1
|
Li W, Chen G, Wang Y, Jiang Y, Wu N, Hu M, Wu T, Yue W. Functional Analysis of BARD1 Missense Variants on Homology-Directed Repair in Ovarian and Breast Cancers. Mol Carcinog 2024. [PMID: 39387837 DOI: 10.1002/mc.23829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/07/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
Women with germline BRCA1 mutations face an increased risk of developing breast and ovarian cancers. BARD1 (BRCA1 associated RING domain 1) is an essential heterodimeric partner of BRCA1, and mutations in BARD1 are also associated with these cancers. While BARD1 mutations are recognized for their cancer susceptibility, the exact roles of numerous BARD1 missense mutations remain unclear. In this study, we conducted functional assays to assess the homology-directed DNA repair (HDR) activity of all BARD1 missense substitutions identified in 55 breast and ovarian cancer samples, using the real-world data from the COSMIC and cBioPortal databases. Seven BARD1 variants (V85M, P187A, G491R, R565C, P669L, T719R, and Q730L) were confirmed to impair DNA damage repair. Furthermore, cells harboring these BARD1 variants exhibited increased sensitivity to the chemotherapeutic drugs, cisplatin, and olaparib, compared to cells expressing wild-type BARD1. These findings collectively suggest that these seven missense BARD1 variants are likely pathogenic and may respond well to cisplatin-olaparib combination therapy. This study not only enhances our understanding of BARD1's role in DNA damage repair but also offers valuable insights into predicting therapy responses in patients with specific BARD1 missense mutations.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Gynecology and Obstetrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Guansheng Chen
- Department of Gynecology and Obstetrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yongjun Wang
- Department of Gynecology and Obstetrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yuening Jiang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
| | - Nanlin Wu
- Department of Pathology, Chuzhou First People's Hospital, Anhui, China
| | - Mingjie Hu
- School of Life Science, Bengbu Medical University, Anhui, China
| | - Taju Wu
- School of Life Science, Bengbu Medical University, Anhui, China
| | - Wei Yue
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Interdisciplinary Eye Research Institute (EYE-X Institute), Bengbu Medical University, Anhui, China
| |
Collapse
|
2
|
Feng Y, Shan L, Gong Y, Hang W, Sang Z, Sun Y, Tang K, Wang Y, Hu B, Xi X. Identification of ubiquitin markers for survival and prognosis of ovarian cancer. Heliyon 2024; 10:e37288. [PMID: 39315145 PMCID: PMC11417246 DOI: 10.1016/j.heliyon.2024.e37288] [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: 08/30/2023] [Revised: 08/11/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
Ovarian cancer (OC) is one of the most common malignancies and a leading cause of death among women worldwide. The ubiquitin pathway plays an important role in OC development. Using the single nucleotide polymorphism data obtained using the prevalence and dominance strategies, four ubiquitin marker genes were identified according to their expression levels: BARD1, BRCA2, FANCA, and BRCA1. Based on these four genes, a consensus clustering of OC expression data was performed. The significant differences in the survival analysis, ESTIMATE, and CIBERSORT results among the clusters indicated the pivotal role of these four genes in OC development. Of the ubiquitin-representative genes in each cluster, two ubiquitin genes, TOP2A and MYLIP, were identified in the survival risk model after univariate survival, Least Absolute Shrinkage and Selection Operator regression, and multivariate survival analyses. The reliability and robustness of both the training and validation data were confirmed by comparing the significant survival difference between high- and low-risk patients. We further explored the association between our risk model and clinical outcomes as well as predicted potentially interacting drugs. The co-expression network showed clear interactions among the four marker genes and two model genes and between high- and low-risk differentially expressed genes. Significantly enriched genes were found in pathways associated with ion channels, channel activity, and neuroactive ligand-receptor interactions. Therefore, suggesting the involvement of ubiquitin genes in the survival and development of OC through neurohumoral regulation. Our results will provide valuable reference or supplementary information for studies investigating OC diagnosis and therapies.
Collapse
Affiliation(s)
- Yiwen Feng
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Liyun Shan
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Yanping Gong
- Departments of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Wenzhao Hang
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Zhenyu Sang
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Yunyan Sun
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Kefu Tang
- Center for Reproductive Medicne, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200135, PR China
| | - Yulan Wang
- Departments of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Binjie Hu
- Departments of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Xiaowei Xi
- Departments of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| |
Collapse
|
3
|
Hawsawi YM, Khoja B, Aljaylani AO, Jaha R, AlDerbi RM, Alnuman H, Khan MI. Recent progress and applications of single-cell sequencing technology in breast cancer. Front Genet 2024; 15:1417415. [PMID: 39359479 PMCID: PMC11445024 DOI: 10.3389/fgene.2024.1417415] [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: 04/14/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technology enables the precise analysis of individual cell transcripts with high sensitivity and throughput. When integrated with multiomics technologies, scRNA-seq significantly enhances the understanding of cellular diversity, particularly within the tumor microenvironment. Similarly, single-cell DNA sequencing has emerged as a powerful tool in cancer research, offering unparalleled insights into the genetic heterogeneity and evolution of tumors. In the context of breast cancer, this technology holds substantial promise for decoding the intricate genomic landscape that drives disease progression, treatment resistance, and metastasis. By unraveling the complexities of tumor biology at a granular level, single-cell DNA sequencing provides a pathway to advancing our comprehension of breast cancer and improving patient outcomes through personalized therapeutic interventions. As single-cell sequencing technology continues to evolve and integrate into clinical practice, its application is poised to revolutionize the diagnosis, prognosis, and treatment strategies for breast cancer. This review explores the potential of single-cell sequencing technology to deepen our understanding of breast cancer, highlighting key approaches, recent advancements, and the role of the tumor microenvironment in disease plasticity. Additionally, the review discusses the impact of single-cell sequencing in paving the way for the development of personalized therapies.
Collapse
Affiliation(s)
- Yousef M Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| | - Basmah Khoja
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | | | - Raniah Jaha
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Rasha Mohammed AlDerbi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Huda Alnuman
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Mohammed I Khan
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
McGrath J, Rhatigan DJ, Donahue D, Fannin M, Lawson N. Infiltrating Ductal Breast Carcinoma in a Male Patient With Associated BARD1 Mutation. Cureus 2024; 16:e66216. [PMID: 39233942 PMCID: PMC11374429 DOI: 10.7759/cureus.66216] [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: 04/05/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Male breast cancer is an uncommon diagnosis with limited research on management and prognosis due to its rarity. We discuss a case of a 55-year-old male with a non-contributory past medical history who presented with an enlarging palpable mass of his right breast tissue at the 10:00 position. The ultrasound of the right breast showed a 2.8 cm heterogenous mass with irregular borders highly suspicious for malignancy. The follow-up sonogram-guided core biopsy was performed, and the pathology of the mass confirmed high-grade infiltrating ductal carcinoma. A modified radical mastectomy of the right breast with extensive axillary lymph node excision was performed. Genetic testing of the excised tumor revealed a MUTYH gene mutation and a BARD1 (BRCA1-associated RING domain 1) gene mutation of unknown significance. Histopathological analysis confirmed a Grade 2, ER/PR-positive, KI 67-positive, and HER2-negative tumor.
Collapse
Affiliation(s)
- Joseph McGrath
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Dillin J Rhatigan
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Danielle Donahue
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Mackenzie Fannin
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Nuria Lawson
- General Surgery, Palmetto General Hospital, Miami, USA
| |
Collapse
|
5
|
Novelli F, Yoshikawa Y, Vitto VAM, Modesti L, Minaai M, Pastorino S, Emi M, Kim JH, Kricek F, Bai F, Onuchic JN, Bononi A, Suarez JS, Tanji M, Favaron C, Zolondick AA, Xu R, Takanishi Y, Wang Z, Sakamoto G, Gaudino G, Grzymski J, Grosso F, Schrump DS, Pass HI, Atanesyan L, Smout J, Savola S, Sarin KY, Abolhassani H, Hammarström L, Pan-Hammarström Q, Giorgi C, Pinton P, Yang H, Carbone M. Germline BARD1 variants predispose to mesothelioma by impairing DNA repair and calcium signaling. Proc Natl Acad Sci U S A 2024; 121:e2405231121. [PMID: 38990952 PMCID: PMC11260134 DOI: 10.1073/pnas.2405231121] [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: 03/13/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
We report that ~1.8% of all mesothelioma patients and 4.9% of those younger than 55, carry rare germline variants of the BRCA1 associated RING domain 1 (BARD1) gene that were predicted to be damaging by computational analyses. We conducted functional assays, essential for accurate interpretation of missense variants, in primary fibroblasts that we established in tissue culture from a patient carrying the heterozygous BARD1V523A mutation. We found that these cells had genomic instability, reduced DNA repair, and impaired apoptosis. Investigating the underlying signaling pathways, we found that BARD1 forms a trimeric protein complex with p53 and SERCA2 that regulates calcium signaling and apoptosis. We validated these findings in BARD1-silenced primary human mesothelial cells exposed to asbestos. Our study elucidated mechanisms of BARD1 activity and revealed that heterozygous germline BARD1 mutations favor the development of mesothelioma and increase the susceptibility to asbestos carcinogenesis. These mesotheliomas are significantly less aggressive compared to mesotheliomas in asbestos workers.
Collapse
Affiliation(s)
- Flavia Novelli
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Yoshie Yoshikawa
- Department of Genetics, School of Medicine, Hyogo Medical University, Hyogo663-8501, Japan
| | - Veronica Angela Maria Vitto
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara44121, Italy
| | - Lorenzo Modesti
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara44121, Italy
| | - Michael Minaai
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Sandra Pastorino
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Mitsuru Emi
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Jin-Hee Kim
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Franz Kricek
- NBS-C Bioscience & Consulting GmbH, Vienna1230, Austria
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies, Shanghai Tech University, Shanghai201210, China
| | - José N. Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX77005
| | - Angela Bononi
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Joelle S. Suarez
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Mika Tanji
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Cristina Favaron
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Alicia A. Zolondick
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI96822
| | - Ronghui Xu
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Yasutaka Takanishi
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Zhanwei Wang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Greg Sakamoto
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Giovanni Gaudino
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | | | - Federica Grosso
- Mesothelioma Unit, Azienda Ospedaliera Santo Antonio and Santo Biagio (SS) Antonio e Biagio e Cesare Arrigo, Alessandria15121, Italy
| | - David S. Schrump
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD20892-1201
| | - Harvey I. Pass
- Department of Cardiothoracic Surgery, New York University, New York, NY10016
| | - Lilit Atanesyan
- Department of Oncogenetics, MRC Holland, Amsterdam 1057, the Netherlands
| | - Jan Smout
- Department of Oncogenetics, MRC Holland, Amsterdam 1057, the Netherlands
| | - Suvi Savola
- Department of Oncogenetics, MRC Holland, Amsterdam 1057, the Netherlands
| | - Kavita Y. Sarin
- Department of Dermatology, Stanford University Medical Center, Stanford, CA94305
| | - Hassan Abolhassani
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Lennart Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm17165, Sweden
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara44121, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara44121, Italy
| | - Haining Yang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| | - Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96816
| |
Collapse
|
6
|
Lu Z, Fan P, Huo W, Feng Y, Wang R. Genomic profiles and their relationships with clinical characteristics and immune features in cervical cancer. Transl Oncol 2024; 44:101923. [PMID: 38432114 PMCID: PMC10920960 DOI: 10.1016/j.tranon.2024.101923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/18/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the genomic alteration profiles of cervical cancer patients, examine the correlation between mutation patterns and clinical and immune attributes, and discover novel targets for treatment of individuals with cervical cancer. METHODS We performed targeted next-generation sequencing of tumor tissues and blood samples obtained from 45 cervical cancer patients to analyze somatic alterations, mutation patterns, and HLA alleles comprehensively. Additionally, we used flow cytometry to assess expression levels of immune checkpoint genes. RESULTS Notably, genes such as AR (78%), KMT2D (76%), and NOTCH1 (62%) exhibited higher mutation frequencies. Moreover, the tumor mutation burden (TMB) was significantly greater in HPV-positive cervical cancer patients than in HPV-negative patients (P=0.029). BMI (P=0.047) and mutations in BARD1 (P=0.034), CEP290 (P=4E-04), and SLX4 (P=0.0128) were identified as predictors of shorter overall survival in cervical cancer patients. Furthermore, the present study revealed significant upregulation of PD-1 (P=0.027) and Tim-3 (P=0.048) in the high mutant-allele tumor heterogeneity (MATH) cohort. In the elderly cervical cancer patient population, HLA-A03:01 emerged as a high-risk allele (OR=3.2, P<0.0001); HLA-C07:02 (OR=0.073, P=0.02) and HLA-B*07:02 (OR=0.257, P=0.037) were associated with a reduced risk among patients with low TMB. CONCLUSIONS This study offers insights into the mutation characteristics of cervical cancer patients and identifies potential therapeutic.
Collapse
Affiliation(s)
- Zinan Lu
- Xinjiang Medical University Affiliated Tumor Hospital, Key Laboratory of Cancer Immunotherapy and Radiotherapy, Chinese Academy of Medical Sciences, Urumqi, Xinjiang 830011, China; Key Laboratory of Oncology of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830011, China
| | - Peiwen Fan
- Xinjiang Medical University Affiliated Tumor Hospital, Key Laboratory of Cancer Immunotherapy and Radiotherapy, Chinese Academy of Medical Sciences, Urumqi, Xinjiang 830011, China; Key Laboratory of Oncology of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830011, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Wen Huo
- Xinjiang Medical University Affiliated Tumor Hospital, Key Laboratory of Cancer Immunotherapy and Radiotherapy, Chinese Academy of Medical Sciences, Urumqi, Xinjiang 830011, China; Key Laboratory of Oncology of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830011, China
| | - Yaning Feng
- Xinjiang Medical University Affiliated Tumor Hospital, Key Laboratory of Cancer Immunotherapy and Radiotherapy, Chinese Academy of Medical Sciences, Urumqi, Xinjiang 830011, China; Key Laboratory of Oncology of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830011, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Ruozheng Wang
- Xinjiang Medical University Affiliated Tumor Hospital, Key Laboratory of Cancer Immunotherapy and Radiotherapy, Chinese Academy of Medical Sciences, Urumqi, Xinjiang 830011, China; Key Laboratory of Oncology of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang 830011, China; Xinjiang Uygur Autonomous Region Radiotherapy Clinical Research and Training Center, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China.
| |
Collapse
|
7
|
Cai Q, Fu Y, Lyu C, Wang Z, Rao S, Alvarez JA, Bai Y, Kang J, Yu T. A new framework for exploratory network mediator analysis in omics data. Genome Res 2024; 34:642-654. [PMID: 38719472 PMCID: PMC11146592 DOI: 10.1101/gr.278684.123] [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: 11/02/2023] [Accepted: 04/11/2024] [Indexed: 06/01/2024]
Abstract
Omics methods are widely used in basic biology and translational medicine research. More and more omics data are collected to explain the impact of certain risk factors on clinical outcomes. To explain the mechanism of the risk factors, a core question is how to find the genes/proteins/metabolites that mediate their effects on the clinical outcome. Mediation analysis is a modeling framework to study the relationship between risk factors and pathological outcomes, via mediator variables. However, high-dimensional omics data are far more challenging than traditional data: (1) From tens of thousands of genes, can we overcome the curse of dimensionality to reliably select a set of mediators? (2) How do we ensure that the selected mediators are functionally consistent? (3) Many biological mechanisms contain nonlinear effects. How do we include nonlinear effects in the high-dimensional mediation analysis? (4) How do we consider multiple risk factors at the same time? To meet these challenges, we propose a new exploratory mediation analysis framework, medNet, which focuses on finding mediators through predictive modeling. We propose new definitions for predictive exposure, predictive mediator, and predictive network mediator, using a statistical hypothesis testing framework to identify predictive exposures and mediators. Additionally, two heuristic search algorithms are proposed to identify network mediators, essentially subnetworks in the genome-scale biological network that mediate the effects of single or multiple exposures. We applied medNet on a breast cancer data set and a metabolomics data set combined with food intake questionnaire data. It identified functionally consistent network mediators for the exposures' impact on the outcome, facilitating data interpretation.
Collapse
Affiliation(s)
- Qingpo Cai
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia 30322, USA
| | - Yinghao Fu
- Shenzhen Research Institute of Big Data, School of Data Science, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
- School of Medicine, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Cheng Lyu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia 30322, USA
| | - Zihe Wang
- Shenzhen Research Institute of Big Data, School of Data Science, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Shun Rao
- Shenzhen Research Institute of Big Data, School of Data Science, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Jessica A Alvarez
- Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Yun Bai
- School of Medicine, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Jian Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Tianwei Yu
- Shenzhen Research Institute of Big Data, School of Data Science, the Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China;
| |
Collapse
|
8
|
Chen S, Xu D, Huang R, Lin Y, Li L. Correlation of BARD1 gene polymorphisms with risk of neuroblastoma: a meta-analysis. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024:1-19. [PMID: 38619196 DOI: 10.1080/15257770.2024.2336215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 03/21/2024] [Indexed: 04/16/2024]
Abstract
BRCA1-associated RING domain protein 1 (BARD1) gene polymorphisms may be associated with neuroblastoma (NB) susceptibility. However, the results remain controversial. Relevant studies were identified by searching PubMed, Web of Science, Embase, China National Knowledge Infrastructure databases up to March 5, 2023. The strength of the association between BARD1 polymorphisms and susceptibility of NB was assessed by calculating odds ratios (ORs) and 95% confidence intervals (95% CIs) through the fixed- or random-effects model. Eight articles involving 12 studies were finally included. We found that rs6435862 T > G, rs3768716 A > G, rs17487792 C > T and rs7587476 C > T variant increase the risk of NB in allelic, dominant, recessive, homozygous and heterozygous genetic models, while rs7585356 G > A variant appeared protective against NB. When stratified by ethnicity, subgroup analysis indicated that the above association remained significant in Caucasian populations in all genetic models, except for rs7585356G > A polymorphism in Asians. In Asian populations, we found the similar results in the allelic and dominant model of rs6435862 T > G, rs3768716 A > G, rs17487792 C > T and rs7587476 C > T as in Caucasians, while there lacked a significant association in the other three model. In addition, rs7585356 G > A was not associated with an increased risk of NB in the Asian population. After Bonferroni correction, significant associations for rs7585356 G > A disappeared in both Asian and Caucasian populations, with no significant association found for rs7587476 in the allelic and dominant models among Asians. BARD1 polymorphisms might be significantly associated with NB susceptibility. It is crucial that these finding should be further confirmed through extensive and well-planned studies.
Collapse
Affiliation(s)
- Shan Chen
- Department of Laboratory, Fuzhou Second General Hospital, Fuzhou, Fujian, China
| | - Di Xu
- Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Rongdong Huang
- Fujian Center for Disease Control and Prevention, Fuzhou, Fujian, China
| | - Yang Lin
- Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Lizhi Li
- Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
9
|
Wang S, Zhang Q, Mao X. Invasive papillary carcinoma of the breast. Front Oncol 2024; 14:1374091. [PMID: 38601769 PMCID: PMC11004302 DOI: 10.3389/fonc.2024.1374091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024] Open
Abstract
Invasive papillary carcinoma is a rare form of breast cancer that is more likely to occur in postmenopausal women. Previous studies have been limited to case reports and small retrospective studies, leading to low awareness of this type of tumor and difficult clinical management. According to the available literature, invasive papillary carcinoma exhibits unique pathological features and biological behaviors. Invasive papillary carcinoma is mostly luminal type, with a low rate of lymph node metastasis, which underlies its favorable prognosis. The effectiveness of adjuvant therapy in reducing tumor burden and improving prognosis in patients with invasive papillary carcinoma remains uncertain. Due to the rarity of the lesion, conducting prospective clinical trials is impractical. The use of biological models, such as organoids, can help alleviate the impact of the scarcity of this condition on research. In addition, invasive papillary carcinoma is affected by specific genomic events, and more extensive studies of gene expression profiling may provide molecular-level insights to make optimal therapeutic decisions.
Collapse
Affiliation(s)
- Shijing Wang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qingfu Zhang
- Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Chen F, Zhang Y, Sedlazeck FJ, Creighton CJ. Germline structural variation globally impacts the cancer transcriptome including disease-relevant genes. Cell Rep Med 2024; 5:101446. [PMID: 38442712 PMCID: PMC10983041 DOI: 10.1016/j.xcrm.2024.101446] [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: 09/26/2023] [Revised: 01/01/2024] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
Germline variation and somatic alterations contribute to the molecular profile of cancers. We combine RNA with whole genome sequencing across 1,218 cancer patients to determine the extent germline structural variants (SVs) impact expression of nearby genes. For hundreds of genes, recurrent and common germline SV breakpoints within 100 kb associate with increased or decreased expression in tumors spanning various tissues of origin. A significant fraction of germline SV expression associations involves duplication of intergenic enhancers or 3' UTR disruption. Genes altered by both somatic and germline SVs include ATRX and CEBPA. Genes essential in cancer cell lines include BARD1 and IRS2. Genes with both expression and germline SV breakpoint patterns associated with patient survival include GCLM. Our results capture a class of phenotypic variation at work in the disease setting, including genes with cancer roles. Specific germline SVs represent potential cancer risk variants for genetic testing, including those involving genes with targeting implications.
Collapse
Affiliation(s)
- Fengju Chen
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Computer Science, Rice University, Houston, TX 77005, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Takaiso N, Imoto I, Yoshimura A, Ouchi A, Komori K, Iwata H, Shimizu Y. BARD1 deletion in a patient with suspected hereditary colorectal cancer. Hum Genome Var 2024; 11:11. [PMID: 38485918 PMCID: PMC10940602 DOI: 10.1038/s41439-024-00267-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/28/2024] [Accepted: 02/06/2024] [Indexed: 03/18/2024] Open
Abstract
Deleterious germline variants in the BRCA1-associated ring domain (BARD1) gene moderately elevate breast cancer risk; however, their potential association with other neoplasms remains unclear. Here, we present the case of a 43-year-old female patient diagnosed with sigmoid colon adenocarcinoma whose maternal family members met the Amsterdam Criteria II for Lynch syndrome. Comprehensive multigene panel testing revealed a heterozygous BARD1 exon 3 deletion.
Collapse
Affiliation(s)
- Nobue Takaiso
- Risk Assessment Unit, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Issei Imoto
- Risk Assessment Unit, Aichi Cancer Center Hospital, Nagoya, Japan.
- Aichi Cancer Center Research Institute, Nagoya, Japan.
| | - Akiyo Yoshimura
- Risk Assessment Unit, Aichi Cancer Center Hospital, Nagoya, Japan
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Akira Ouchi
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Koji Komori
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| |
Collapse
|
12
|
Liu S, Liang Z, Wang Y, Ren Y, Gu Y, Qiao Y, He H, Li Y, Cheng Y, Liu Y. MCM2 is involved in subtyping and tamoxifen resistance of ERα-positive breast cancer by acting as the downstream factor of ERα. Biotechnol J 2024; 19:e2300560. [PMID: 38403459 DOI: 10.1002/biot.202300560] [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: 10/16/2023] [Revised: 12/16/2023] [Accepted: 12/27/2023] [Indexed: 02/27/2024]
Abstract
Tamoxifen (TAM) resistance is finally developed in over 40% of patients with estrogen receptor α-positive breast cancer (ERα+ -BC), documenting that discovering new molecular subtype is needed to confer perception to the heterogeneity of ERα+ -BC. We obtained representative gene sets subtyping ERα+ -BC using gene set variation analysis (GSVA), non-negative matrix factorization (NMF), and COX regression methods on the basis of METABRIC, TCGA, and GEO databases. Furthermore, the risk score of ERα+ -BC subtyping was established using least absolute shrinkage and selection operator (LASSO) regression on the basis of genes in the representative gene sets, thereby generating the two subtypes of ERα+ -BC. We further found that minichromosome maintenance complex component 2 (MCM2) functioned as the hub gene subtyping ERα+ -BC using GO, KEGG, and MCODE. MCM2 expression was capable for specifically predicting 1-year overall survival (OS) of ERα+ -BC and correlated with T stage, AJCC stage, and tamoxifen (TAM) sensitivity of ERα+ -BC. The downregulation of MCM2 expression inhibited proliferation, migration, and invasion of TAM-resistant cells and promoted G0/G1 arrest. Altogether, tamoxifen resistance entails that MCM2 is a hub gene subtyping ERα+ -BC, providing a novel dimension for discovering a potential target of TAM-resistant BC.
Collapse
Affiliation(s)
- Sainan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Zhuoshuai Liang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Yujian Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Yaxuan Ren
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Yulu Gu
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Yichun Qiao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Huan He
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Yong Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Yi Cheng
- Institute of Translational Medicine, the First Hospital of Jilin University, Changchun, China
| | - Yawen Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
13
|
Zhao S, Wang Q, Ni K, Zhang P, Liu Y, Xie J, Ji W, Cheng C, Zhou Q. Combining single-cell sequencing and spatial transcriptome sequencing to identify exosome-related features of glioblastoma and constructing a prognostic model to identify BARD1 as a potential therapeutic target for GBM patients. Front Immunol 2023; 14:1263329. [PMID: 37727789 PMCID: PMC10505933 DOI: 10.3389/fimmu.2023.1263329] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
Background Glioblastoma (GBM) is a malignant primary brain tumor. This study focused on exploring the exosome-related features of glioblastoma to better understand its cellular composition and molecular characteristics. Methods Single-cell RNA sequencing (scRNA-seq) and spatial transcriptome RNA sequencing (stRNA-seq) were used to analyze the heterogeneity of glioblastomas. After data integration, cell clustering, and annotation, five algorithms were used to calculate scores for exosome-related genes(ERGs). Cell trajectory analysis and intercellular communication analysis were performed to explore exosome-related communication patterns. Spatial transcriptome sequencing data were analyzed to validate the findings. To further utilize exosome-related features to aid in clinical decision-making, a prognostic model was constructed using GBM's bulk RNA-seq. Results Different cell subpopulations were observed in GBM, with Monocytes/macrophages and malignant cells in tumor samples showing higher exosome-related scores. After identifying differentially expressed ERGs in malignant cells, pseudotime analysis revealed the cellular status of malignant cells during development. Intercellular communication analysis highlighted signaling pathways and ligand-receptor interactions. Spatial transcriptome sequencing confirmed the high expression of exosome-related gene features in the tumor core region. A prognostic model based on six ERGs was shown to be predictive of overall survival and immunotherapy outcome in GBM patients. Finally, based on the results of scRNA-seq and prognostic modeling as well as a series of cell function experiments, BARD1 was identified as a novel target for the treatment of GBM. Conclusion This study provides a comprehensive understanding of the exosome-related features of GBM in both scRNA-seq and stRNA-seq, with malignant cells with higher exosome-related scores exhibiting stronger communication with Monocytes/macrophages. In terms of spatial data, highly scored malignant cells were also concentrated in the tumor core region. In bulk RNA-seq, patients with a high exosome-related index exhibited an immunosuppressive microenvironment, which was accompanied by a worse prognosis as well as immunotherapy outcomes. Prognostic models constructed using ERGs are expected to be independent prognostic indicators for GBM patients, with potential implications for personalized treatment strategies for GBM. Knockdown of BARD1 in GBM cell lines reduces the invasive and value-added capacity of tumor cells, and thus BARD1-positively expressing malignant cells are a risk factor for GBM patients.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Neurosurgery, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kaixiang Ni
- Department of Neurosurgery, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuan Liu
- Department of General Surgery, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Ji
- Department of Neurosurgery, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Chao Cheng
- Department of Neurosurgery, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Qiang Zhou
- Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, China
| |
Collapse
|
14
|
Albarakati N, Al-Ghamdi H, Al-Sowayan B, Alshareeda A. Homologous recombination mRNAs (RAD21, RAD50 and BARD1) have a potentially poor prognostic role in ERBB2-low bladder cancer patients. Sci Rep 2023; 13:11738. [PMID: 37474724 PMCID: PMC10359419 DOI: 10.1038/s41598-023-38923-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2/ERBB2) factor is known to be implicated in many malignancies and the potential of it as a prognostic biomarker was reported years ago. Molecular subtypes of HER2/ERBB2 negative and positive with distinct clinical outcomes have been identified in recent years; however, it is still under investigation for bladder cancer. This study evaluates the biological and prognostic significance of RAD21, RAD50 and BARD1 (homologous recombination biomarkers) mRNA levels with ERBB2 low and high expression to explore their impact on bladder cancer patient survival and cancer aggressiveness. The expression of ERBB2, RAD21, RAD50 and BARD1 mRNA levels was assessed in The Cancer Genome Atlas (TCGA) bladder cancer dataset along with four validation cohorts. Outcome analysis was evaluated using disease-free survival (DFS) and overall survival (OS). Univariate and multivariate analysis were used to evaluate the relationship between RAD21, RAD50, BARD1 and ERBB2 expression and clinicopathological variables. A significant increase in mRNA expression levels of RAD21, RAD50 and BARD1 was noticed in ERBB2-low patients compared to ERBB2-high patients. This overexpression of the homologous recombination repair transcripts was associated with poor outcome in ERBB2-low tumors, not in ERBB2-high tumors. Furthermore, the combined expression of high RAD21/RAD50, high RAD21/BARD1 or high RAD50/BARD1 were significantly associated with worse DFS and a better outcome for those with low co-expression in the ERBB2-low cohort. High expression of either RAD21/RAD50 or RAD21/BARD1 in ERBB2-low cohort associated with higher chance of metastasis. In addition, gene expression of BARD1 alone or in combination with RAD50 acted as an independent prognostic factor for worst survival. The data presented in this study reveal a connection between RAD21, RAD50, BARD1 and ERBB2 and patient survival. Importantly, it provided novel findings and potential prognostic markers, particularly in ERBB2-low bladder cancer.
Collapse
Affiliation(s)
- Nada Albarakati
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Jeddah, Kingdom of Saudi Arabia.
- King Saud Bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia.
| | - Hanin Al-Ghamdi
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Jeddah, Kingdom of Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Batla Al-Sowayan
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Alaa Alshareeda
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
George M, Masamba P, Iwalokun BA, Kappo AP. Zooming into the structure-function of RING finger proteins for anti-cancer therapeutic applications. Am J Cancer Res 2023; 13:2773-2789. [PMID: 37559981 PMCID: PMC10408477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/22/2023] [Indexed: 08/11/2023] Open
Abstract
Cancer is one of the most common and widely diagnosed diseases worldwide. With an increase in prevalence and incidence, many studies in cancer biology have been looking at the role pro-cancer proteins play. One of these proteins is the Really Interesting New Gene (RING), which has been studied extensively due to its structure and functions such as apoptosis, neddylation, and its role in ubiquitination. The RING domain is a cysteine-rich domain known to bind Cysteine and Histidine residues. It also binds two zinc ions that help stabilize the protein in various patterns, often with a 'cross-brace' topology. Different RING finger proteins have been studied and found to have suitable targets for developing anti-cancer therapeutics. These identified candidate proteins include Parkin, COP1, MDM2, BARD1, BRCA-1, PIRH2, c-CBL, SIAH1, RBX1 and RNF8. Inhibiting these candidate proteins provides opportunities for shutting down pathways associated with tumour development and metastasis.
Collapse
Affiliation(s)
- Mary George
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway CampusAuckland Park, Johannesburg, South Africa
| | - Priscilla Masamba
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway CampusAuckland Park, Johannesburg, South Africa
| | - Bamidele Abiodun Iwalokun
- Department of Molecular Biology and Biotechnology, Nigerian Institute of Medical Research (NIMR)Yaba, Lagos, Nigeria
| | - Abidemi Paul Kappo
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway CampusAuckland Park, Johannesburg, South Africa
| |
Collapse
|
16
|
Kurochkina N, Sapio MR, Iadarola MJ, Hall BE, Kulkarni AB. Multiprotein Assemblies, Phosphorylation and Dephosphorylation in Neuronal Cytoskeleton. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.21.545989. [PMID: 37502949 PMCID: PMC10370197 DOI: 10.1101/2023.06.21.545989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Filament systems are comprised of fibrous and globular cytoskeletal proteins and are key elements regulating cell shape, rigidity, and dynamics. The cellular localization and assembly of neurofilaments depend on phosphorylation by kinases. The involvement of the BRCA1 (Breast cancer associated protein 1)/BARD1 (BRCA1-associated RING domain 1) pathways in Alzheimer disease (AD) is suggested by colocalization studies. In particular, BRCA1 accumulation within neurofibrillary tangles and colocalization with tau aggregates in the cytoplasm of AD patients implicates the involvement of mutant forms of BRCA1/BARD1 proteins in disease pathogenesis. The purpose of this study is to show that the location of mutations in the translated BARD1, specifically within ankyrin repeats, has strong correlation with the Cdk5 motifs for phosphorylation. Mapping of the mutation sites on the protein's three-dimensional structure and estimation of the backbone dihedral angles show transitions between the canonical helical and extended conformations of the tetrapeptide sequence of ankyrin repeats. Clustering of mutations in BARD1 ankyrin repeats near the N-termini of the helices with T/SXXH motifs provides a basis for conformational transitions that might be necessary to ensure the compatibility of the substrate with active site geometry and accessibility of the substrate to the kinase. Ankyrin repeats are interaction sites for phosphorylation-dependent dynamic assembly of proteins including those involved in transcription regulation and signaling, and present potential targets for the design of new drugs.
Collapse
Affiliation(s)
- Natalya Kurochkina
- Department of Biophysics, The School of Theoretical Modeling, Washington, DC, 20006, USA
| | - Matthew R. Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Michael J. Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Bradford E Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Ashok B. Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
17
|
Carrera S, Rodríguez-Martínez AB, Garin I, Sarasola E, Martínez C, Maortua H, Callejo A, Ruiz de Lobera A, Muñoz A, Miñambres N, Jiménez-Labaig P. Germline heterozygous exons 8-11 pathogenic BARD1 gene deletion reported for the first time in a family with suspicion of a hereditary colorectal cancer syndrome: more than an incidental finding? Hered Cancer Clin Pract 2023; 21:2. [PMID: 36709314 PMCID: PMC9883939 DOI: 10.1186/s13053-023-00246-4] [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: 10/21/2022] [Accepted: 01/24/2023] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a highly prevalent disease in developed countries. Inherited Mendelian causes account for approximately 5% of CRC cases, with Lynch syndrome and familial adenomatous polyposis being the most prevalent forms. Scientific efforts are focused on the discovery of new candidate genes associated with CRC and new associations of phenotypes with well-established cancer-related genes. BRCA1-associated ring domain (BARD1) gene deleterious germline variants are associated with a moderate increase in the relative risk of breast cancer, but their association with other neoplasms, such as CRC, remains unclear. CASE PRESENTATION We present the case of a 49-year-old male diagnosed with rectal adenocarcinoma whose maternal family fulfilled Amsterdam clinical criteria for Lynch syndrome. Genetic test confirmed the presence in heterozygosis of a germline pathogenic deletion of exons 8-11 in BARD1 gene. The predictive genetic study of the family revealed the presence of this pathogenic variant in his deceased cancer affected relatives, confirming co-segregation of the deletion with the disease. CONCLUSIONS To the best of our knowledge, this is the first published work in which this BARD1 deletion is detected in a family with familial colorectal cancer type X (FCCTX) syndrome, in which the clinical criteria for Lynch syndrome without alteration of the DNA mismatch repair (MMR) system are fulfilled. Whether this incidental germline finding is the cause of familial colorectal aggregation remains to be elucidated in scientific forums. Patients should be carefully assessed in specific cancer genetic counseling units to account for hypothetical casual findings in other genes, in principle unrelated to the initial clinical suspicion, but with potential impact on their health.
Collapse
Affiliation(s)
- Sergio Carrera
- grid.411232.70000 0004 1767 5135Hereditary Cancer Genetic Counseling Unit- Medical Oncology Department, Cruces University Hospital, Plaza de Cruces S/N. 48903, Baracaldo, Bizkaia Spain
| | | | - Intza Garin
- grid.411232.70000 0004 1767 5135Molecular Genetics Laboratory, Cruces University Hospital, Baracaldo, Spain
| | - Esther Sarasola
- grid.414269.c0000 0001 0667 6181Molecular Genetics Laboratory, Basurto University Hospital, Bilbao, Spain
| | - Cristina Martínez
- grid.411232.70000 0004 1767 5135Molecular Genetics Laboratory, Cruces University Hospital, Baracaldo, Spain
| | - Hiart Maortua
- grid.411232.70000 0004 1767 5135Molecular Genetics Laboratory, Cruces University Hospital, Baracaldo, Spain
| | - Almudena Callejo
- grid.411232.70000 0004 1767 5135Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| | - Abigail Ruiz de Lobera
- grid.411232.70000 0004 1767 5135Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| | - Alberto Muñoz
- grid.411232.70000 0004 1767 5135Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| | - Nagore Miñambres
- grid.411232.70000 0004 1767 5135Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| | - Pablo Jiménez-Labaig
- grid.411232.70000 0004 1767 5135Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| |
Collapse
|
18
|
Mounai Y, Yoshida T, Ito S, Fukuda K, Shimazu K, Taguchi D, Shinozaki H, Takagi D, Imai K, Yamamoto H, Minamiya Y, Nanjyo H, Shibata H. Pulmonary Artery Intimal Sarcoma in a Patient with Lynch Syndrome: Response to an Immune Checkpoint Inhibitor. Case Rep Oncol 2023; 16:21-29. [PMID: 36743879 PMCID: PMC9891846 DOI: 10.1159/000528682] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/06/2022] [Indexed: 01/28/2023] Open
Abstract
Intimal sarcoma is an extremely rare mesenchymal tumor arising in the great vessels. To date, intimal sarcoma has not been reported in patients with Lynch syndrome (LS), even though this syndrome lacks DNA mismatch repair ability genetically and is prone to various malignancies. This patient was diagnosed with LS by the Revised Amsterdam Criteria II, and she suffered from intimal sarcoma in the left pulmonary artery. She had a germline missense variant of PMS2 (c.1399G>A, pV467I) which is classified as a variant of unknown significance. In her intimal sarcoma, PMS2 expression was decreased. Additionally, it exhibited microsatellite instability and a high tumor mutational burden (69 mutations/Mb) which are features of mismatch repair deficiency, although PMS2 (c.1399G>A, pV467I) missense is a variant of unknown significance. The metastatic lesions of intimal sarcoma in this patient responded heterogeneously to pembrolizumab, an immune checkpoint inhibitor. Cytotoxic agents and radiation were also effective for some metastatic lesions, but some lesions, including her liver metastases, were resistant. The hypermutable nature of the LS genotype might acquire resistance to an immune checkpoint inhibitor and other cytotoxic agents such as occurred with her liver metastases.
Collapse
Affiliation(s)
- Yue Mounai
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Taichi Yoshida
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Shogo Ito
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Koji Fukuda
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Kazuhiro Shimazu
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Daiki Taguchi
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Hanae Shinozaki
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Daichi Takagi
- Department of Cardiovascular Surgery, Graduate School of Medicine, Akita University, Akita, Japan
| | - Kazuhiro Imai
- Department of Thoracic Surgery, Graduate School of Medicine, Akita University, Akita, Japan
| | - Hiroshi Yamamoto
- Department of Cardiovascular Surgery, Graduate School of Medicine, Akita University, Akita, Japan
| | - Yoshihiro Minamiya
- Department of Thoracic Surgery, Graduate School of Medicine, Akita University, Akita, Japan
| | - Hiroshi Nanjyo
- Department of Pathology, Akita University Hospital, Akita, Japan
| | - Hiroyuki Shibata
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Akita, Japan
| |
Collapse
|
19
|
Ottaiano A, Santorsola M, Circelli L, Cascella M, Petrillo N, Perri F, Casillo M, Granata V, Ianniello M, Izzo F, Picone C, Correra M, Petrillo A, Sirica R, Misso G, Delrio P, Nasti G, Savarese G, Caraglia M. Genetic landscape of colorectal cancer patients manifesting tumor shrinkage during SARS-Cov-2 infection. Ther Adv Med Oncol 2022; 14:17588359221138388. [PMCID: PMC9742504 DOI: 10.1177/17588359221138388] [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: 08/10/2022] [Accepted: 10/26/2022] [Indexed: 12/13/2022] Open
Abstract
We previously described three patients affected by metastatic colorectal cancer (mCRC) who experienced spontaneous tumour shrinkage during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Thereafter, the patients were closely monitored and no systemic treatments were applied. Here, we report follow-up clinical information about these patients as well as genetic characterization of their primary tumours through the TruSigt™Oncology 500 Next Generation Sequencing test targeting 523 cancer-relevant genes. An Illumina NovaSeq 6000 platform was used to perform sequencing. Time-to-progression was 23 and 2 months, respectively, in Patients 2 and 3 while it was not reached in Patient 1. Patients 1 and 2 had the greatest anti-SARS-CoV-2 IgG titres. Assessment of genetic landscapes evidenced common mutation in BARD1 gene (p.Val507Met) in Patients 1 and 2. Although our report is descriptive in its nature, we suggest that complex and unexplored interactions between genetic background and components of the immune response to SARS-CoV-2 infection could be responsible of unexpected rare mCRC shrinkage.
Collapse
Affiliation(s)
| | | | | | - Marco Cascella
- Unit of Anestesiology and Pain Therapy, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Nadia Petrillo
- AMES, Centro Polidiagnostico Strumentale, Casalnuovo Di Napoli, Naples, Italy
| | - Francesco Perri
- Medical and Experimental Head and Neck Oncology Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Marika Casillo
- AMES, Centro Polidiagnostico Strumentale, Casalnuovo Di Napoli, Naples, Italy
| | - Vincenza Granata
- Department of Radiology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Monica Ianniello
- AMES, Centro Polidiagnostico Strumentale, Casalnuovo Di Napoli, Naples, Italy
| | - Francesco Izzo
- Hepato-Biliary Surgey Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Carmine Picone
- Department of Radiology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Marco Correra
- Department of Radiology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Antonella Petrillo
- Department of Radiology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Roberto Sirica
- AMES, Centro Polidiagnostico Strumentale, Casalnuovo Di Napoli, Naples, Italy
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Paolo Delrio
- Colorectal Surgical Oncology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Guglielmo Nasti
- SSD-Innovative Therapies for Abdominal Metastases, Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | | | | |
Collapse
|