrarenal features, eventually leading to end-stage renal disease (ESRD) by the median age of 60 years worldwide. Approximately 89% of ADPKD patients had either PKD1 or PKD2 gene mutations. The majority (85%) of the mutations are in the PKD1 gene, especially in the context of family history. Objectives: This study investigated the genetic basis and the undiscovered genes that are involved in ADPKD development among the Saudi population. Materials and Methods: In this study, 11 patients with chronic kidney disease were enrolled. The diagnosis of ADPKD was based on history and diagnostic images: CT images include enlargement of renal outlines, renal echogenicity, and presence of multiple renal cysts with dilated collecting ducts, loss of corticomedullary differentiation, and changes in GFR and serum creatinine levels. Next-generation whole-exome sequencing was conducted using the Ion Torrent PGM platform. Results: Of the 11 Saudi patients diagnosed with chronic kidney disease (CKD) and ADPKD, the most common heterozygote nonsynonymous variant in the PKD1 gene was exon15: (c.4264G > A). Two missense mutations were identified with a PKD1 (c.1758A > C and c.9774T > G), and one patient had a PKD2 mutation (c.1445T > G). Three detected variants were novel, identified at PKD1 (c.1758A > C), PKD2L2 (c.1364A > T), and TSC2 (deletion of a’a at the 3’UTR, R1680C) genes. Other variants in PKD1L1 (c.3813_381 4delinsTG) and PKD1L2 (c.404C > T) were also detected. The median age of end-stage renal disease for ADPK patients in Saudi Arabia was 30 years. Conclusion: This study reported a common variant in the PKD1 gene in Saudi patients with typical ADPKD. We also reported (to our knowledge) for the first time two novel missense variants in PKD1 and PKD2L2 genes and one indel mutation at the 3’UTR of the TSC2 gene. This study establishes that the reported mutations in the affected genes resulted in ADPKD development in the Saudi population by a median age of 30. Nevertheless, future protein−protein interaction studies to investigate the influence of these mutations on PKD1 and PKD2 functions are required. Furthermore, large-scale population-based studies to verify these findings are recommended.
Collapse
|
11
|
Alsagaby SA, Iqbal D, Ahmad I, Patel H, Mir SA, Madkhali YA, Oyouni AAA, Hawsawi YM, Alhumaydhi FA, Alshehri B, Alturaiki W, Alanazi B, Mir MA, Al Abdulmonem W. In silico investigations identified Butyl Xanalterate to competently target CK2α (CSNK2A1) for therapy of chronic lymphocytic leukemia. Sci Rep 2022; 12:17648. [PMID: 36271116 PMCID: PMC9587039 DOI: 10.1038/s41598-022-21546-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is an incurable malignancy of B-cells. In this study, bioinformatics analyses were conducted to identify possible pathogenic roles of CK2α, which is a protein encoded by CSNK2A1, in the progression and aggressiveness of CLL. Furthermore, various computational tools were used to search for a competent inhibitor of CK2α from fungal metabolites that could be proposed for CLL therapy. In CLL patients, high-expression of CSNK2A1 was associated with early need for therapy (n = 130, p < 0.0001) and short overall survival (OS; n = 107, p = 0.005). Consistently, bioinformatics analyses showed CSNK2A1 to associate with/play roles in CLL proliferation and survival-dependent pathways. Furthermore, PPI network analysis identified interaction partners of CK2α (PPI enrichment p value = 1 × 10-16) that associated with early need for therapy (n = 130, p < 0.003) and have been known to heavily impact on the progression of CLL. These findings constructed a rational for targeting CK2α for CLL therapy. Consequently, computational analyses reported 35 fungal metabolites out of 5820 (filtered from 19,967 metabolites) to have lower binding energy (ΔG: - 10.9 to - 11.7 kcal/mol) and better binding affinity (Kd: 9.77 × 107 M-1 to 3.77 × 108 M-1) compared with the native ligand (ΔG: - 10.8, Kd: 8.3 × 107 M--1). Furthermore, molecular dynamics simulation study established that Butyl Xanalterate-CK2α complex continuously remained stable throughout the simulation time (100 ns). Moreover, Butyl Xanalterate interacted with most of the catalytic residues, where complex was stabilized by more than 65% hydrogen bond interactions, and a significant hydrophobic interaction with residue Phe113. Here, high-expression of CSNK2A1 was implicated in the progression and poor prognosis of CLL, making it a potential therapeutic target in the disease. Butyl Xanalterate showed stable and strong interactions with CK2α, thus we propose it as a competitive inhibitor of CK2α for CLL therapy.
Collapse
Affiliation(s)
- Suliman A. Alsagaby
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Danish Iqbal
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Iqrar Ahmad
- grid.412233.50000 0001 0641 8393Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Harun Patel
- grid.412233.50000 0001 0641 8393Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Shabir Ahmad Mir
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Yahya Awaji Madkhali
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Atif Abdulwahab A. Oyouni
- grid.440760.10000 0004 0419 5685Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia ,grid.440760.10000 0004 0419 5685Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Yousef M. Hawsawi
- grid.415310.20000 0001 2191 4301Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 40047, Jeddah, 21499 Kingdom of Saudi Arabia ,grid.411335.10000 0004 1758 7207College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh, 11533 Kingdom of Saudi Arabia
| | - Fahad A. Alhumaydhi
- grid.412602.30000 0000 9421 8094Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Bader Alshehri
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Wael Alturaiki
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Bader Alanazi
- grid.415277.20000 0004 0593 1832Biomedical Research Administration, Research Center, King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia ,Prince Mohammed bin Abdulaziz Medical City, AlJouf, Kingdom of Saudi Arabia
| | - Manzoor Ahmad Mir
- grid.412997.00000 0001 2294 5433Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Waleed Al Abdulmonem
- grid.412602.30000 0000 9421 8094Department of Pathology, College of Medicine, Qassim University, Qassim, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Lan C, Yamashita YI, Hayashi H, Nakagawa S, Imai K, Mima K, Kaida T, Matsumoto T, Maruno M, Liu Z, Wu X, Wei F, Baba H. High Expression of Bloom Syndrome Helicase is a Key Factor for Poor Prognosis and Advanced Malignancy in Patients with Pancreatic Cancer: A Retrospective Study. Ann Surg Oncol 2022; 29:3551-3564. [PMID: 35419757 DOI: 10.1245/s10434-022-11500-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND Bloom syndrome helicase (BLM) is overexpressed in multiple types of cancers and its overexpression may induce genomic instability. This study aimed to determine the function of BLM expression in pancreatic cancer. METHODS BLM messenger RNA (mRNA) expression was analyzed using public datasets to determine its relationship with pancreatic cancer prognosis. Overall, 182 patients with pancreatic cancer who underwent radical resection at our institution were enrolled. BLM expression was evaluated by immunohistochemistry (IHC). We explored the effect of BLM on the proliferation, invasion, migration, and chemoresistance of pancreatic cancer cells via small-interfering RNAs and performed pathway analysis using gene set enrichment analysis. RESULTS BLM mRNA expression was higher in tumor tissue than in normal tissue and had a prognostic effect on overall survival (OS) and recurrence-free survival. The same results were validated by IHC. Multivariate analysis showed that high BLM expression was an independent poor prognostic factor for OS (hazard ratio [HR] 1.678, p = 0.029). In subgroup analysis, the effect of high BLM expression was more significant on OS in patients with younger age (HR 2.27, p = 0.006), male sex (HR 2.39, p = 0.002), high cancer antigen 19-9 level (HR 2.44, p = 0.001), advanced tumor stage (HR 2.25, p = 0.001), lymph node metastasis (HR 2.51, p = 0.001), nerve invasion (HR 2.07, p = 0.002), and adjuvant chemotherapy (HR 2.66, p < 0.001). In vitro, BLM suppression resulted in reduced tumor proliferation, invasion, migration, and chemoresistance. Mechanistically, BLM expression may be associated with E2F1 and E2F2. CONCLUSION BLM expression is a prognostic factor for patients with pancreatic cancer, especially in those with advanced malignancies and receiving chemotherapy.
Collapse
Affiliation(s)
- Chuan Lan
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Hepatobiliary Surgery and Center of Severe Acute Pancreatitis, The Affiliated Hospital of North Sichuan Medical College, Nanchong, People's Republic of China
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takayoshi Kaida
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Matsumoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masataka Maruno
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Zhao Liu
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Xiyu Wu
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Feng Wei
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
13
|
Ajucarmelprecilla A, Pandi J, Dhandapani R, Ramanathan S, Chinnappan J, Paramasivam R, Thangavelu S, Mohammed Ghilan AK, Aljohani SAS, Oyouni AAA, Farasani A, Altayar MA, Althagafi HAE, Alzahrani OR, Durairaj K, Shrestha A. In Silico Identification of Hub Genes as Observing Biomarkers for Gastric Cancer Metastasis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6316158. [PMID: 35535159 PMCID: PMC9078768 DOI: 10.1155/2022/6316158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/03/2022] [Indexed: 12/02/2022]
Abstract
Perception of hub genes engaged in metastatic gastric cancer (mGC) promotes novel ways to diagnose and treat the illness. The goal of this investigation is to recognize the hub genes and reveal its molecular mechanism. In order to explore the potential facts for gastric cancer, the expression profiles of two different datasets were used (GSE161533 and GSE54129). The genes were confirmed to be part of the PPI network for gastric cancer pathogenesis and prognosis. In Cytoscape, the CytoHubba module was used to discover the hub genes. Responsible hub genes were identified. Data from Kaplan-Meier plotter confirmed the predictive value of these distinct genes in various stages of gastric malignancy. Upregulated and downregulated genes were identified to utilize for further analysis. Positive regulation by a host of viral process, positive regulation of granulocyte differentiation, negative regulation of histone H3-K9 methylation were found in DEGs analysis. In addition, five KEGG pathways were identified as an essential enhancer that include nucleotide excision repair; base excision repair; DNA replication; homologous recombination; and complement and coagulation cascades. POLE, BUB1B, POLD4, C3, BLM, CCT7, PRPF31, APEX1, PSMA7, and CDC45 were chosen as hub genes after combining the PPI results. Our study recommends that BUB1B, CCT7, APEX1, PSMA7, and CDC45 might be potential biomarkers for gastric cancer. These biomarkers are upregulated genes. Therefore, suppression of these genes will increase the survival rate in gastric cancer patients.
Collapse
Affiliation(s)
| | - Jhansi Pandi
- Medical Microbiology Unit, Department of Microbiology, Alagappa University, Karaikudi, Tamil Nadu, India
- Chimertech Private Limited, Chennai, India
| | | | - Saikishore Ramanathan
- Medical Microbiology Unit, Department of Microbiology, Alagappa University, Karaikudi, Tamil Nadu, India
| | | | | | - Sathiamoorthi Thangavelu
- Medical Microbiology Unit, Department of Microbiology, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Abdul-Kareem Mohammed Ghilan
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Saad Ali S. Aljohani
- Department of Basic Medical Sciences, Faculty of Medicine, Alrayan Colleges, Almadinah Almunawarah, Saudi Arabia
| | - Atif Abdulwahab A. Oyouni
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdullah Farasani
- Biomedical Research Unit, Medical Research Center, Jazan University, Jazan 45142, Saudi Arabia
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Malik A. Altayar
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | | | - Othman R. Alzahrani
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Kaliannan Durairaj
- Zoonosis Research Center, School of Medicine, Wonkwang University, Iksan, Republic of Korea
| | - Anupama Shrestha
- Research Institute of Agriculture and Applied Science, Tokha Kathmandu 2356, Nepal
- Department of Plant Protection, Himalayan College of Agricultural Sciences and Technology, Kalanki, 44600 Kathmandu, Nepal
| |
Collapse
|
14
|
Maity J, Horibata S, Zurcher G, Lee JM. Targeting of RecQ Helicases as a Novel Therapeutic Strategy for Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14051219. [PMID: 35267530 PMCID: PMC8909030 DOI: 10.3390/cancers14051219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
RecQ helicases are essential for DNA replication, recombination, DNA damage repair, and other nucleic acid metabolic pathways required for normal cell growth, survival, and genome stability. More recently, RecQ helicases have been shown to be important for replication fork stabilization, one of the major mechanisms of PARP inhibitor resistance. Cancer cells often have upregulated helicases and depend on these enzymes to repair rapid growth-promoted DNA lesions. Several studies are now evaluating the use of RecQ helicases as potential biomarkers of breast and gynecologic cancers. Furthermore, RecQ helicases have attracted interest as possible targets for cancer treatment. In this review, we discuss the characteristics of RecQ helicases and their interacting partners that may be utilized for effective treatment strategies (as cancers depend on helicases for survival). We also discuss how targeting helicase in combination with DNA repair inhibitors (i.e., PARP and ATR inhibitors) can be used as novel approaches for cancer treatment to increase sensitivity to current treatment to prevent rise of treatment resistance.
Collapse
Affiliation(s)
- Jyotirindra Maity
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.M.); (G.Z.)
| | - Sachi Horibata
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Correspondence: (S.H.); (J.M.L.)
| | - Grant Zurcher
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.M.); (G.Z.)
| | - Jung-Min Lee
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.M.); (G.Z.)
- Correspondence: (S.H.); (J.M.L.)
| |
Collapse
|
15
|
Alrefaei AF, Hawsawi YM, Almaleki D, Alafif T, Alzahrani FA, Bakhrebah MA. Genetic data sharing and artificial intelligence in the era of personalized medicine based on a cross-sectional analysis of the Saudi human genome program. Sci Rep 2022; 12:1405. [PMID: 35082362 PMCID: PMC8791994 DOI: 10.1038/s41598-022-05296-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/07/2022] [Indexed: 12/21/2022] Open
Abstract
The success of the Saudi Human Genome Program (SHGP), one of the top ten genomic programs worldwide, is highly dependent on the Saudi population embracing the concept of participating in genetic testing. However, genetic data sharing and artificial intelligence (AI) in genomics are critical public issues in medical care and scientific research. The present study was aimed to examine the awareness, knowledge, and attitude of the Saudi society towards the SHGP, the sharing and privacy of genetic data resulting from the SHGP, and the role of AI in genetic data analysis and regulations. Results of a questionnaire survey with 804 respondents revealed moderate awareness and attitude towards the SHGP and minimal knowledge regarding its benefits and applications. Respondents demonstrated a low level of knowledge regarding the privacy of genetic data. A generally positive attitude was found towards the outcomes of the SHGP and genetic data sharing for medical and scientific research. The highest level of knowledge was detected regarding AI use in genetic data analysis and privacy regulation. We recommend that the SHGP’s regulators launch awareness campaigns and educational programs to increase and improve public awareness and knowledge regarding the SHGP’s benefits and applications. Furthermore, we propose a strategy for genetic data sharing which will facilitate genetic data sharing between institutions and advance Personalized Medicine in genetic diseases’ diagnosis and treatment.
Collapse
Affiliation(s)
- Abdulmajeed F Alrefaei
- Department of Biology, Genetic and Molecular Biology Central Lab, Jamoum University College, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
| | - Yousef M Hawsawi
- Research Centre, King Faisal Specialist Hospital and Research Centre, P.O. Box 40047, Jeddah, 21499, Saudi Arabia.,MBC: J04/ College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Deyab Almaleki
- Department of Evaluation, Measurement, and Research, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Tarik Alafif
- Computer Science Department, Jamoum University College, Umm Al-Qura University, Jamoum, 25375, Saudi Arabia
| | - Faisal A Alzahrani
- Department of Biochemistry, Faculty of Science, Embryonic Stem Cells Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Muhammed A Bakhrebah
- King Abdulaziz City for Science and Technology (KACST), Life Science and Environment Research Institute, P.O. Box 6086, Riyadh, 11442, Saudi Arabia
| |
Collapse
|
16
|
OUP accepted manuscript. Hum Mol Genet 2022; 31:2236-2261. [DOI: 10.1093/hmg/ddac029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/12/2022] Open
|
17
|
Al-Amer OM, Oyouni AAA, Alshehri MA, Alasmari A, Alzahrani OR, Aljohani SAS, Alasmael N, Theyab A, Algahtani M, Al Sadoun H, Alsharif KF, Hamad A, Abdali WA, Hawasawi YM. Association of SNPs within TMPRSS6 and BMP2 genes with iron deficiency status in Saudi Arabia. PLoS One 2021; 16:e0257895. [PMID: 34780475 PMCID: PMC8592490 DOI: 10.1371/journal.pone.0257895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 09/13/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Globally, iron-deficiency anemia (IDA) remains a major health obstacle. This health condition has been identified in 47% of pre-school students (aged 0 to 5 years), 42% of pregnant females, and 30% of non-pregnant females (aged 15 to 50 years) worldwide according to the WHO. Environmental and genetic factors play a crucial role in the development of IDA; genetic testing has revealed the association of a number of polymorphisms with iron status and serum ferritin. AIM The current study aims to reveal the association of TMPRSS6 rs141312 and BMP2 rs235756 with the iron status of females in Saudi Arabia. METHODS A cohort of 108 female university students aged 18-25 years was randomly selected to participate: 50 healthy and 58 classified as iron deficient. A 3-5 mL sample of blood was collected from each one and analyzed based on hematological and biochemical iron status followed by genotyping by PCR. RESULTS The genotype distribution of TMPRSS6 rs141312 was 8% (TT), 88% (TC) and 4% (CC) in the healthy group compared with 3.45% (TT), 89.66% (TC) and 6.89% (CC) in the iron-deficient group (P = 0.492), an insignificant difference in the allelic distribution. The genotype distribution of BMP2 rs235756 was 8% (TT), 90% (TC) and 2% (CC) in the healthy group compared with 3.45% (TT), 82.76% (TC) and 13.79% (CC) in iron-deficient group (P = 0.050) and was significantly associated with decreased ferritin status (P = 0.050). In addition, TMPRSS6 rs141312 is significantly (P<0.001) associated with dominant genotypes (TC+CC) and increased risk of IDA while BMP2 rs235756 is significantly (P<0.026) associated with recessive homozygote CC genotypes and increased risk of IDA. CONCLUSION Our finding potentially helps in the early prediction of iron deficiency in females through the genetic testing.
Collapse
Affiliation(s)
- Osama M. Al-Amer
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Atif Abdulwahab A. Oyouni
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Mohammed Ali Alshehri
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Abdulrahman Alasmari
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Othman R. Alzahrani
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Saad Ali S. Aljohani
- Department of Basic Medical Sciences, Faculty of Medicine, Alrayan Colleges, Almadinah Almunawarah, Kingdom of Saudi Arabia
| | - Noura Alasmael
- King Abdullah University for Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Abdulrahman Theyab
- Department of Laboratory Medicine, Security Forces Hospital, Mecca, Kingdom of Saudi Arabia
| | - Mohammad Algahtani
- Department of Laboratory Medicine, Security Forces Hospital, Mecca, Kingdom of Saudi Arabia
| | - Hadeel Al Sadoun
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Abdullah Hamad
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Wed A. Abdali
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia
| | - Yousef MohammedRabaa Hawasawi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia
- College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
18
|
Alzahrani FA, Hawsawi YM, Altayeb HN, Alsiwiehri NO, Alzahrani OR, Alatwi HE, Al‐Amer OM, Alomar S, Mansour L. In silico modeling of the interaction between TEX19 and LIRE1, and analysis of TEX19 gene missense SNPs. Mol Genet Genomic Med 2021; 9:e1707. [PMID: 34036740 PMCID: PMC8372073 DOI: 10.1002/mgg3.1707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/19/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Testis expressed 19 (TEX19) is a specific human stem cell gene identified as cancer-testis antigen (CTA), which emerged as a potential therapeutic drug target. TEX19.1, a mouse paralog of human TEX19, can interact with LINE-1 retrotransposable element ORF1 protein (LIRE1) and subsequently restrict mobilization of LINE-1 elements in the genome. AIM This study aimed to predict the interaction of TEX19 with LIRE1 and analyze TEX19 missense polymorphisms. TEX19 model was generated using I-TASSER and the interaction between TEX19 and LIRE1 was studied using the HADDOCK software. METHODS The stability of the docking formed complex was studied through the molecular dynamic simulation using GROMACS. Missense SNPs (n=102) of TEX19 were screened for their potential effects on protein structure and function using different software. RESULTS Outcomes of this study revealed amino acids that potentially stabilize the predicted interaction interface between TEX19 and LIRE1. Of these SNPs, 37 were predicted to play a probably damaging role for the protein, three of them (F35S, P61R, and E55L) located at the binding site of LIRE1 and could disturb this binding affinity. CONCLUSION This information can be verified by further in vitro and in vivo experimentations and could be exploited for potential therapeutic targets.
Collapse
Affiliation(s)
- Faisal A. Alzahrani
- Department of BiochemistryFaculty of ScienceEmbryonic Stem Cell UnitKing Fahad Center for Medical ResearchKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Yousef MohammedRabaa Hawsawi
- Research Center at King Faisal Specialist Hospital and Research CenterJeddahSaudi Arabia
- College of MedicineAl‐Faisal UniversityRiyadhSaudi Arabia
| | - Hisham N. Altayeb
- Department of BiochemistryFaculty of ScienceEmbryonic Stem Cell UnitKing Fahad Center for Medical ResearchKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Naif O. Alsiwiehri
- Department of Clinical Laboratory ScienceFaculty of Applied Medical ScienceTaif UniversityTaifSaudi Arabia
| | - Othman R. Alzahrani
- Department of BiologyFaculty of SciencesUniversity of TabukTabukSaudi Arabia
- Genome and Biotechnology UnitFaculty of ScienceUniversity of TabukTabukSaudi Arabia
| | - Hanan E. Alatwi
- Department of BiologyFaculty of SciencesUniversity of TabukTabukSaudi Arabia
- Genome and Biotechnology UnitFaculty of ScienceUniversity of TabukTabukSaudi Arabia
| | - Osama M. Al‐Amer
- Genome and Biotechnology UnitFaculty of ScienceUniversity of TabukTabukSaudi Arabia
- Department of Medical Laboratory TechnologyFaculty of Applied Medical SciencesUniversity of TaboukTabukSaudi Arabia
| | - Suliman Alomar
- Doping Research ChairDepartment of ZoologyCollege of ScienceKing Saud UniversityRiyadhSaudi Arabia
| | - Lamjed Mansour
- Doping Research ChairDepartment of ZoologyCollege of ScienceKing Saud UniversityRiyadhSaudi Arabia
- Department of ZoologyCollege of ScienceKing Saud UniversityRiyadhSaudi Arabia
| |
Collapse
|
19
|
Barnawi I, Hawsawi Y, Dash P, Oyouni AAA, Mustafa SK, Hussien NA, Al-Amer O, Alomar S, Mansour L. Nitric Oxide Synthase Potentiates the Resistance of Cancer Cell Lines to Anticancer Chemotherapeutics. Anticancer Agents Med Chem 2021; 22:1397-1406. [PMID: 34165414 DOI: 10.2174/1871520621666210623094526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Despite the advancement in the fields of medical science and molecular biology, cancer is still the leading cause of death worldwide. Chemotherapy is a choice for treatment; however, the acquisition of chemo-resistance is a major impediment to cancer management. Many mechanisms have been postulated regarding the acquisition of chemo-resistance in breast cancer the impact on cellular signaling and the induction of apoptosis in tumour cells. The mechanism of the apoptotic mutation of p53 and bcl-2 proteins is commonly associated with increased resistance to apoptosis and, therein, to chemotherapy. OBJECTIVES The current study was aimed to investigate A172 and MDA-MB-231 cancer cells' sensitivity against chemotherapeutic drugs, including cisplatin, doxorubicin, and paclitaxel with different doses. Moreover, it estimates the resistance of cancer cells by evaluating nitric oxide synthase (NOS) expression and evaluate its correlation with the expression profile proteins of the apoptosis regulating Bcl-2 family. METHODS Dose-dependent sensitivity to cisplatin, doxorubicin, or paclitaxel was evaluated on spheroid cultured A172 and MDA-MB-231 cells lines as measured by time-lapse microscopy over a 72h period. Expressions of two nitric oxides (NO) synthases isoforms (iNOS, eNOS), anti-apoptotic (Bcl-2, phospho-Bcl-2, Mcl-1, and Bcl-xL), and pro-apoptotic (BID, Bim, Bok, Bad, Puma, and Bax) were evaluated by Western blot. The effect of NO modulation on anti- and pro-apoptotic molecule expression was also studied using Western blot. RESULT A172 cells show more resistance to chemotherapy drugs than MDA-MB-231 cancer cells. Therefore, they need higher doses for apoptosis. Resistance of gliomas might be returned to the higher significant expression of endothelial eNOS expression. It was clear that there is not a significant effect of NO modulation on the expression of pro-and anti-apoptotic proteins on both cell lines. CONCLUSION The present work provides a putative mechanism for the acquisition of drug resistance in breast cancer and glioma, which might be significant for clinical outcomes.
Collapse
Affiliation(s)
- Ibrahim Barnawi
- Department of Biology, Faculty of Sciences, University of Taiba, Madina, Saudi Arabia
| | - Yousef Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah 21499, P.O. Box 40047, Saudi Arabia
| | - Philip Dash
- University of Reading Faculty of Life Sciences, school of science, Reading, Reading, United Kingdom
| | | | - Syed Khalid Mustafa
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Nahed A Hussien
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Osama Al-Amer
- Department of Medical Laboratory Technology, Faculty of Applied Medical Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Suliman Alomar
- Doping Research Chair, Department of Zoology, College of Science, King Saud University PO. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Lamjed Mansour
- Doping Research Chair, Department of Zoology, College of Science, King Saud University PO. Box: 2455, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
20
|
Kaur E, Agrawal R, Sengupta S. Functions of BLM Helicase in Cells: Is It Acting Like a Double-Edged Sword? Front Genet 2021; 12:634789. [PMID: 33777104 PMCID: PMC7994599 DOI: 10.3389/fgene.2021.634789] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
DNA damage repair response is an important biological process involved in maintaining the fidelity of the genome in eukaryotes and prokaryotes. Several proteins that play a key role in this process have been identified. Alterations in these key proteins have been linked to different diseases including cancer. BLM is a 3′−5′ ATP-dependent RecQ DNA helicase that is one of the most essential genome stabilizers involved in the regulation of DNA replication, recombination, and both homologous and non-homologous pathways of double-strand break repair. BLM structure and functions are known to be conserved across many species like yeast, Drosophila, mouse, and human. Genetic mutations in the BLM gene cause a rare, autosomal recessive disorder, Bloom syndrome (BS). BS is a monogenic disease characterized by genomic instability, premature aging, predisposition to cancer, immunodeficiency, and pulmonary diseases. Hence, these characteristics point toward BLM being a tumor suppressor. However, in addition to mutations, BLM gene undergoes various types of alterations including increase in the copy number, transcript, and protein levels in multiple types of cancers. These results, along with the fact that the lack of wild-type BLM in these cancers has been associated with increased sensitivity to chemotherapeutic drugs, indicate that BLM also has a pro-oncogenic function. While a plethora of studies have reported the effect of BLM gene mutations in various model organisms, there is a dearth in the studies undertaken to investigate the effect of its oncogenic alterations. We propose to rationalize and integrate the dual functions of BLM both as a tumor suppressor and maybe as a proto-oncogene, and enlist the plausible mechanisms of its deregulation in cancers.
Collapse
Affiliation(s)
- Ekjot Kaur
- Signal Transduction Laboratory-2, National Institute of Immunology, New Delhi, India
| | - Ritu Agrawal
- Signal Transduction Laboratory-2, National Institute of Immunology, New Delhi, India
| | - Sagar Sengupta
- Signal Transduction Laboratory-2, National Institute of Immunology, New Delhi, India
| |
Collapse
|
21
|
Ahmed F, Sharma M, Al-Ghamdi AA, Al-Yami SM, Al-Salami AM, Refai MY, Warsi MK, Howladar SM, Baeshen MN. A Comprehensive Analysis of cis-Acting RNA Elements in the SARS-CoV-2 Genome by a Bioinformatics Approach. Front Genet 2020; 11:572702. [PMID: 33424918 PMCID: PMC7786107 DOI: 10.3389/fgene.2020.572702] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/14/2020] [Indexed: 01/08/2023] Open
Abstract
The emergence of a new coronavirus (CoV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for severe respiratory disease in humans termed coronavirus disease of 2019 (COVID-19), became a new global threat for health and the economy. The SARS-CoV-2 genome is about a 29,800-nucleotide-long plus-strand RNA that can form functionally important secondary and higher-order structures called cis-acting RNA elements. These elements can interact with viral proteins, host proteins, or other RNAs and be involved in regulating translation and replication processes of the viral genome and encapsidation of the virus. However, the cis-acting RNA elements and their biological roles in SARS-CoV-2 as well as their comparative analysis in the closely related viral genome have not been well explored, which is very important to understand the molecular mechanism of viral infection and pathogenies. In this study, we used a bioinformatics approach to identify the cis-acting RNA elements in the SARS-CoV-2 genome. Initially, we aligned the full genomic sequence of six different CoVs, and a phylogenetic analysis was performed to understand their evolutionary relationship. Next, we predicted the cis-acting RNA elements in the SARS-CoV-2 genome using the structRNAfinder tool. Then, we annotated the location of these cis-acting RNA elements in different genomic regions of SARS-CoV-2. After that, we analyzed the sequence conservation patterns of each cis-acting RNA element among the six CoVs. Finally, the presence of cis-acting RNA elements across different CoV genomes and their comparative analysis was performed. Our study identified 12 important cis-acting RNA elements in the SARS-CoV-2 genome; among them, Corona_FSE, Corona_pk3, and s2m are highly conserved across most of the studied CoVs, and Thr_leader, MAT2A_D, and MS2 are uniquely present in SARS-CoV-2. These RNA structure elements can be involved in viral translation, replication, and encapsidation and, therefore, can be potential targets for better treatment of COVID-19. It is imperative to further characterize these cis-acting RNA elements experimentally for a better mechanistic understanding of SARS-CoV-2 infection and therapeutic intervention.
Collapse
Affiliation(s)
- Firoz Ahmed
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
- University of Jeddah Center for Scientific and Medical Research, University of Jeddah, Jeddah, Saudi Arabia
| | - Monika Sharma
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | | | | | | | - Mohammed Y. Refai
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
- University of Jeddah Center for Scientific and Medical Research, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohiuddin Khan Warsi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
- University of Jeddah Center for Scientific and Medical Research, University of Jeddah, Jeddah, Saudi Arabia
| | - Saad M. Howladar
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammed N. Baeshen
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
22
|
Ahmed F. A Network-Based Analysis Reveals the Mechanism Underlying Vitamin D in Suppressing Cytokine Storm and Virus in SARS-CoV-2 Infection. Front Immunol 2020; 11:590459. [PMID: 33362771 PMCID: PMC7756074 DOI: 10.3389/fimmu.2020.590459] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/30/2020] [Indexed: 01/08/2023] Open
Abstract
Background SARS-CoV-2 causes ongoing pandemic coronavirus disease of 2019 (COVID-19), infects the cells of the lower respiratory tract that leads to a cytokine storm in a significant number of patients resulting in severe pneumonia, shortness of breathing, respiratory and organ failure. Extensive studies suggested the role of Vitamin D in suppressing cytokine storm in COVID-19 and reducing viral infection; however, the precise molecular mechanism is not clearly known. In this work, bioinformatics and systems biology approaches were used to understand SARS-CoV-2 induced cytokine pathways and the potential mechanism of Vitamin D in suppressing cytokine storm and enhancing antiviral response. Results This study used transcriptome data and identified 108 differentially expressed host genes (DEHGs) in SARS-CoV-2 infected normal human bronchial epithelial (NHBE) cells compared to control. Then, the DEHGs was integrated with the human protein-protein interaction data to generate a SARS-CoV-2 induced host gene regulatory network (SiHgrn). Analysis of SiHgrn identified a sub-network "Cluster 1" with the highest MCODE score, 31 up-regulated genes, and predominantly associated immune and inflammatory response. Interestingly, the iRegulone tool identified that "Cluster 1" is under the regulation of transcription factors STAT1, STAT2, STAT3, POU2F2, and NFkB1, collectively referred to as "host response signature network". Functional enrichment analysis with NDEx revealed that the "host response signature network" is predominantly associated with critical pathways, including "cytokines and inflammatory response", "non-genomic action of Vitamin D", "the human immune response to tuberculosis", and "lung fibrosis". Finally, in-depth analysis and literature mining revealed that Vitamin D binds with its receptor and could work through two different pathways: (i) it inhibits the expression of pro-inflammatory cytokines through blocking the TNF induced NFkB1 signaling pathway; and (ii) it initiates the expression of interferon-stimulating genes (ISGs) for antiviral defense program through activating the IFN-α induced Jak-STAT signaling pathway. Conclusion This comprehensive study identified the pathways associated with cytokine storm in SARS-CoV-2 infection. The proposed underlying mechanism of Vitamin D could be promising in suppressing the cytokine storm and inducing a robust antiviral response in severe COVID-19 patients. The finding in this study urgently needs further experimental validations for the suitability of Vitamin D in combination with IFN-α to control severe COVID-19.
Collapse
Affiliation(s)
- Firoz Ahmed
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia.,University of Jeddah Center for Scientific and Medical Research, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|